Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 81
1.
Alzheimers Dement ; 2022 Dec 08.
Article En | MEDLINE | ID: mdl-36479795

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.

2.
Int J Mol Sci ; 23(20)2022 Oct 17.
Article En | MEDLINE | ID: mdl-36293260

Abnormal glucose metabolism is central to neurodegeneration, and considerable evidence suggests that abnormalities in key enzymes of the tricarboxylic acid (TCA) cycle underlie the metabolic deficits. Significant recent advances in the role of metabolism in cancer provide new insight that facilitates our understanding of the role of metabolism in neurodegeneration. Research indicates that the rate-limiting step of the TCA cycle, the α-ketoglutarate dehydrogenase complex (KGDHC) and its substrate alpha ketoglutarate (KG), serve as a signaling hub that regulates multiple cellular processes: (1) is the rate-limiting step of the TCA cycle, (2) is sensitive to reactive oxygen species (ROS) and produces ROS, (3) determines whether KG is used for energy or synthesis of compounds to support growth, (4) regulates the cellular responses to hypoxia, (5) controls the post-translational modification of hundreds of cell proteins in the mitochondria, cytosol, and nucleus through succinylation, (6) controls critical aspects of transcription, (7) modulates protein signaling within cells, and (8) modulates cellular calcium. The primary focus of this review is to understand how reductions in KGDHC are translated to pathologically important changes that underlie both neurodegeneration and cancer. An understanding of each role is necessary to develop new therapeutic strategies to treat neurodegenerative disease.


Ketoglutarate Dehydrogenase Complex , Neurodegenerative Diseases , Humans , Ketoglutarate Dehydrogenase Complex/metabolism , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism , Calcium/metabolism , Ketoglutaric Acids , Glucose , Tricarboxylic Acids
4.
Nat Commun ; 13(1): 159, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013160

Abnormalities in brain glucose metabolism and accumulation of abnormal protein deposits called plaques and tangles are neuropathological hallmarks of Alzheimer's disease (AD), but their relationship to disease pathogenesis and to each other remains unclear. Here we show that succinylation, a metabolism-associated post-translational protein modification (PTM), provides a potential link between abnormal metabolism and AD pathology. We quantified the lysine succinylomes and proteomes from brains of individuals with AD, and healthy controls. In AD, succinylation of multiple mitochondrial proteins declined, and succinylation of small number of cytosolic proteins increased. The largest increases occurred at critical sites of amyloid precursor protein (APP) and microtubule-associated tau. We show that in vitro, succinylation of APP disrupted its normal proteolytic processing thereby promoting Aß accumulation and plaque formation and that succinylation of tau promoted its aggregation to tangles and impaired microtubule assembly. In transgenic mouse models of AD, elevated succinylation associated with soluble and insoluble APP derivatives and tau. These findings indicate that a metabolism-linked PTM may be associated with AD.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/metabolism , Protein Processing, Post-Translational , Succinic Acid/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Protein Precursor/genetics , Animals , Autopsy , Brain/metabolism , Brain/pathology , Case-Control Studies , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Protein Aggregates , Proteolysis , Proteome/genetics , Proteome/metabolism , tau Proteins/genetics
5.
Int J Mol Sci ; 22(24)2021 Dec 07.
Article En | MEDLINE | ID: mdl-34947984

Serum metabolomics and lipidomics are powerful approaches for discovering unique biomarkers in various diseases and associated therapeutics and for revealing metabolic mechanisms of both. Treatment with Benfotiamine (BFT), a thiamine prodrug, for one year produced encouraging results for patients with mild cognitive impairment and mild Alzheimer's disease (AD). In this study, a parallel metabolomics and lipidomics approach was applied for the first exploratory investigation on the serum metabolome and lipidome of patients treated with BFT. A total of 315 unique metabolites and 417 lipids species were confidently identified and relatively quantified. Rigorous statistical analyses revealed significant differences between the placebo and BFT treatment groups in 25 metabolites, including thiamine, tyrosine, tryptophan, lysine, and 22 lipid species, mostly belonging to phosphatidylcholines. Additionally, 10 of 11 metabolites and 14 of 15 lipid species reported in previous literature to follow AD progression changed in the opposite direction to those reported to reflect AD progression. Enrichment and pathway analyses show that significantly altered metabolites by BFT are involved in glucose metabolism and biosynthesis of aromatic amino acids. Our study discovered that multiple novel biomarkers and multiple mechanisms that may underlie the benefit of BFT are potential therapeutic targets in AD and should be validated in studies with larger sample sizes.


Alzheimer Disease/drug therapy , Biomarkers/blood , Metabolomics/methods , Thiamine/analogs & derivatives , Alzheimer Disease/blood , Case-Control Studies , Chromatography, Liquid , Humans , Lipids/blood , Mass Spectrometry , Metabolic Networks and Pathways , Pilot Projects , Thiamine/administration & dosage , Thiamine/pharmacology
6.
J Neurochem ; 158(2): 282-296, 2021 07.
Article En | MEDLINE | ID: mdl-33905124

Metabolic changes that correlate to cognitive changes are well-known in Alzheimer's disease (AD). Metabolism is often linked to functional changes in proteins by post-translational modifications. The importance of the regulation of transcription by acetylation is well documented. Advanced mass spectrometry reveals hundreds of acetylated proteins in multiple tissues, but the acetylome of human brain, its functional significance, and the changes with disease are unknown. Filling this gap is critical for understanding the pathophysiology and development of therapies. To fill this gap, we assessed the human brain acetylome in human brain and its changes with AD. More than 5% of the 4,442 proteins from the human brain global proteome were acetylated. Acetylated proteins were primarily found in the cytosol (148), mitochondria (100), nucleus (91), and plasma membrane (58). The comparison of the brain acetylome in controls to that of patients with AD revealed striking and selective differences in terms of its abundances of acetylated peptides/sites. Acetylation of 18 mitochondrial proteins decreased, while acetylation of two cytosolic proteins, tau and GFAP, increased. Our experiments demonstrate that acetylation at some specific lysine sites alters enzyme function. The results indicate that general activation of de-acetylases (i.e., sirtuins) is not an appropriate therapeutic approach for AD.


Acetylation , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Metabolome/genetics , Mitochondrial Proteins/metabolism , Aged , Aged, 80 and over , Brain Chemistry , Computational Biology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/metabolism , Lysine/metabolism , Male , Protein Processing, Post-Translational , Pyruvate Dehydrogenase Complex/metabolism , Subcellular Fractions/metabolism , tau Proteins/metabolism
7.
J Neurochem ; 156(6): 867-879, 2021 03.
Article En | MEDLINE | ID: mdl-32865230

Mitochondria and releasable endoplasmic reticulum (ER) calcium modulate neuronal calcium signaling, and both change in Alzheimer's disease (AD). The releasable calcium stores in the ER are exaggerated in fibroblasts from AD patients and in multiple models of AD. The activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC), a key mitochondrial enzyme complex, is diminished in brains from AD patients, and can be plausibly linked to plaques and tangles. Our previous studies in cell lines and mouse neurons demonstrate that reductions in KGDHC increase the ER releasable calcium stores. The goal of these studies was to test whether the relationship was true in human iPSC-derived neurons. Inhibition of KGDHC for one or 24 hr increased the ER releasable calcium store in human neurons by 69% and 144%, respectively. The effect was mitochondrial enzyme specific because inhibiting the pyruvate dehydrogenase complex, another key mitochondrial enzyme complex, diminished the ER releasable calcium stores. The link of KGDHC to ER releasable calcium stores was cell type specific as the interaction was not present in iPSC or neural stem cells. Thus, these studies in human neurons verify a link between KGDHC and releasable ER calcium stores, and support the use of human neurons to examine mechanisms and potential therapies for AD.


Calcium/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/enzymology , Neural Stem Cells/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Immunohistochemistry , Ketoglutarate Dehydrogenase Complex/metabolism , Potassium/metabolism , Pyruvate Dehydrogenase Complex/metabolism
8.
J Alzheimers Dis ; 78(3): 989-1010, 2020.
Article En | MEDLINE | ID: mdl-33074237

BACKGROUND: In preclinical models, benfotiamine efficiently ameliorates the clinical and biological pathologies that define Alzheimer's disease (AD) including impaired cognition, amyloid-ß plaques, neurofibrillary tangles, diminished glucose metabolism, oxidative stress, increased advanced glycation end products (AGE), and inflammation. OBJECTIVE: To collect preliminary data on feasibility, safety, and efficacy in individuals with amnestic mild cognitive impairment (aMCI) or mild dementia due to AD in a placebo-controlled trial of benfotiamine. METHODS: A twelve-month treatment with benfotiamine tested whether clinical decline would be delayed in the benfotiamine group compared to the placebo group. The primary clinical outcome was the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). Secondary outcomes were the clinical dementia rating (CDR) score and fluorodeoxyglucose (FDG) uptake, measured with brain positron emission tomography (PET). Blood AGE were examined as an exploratory outcome. RESULTS: Participants were treated with benfotiamine (34) or placebo (36). Benfotiamine treatment was safe. The increase in ADAS-Cog was 43% lower in the benfotiamine group than in the placebo group, indicating less cognitive decline, and this effect was nearly statistically significant (p = 0.125). Worsening in CDR was 77% lower (p = 0.034) in the benfotiamine group compared to the placebo group, and this effect was stronger in the APOEɛ4 non-carriers. Benfotiamine significantly reduced increases in AGE (p = 0.044), and this effect was stronger in the APOEɛ4 non-carriers. Exploratory analysis derivation of an FDG PET pattern score showed a treatment effect at one year (p = 0.002). CONCLUSION: Oral benfotiamine is safe and potentially efficacious in improving cognitive outcomes among persons with MCI and mild AD.


Alzheimer Disease/drug therapy , Brain/diagnostic imaging , Cognitive Dysfunction/drug therapy , Thiamine/analogs & derivatives , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Aniline Compounds , Apolipoprotein E4/genetics , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Disease Progression , Ethylene Glycols , Female , Fluorodeoxyglucose F18 , Glycation End Products, Advanced/blood , Humans , Male , Positron-Emission Tomography , Radiopharmaceuticals , Thiamine/therapeutic use , Treatment Outcome
9.
Basic Res Cardiol ; 115(5): 53, 2020 08 03.
Article En | MEDLINE | ID: mdl-32748289

In heart failure, a functional block of complex I of the respiratory chain provokes superoxide generation, which is transformed to H2O2 by dismutation. The Krebs cycle produces NADH, which delivers electrons to complex I, and NADPH for H2O2 elimination via isocitrate dehydrogenase and nicotinamide nucleotide transhydrogenase (NNT). At high NADH levels, α-ketoglutarate dehydrogenase (α-KGDH) is a major source of superoxide in skeletal muscle mitochondria with low NNT activity. Here, we analyzed how α-KGDH and NNT control H2O2 emission in cardiac mitochondria. In cardiac mitochondria from NNT-competent BL/6N mice, H2O2 emission is equally low with pyruvate/malate (P/M) or α-ketoglutarate (α-KG) as substrates. Complex I inhibition with rotenone increases H2O2 emission from P/M, but not α-KG respiring mitochondria, which is potentiated by depleting H2O2-eliminating capacity. Conversely, in NNT-deficient BL/6J mitochondria, H2O2 emission is higher with α-KG than with P/M as substrate, and further potentiated by complex I blockade. Prior depletion of H2O2-eliminating capacity increases H2O2 emission from P/M, but not α-KG respiring mitochondria. In cardiac myocytes, downregulation of α-KGDH activity impaired dynamic mitochondrial redox adaptation during workload transitions, without increasing H2O2 emission. In conclusion, NADH from α-KGDH selectively shuttles to NNT for NADPH formation rather than to complex I of the respiratory chain for ATP production. Therefore, α-KGDH plays a key role for H2O2 elimination, but is not a relevant source of superoxide in heart. In heart failure, α-KGDH/NNT-dependent NADPH formation ameliorates oxidative stress imposed by complex I blockade. Downregulation of α-KGDH may, therefore, predispose to oxidative stress in heart failure.


Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria, Heart/metabolism , NADP Transhydrogenases/metabolism , NAD/metabolism , Animals , Cell Respiration , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Single-Cell Analysis
10.
Neurochem Res ; 44(10): 2346-2359, 2019 Oct.
Article En | MEDLINE | ID: mdl-30903449

Post-translational modifications (PTMs) are important regulators of protein function, and integrate metabolism with physiological and pathological processes. Phosphorylation and acetylation are particularly well studied PTMs. A relatively recently discovered novel PTM is succinylation in which metabolically derived succinyl CoA modifies protein lysine groups. Succinylation causes a protein charge flip from positive to negative and a relatively large increase in mass compared to other PTMs. Hundreds of protein succinylation sites are present in proteins of multiple tissues and species, and the significance is being actively investigated. The few completed studies demonstrate that succinylation alters rates of enzymes and pathways, especially mitochondrial metabolic pathways. Thus, succinylation provides an elegant and efficient mechanism to coordinate metabolism and signaling by utilizing metabolic intermediates as sensors to regulate metabolism. Even though the brain is one of the most metabolically active organs, an understanding of the role succinylation in the nervous system is largely unknown. Data from other tissues and other PTMs suggest that succinylation provides a coupling between metabolism and protein function in the nervous system and in neurological diseases. This review provides a new insight into metabolism in neurological diseases and suggests that the drug development for these diseases requires a better understanding of succinylation and de-succinylation in the brain and other tissues.


Acyl Coenzyme A/metabolism , Lysine/metabolism , Metabolic Networks and Pathways/physiology , Mitochondria/metabolism , Protein Processing, Post-Translational/physiology , Animals , Humans , Proteome/metabolism
11.
Hum Mol Genet ; 27(16): 2874-2892, 2018 08 15.
Article En | MEDLINE | ID: mdl-29860433

Impaired glucose metabolism, decreased levels of thiamine and its phosphate esters, and reduced activity of thiamine-dependent enzymes, such as pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase and transketolase occur in Alzheimer's disease (AD). Thiamine deficiency exacerbates amyloid beta (Aß) deposition, tau hyperphosphorylation and oxidative stress. Benfotiamine (BFT) rescued cognitive deficits and reduced Aß burden in amyloid precursor protein (APP)/PS1 mice. In this study, we examined whether BFT confers neuroprotection against tau phosphorylation and the generation of neurofibrillary tangles (NFTs) in the P301S mouse model of tauopathy. Chronic dietary treatment with BFT increased lifespan, improved behavior, reduced glycated tau, decreased NFTs and prevented death of motor neurons. BFT administration significantly ameliorated mitochondrial dysfunction and attenuated oxidative damage and inflammation. We found that BFT and its metabolites (but not thiamine) trigger the expression of Nrf2/antioxidant response element (ARE)-dependent genes in mouse brain as well as in wild-type but not Nrf2-deficient fibroblasts. Active metabolites were more potent in activating the Nrf2 target genes than the parent molecule BFT. Docking studies showed that BFT and its metabolites (but not thiamine) bind to Keap1 with high affinity. These findings demonstrate that BFT activates the Nrf2/ARE pathway and is a promising therapeutic agent for the treatment of diseases with tau pathology, such as AD, frontotemporal dementia and progressive supranuclear palsy.


Antioxidant Response Elements/genetics , NF-E2-Related Factor 2/genetics , Protein Aggregation, Pathological/drug therapy , Tauopathies/drug therapy , Thiamine/analogs & derivatives , Amyloid beta-Peptides/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Transgenic , Neuroprotection/drug effects , Oxidative Stress/drug effects , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Signal Transduction/drug effects , Tauopathies/genetics , Tauopathies/physiopathology , Thiamine/administration & dosage , tau Proteins/genetics
12.
Neurochem Int ; 117: 65-76, 2018 07.
Article En | MEDLINE | ID: mdl-28627365

The induction of pluripotent stem cells (iPSC) from differentiated cells such as fibroblasts and their subsequent conversion to neural progenitor cells (NPC) and finally to neurons is intriguing scientifically, and its potential to medicine is nearly infinite, but unrealized. A better understanding of the changes at each step of the transformation will enable investigators to better model neurological disease. Each step of conversion from a differentiated cell to an iPSC to a NPC to neurons requires large changes in glycolysis including aerobic glycolysis, the pentose shunt, the tricarboxylic acid cycle, the electron transport chain and in the production of reactive oxygen species (ROS). These mitochondrial/metabolic changes are required and their manipulation modifies conversions. These same mitochondrial/metabolic processes are altered in common neurological diseases so that factors related to the disease may alter the cellular transformation at each step including the final phenotype. A lack of understanding of these interactions could compromise the validity of the disease comparisons in iPSC derived neurons. Both the complexity and potential of iPSC derived cells for understanding and treating disease remain great.


Cell Differentiation/physiology , Cellular Reprogramming/physiology , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Humans , Reactive Oxygen Species/metabolism
13.
J Neurosci Res ; 95(11): 2244-2252, 2017 11.
Article En | MEDLINE | ID: mdl-28631845

Succinylation of proteins is widespread, modifies both the charge and size of the molecules, and can alter their function. For example, liver mitochondrial proteins have 1,190 unique succinylation sites representing multiple metabolic pathways. Succinylation is sensitive to both increases and decreases of the NAD+ -dependent desuccinylase, SIRT5. Although the succinyl group for succinylation is derived from metabolism, the effects of systematic variation of metabolism on mitochondrial succinylation are not known. Changes in succinylation of mitochondrial proteins following variations in metabolism were compared against the mitochondrial redox state as estimated by the mitochondrial NAD+ /NADH ratio using fluorescent probes. The ratio was decreased by reduced glycolysis and/or glutathione depletion (iodoacetic acid; 2-deoxyglucose), depressed tricarboxylic acid cycle activity (carboxyethyl ester of succinyl phosphonate), and impairment of electron transport (antimycin) or ATP synthase (oligomycin), while uncouplers of oxidative phosphorylation (carbonyl cyanide m-chlorophenyl hydrazine or tyrphostin) increased the NAD+ /NADH ratio. All of the conditions decreased succinylation. In contrast, reducing the oxygen from 20% to 2.4% increased succinylation. The results demonstrate that succinylation varies with metabolic states, is not correlated to the mitochondrial NAD+ /NADH ratio, and may help coordinate the response to metabolic challenge.


Mitochondrial Proteins/metabolism , Succinic Acid/metabolism , Animals , Cell Line, Tumor , Deoxyglucose/pharmacology , Mice , NAD/metabolism , Organophosphonates/metabolism , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , Succinates/metabolism
14.
Neurochem Res ; 42(6): 1636-1648, 2017 Jun.
Article En | MEDLINE | ID: mdl-28181072

Decades of research suggest that alterations in calcium are central to the pathophysiology of Alzheimer's Disease (AD). Highly reproducible changes in calcium dynamics occur in cells from patients with both genetic and non-genetic forms of AD relative to controls. The most robust change is an exaggerated release of calcium from internal stores. Detailed analysis of these changes in animal and cell models of the AD-causing presenilin mutations reveal robust changes in ryanodine receptors, inositol tris-phosphate receptors, calcium leak channels and store activated calcium entry. Similar anomalies in calcium result when AD-like changes in mitochondrial enzymes or oxidative stress are induced experimentally. The calcium abnormalities can be directly linked to the altered tau phosphorylation, amyloid precursor protein processing and synaptic dysfunction that are defining features of AD. A better understanding of these changes is required before using calcium abnormalities as therapeutic targets.


Alzheimer Disease/metabolism , Calcium/physiology , Mitochondria/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/genetics , Oxidative Stress/physiology
15.
PLoS One ; 11(9): e0160384, 2016.
Article En | MEDLINE | ID: mdl-27611087

Metabolic dysfunction is well-documented in Huntington's disease (HD). However, the link between the mutant huntingtin (mHTT) gene and the pathology is unknown. The tricarboxylic acid (TCA) cycle is the main metabolic pathway for the production of NADH for conversion to ATP via the electron transport chain (ETC). The objective of this study was to test for differences in enzyme activities, mRNAs and protein levels related to the TCA cycle between lymphoblasts from healthy subjects and from patients with HD. The experiments utilize the advantages of lymphoblasts to reveal new insights about HD. The large quantity of homogeneous cell populations permits multiple dynamic measures to be made on exactly comparable tissues. The activities of nine enzymes related to the TCA cycle and the expression of twenty-nine mRNAs encoding for these enzymes and enzyme complexes were measured. Cells were studied under baseline conditions and during metabolic stress. The results support our recent findings that the activities of the pyruvate dehydrogenase complex (PDHC) and succinate dehydrogenase (SDH) are elevated in HD. The data also show a large unexpected depression in MDH activities. Furthermore, message levels for isocitrate dehydrogenase 1 (IDH1) were markedly increased in in HD lymphoblasts and were responsive to treatments. The use of lymphoblasts allowed us to clarify that the reported decrease in aconitase activity in HD autopsy brains is likely due to secondary hypoxic effects. These results demonstrate the mRNA and enzymes of the TCA cycle are critical therapeutic targets that have been understudied in HD.


Citric Acid Cycle , Energy Metabolism , Huntington Disease/metabolism , Adult , Case-Control Studies , Cell Death/drug effects , Cell Death/genetics , Cell Line , Female , Gene Expression , Gene Expression Regulation, Enzymologic , Humans , Huntington Disease/genetics , Male , Middle Aged , Mitochondria/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium Cyanide/pharmacology , Stress, Physiological , Trinucleotide Repeats
16.
J Neurochem ; 139(5): 823-838, 2016 12.
Article En | MEDLINE | ID: mdl-27580471

Reductions in metabolism and excess oxidative stress are prevalent in multiple neurodegenerative diseases. The activity of the mitochondrial enzyme α-ketoglutarate dehydrogenase complex (KGDHC) appears central to these abnormalities. KGDHC is diminished in multiple neurodegenerative diseases. KGDHC can not only be rate limiting for NADH production and for substrate level phosphorylation, but is also a source of reactive oxygen species (ROS). The goal of these studies was to determine how changes in KGDHC modify baseline ROS, the ability to buffer ROS, baseline glutathionylation, calcium modulation and cell death in response to external oxidants. In vivo, reducing KGDHC with adeno virus diminished neurogenesis and increased oxidative stress. In vitro, treatments of short duration increased ROS and glutathionylation and enhanced the ability of the cells to diminish the ROS from added oxidants. However, long-term reductions lessened the ability to diminish ROS, diminished glutathionylation and exaggerated oxidant-induced changes in calcium and cell death. Increasing KGDHC enhanced the ability of the cells to diminish externally added ROS and protected against oxidant-induced changes in calcium and cell death. The results suggest that brief periods of diminished KGDHC are protective, while prolonged reductions are harmful. Furthermore, elevated KGDHC activities are protective. Thus, mitogenic therapies that increase KGDHC may be beneficial in neurodegenerative diseases. Read the Editorial Highlight for this article on Page 689.


Ketoglutarate Dehydrogenase Complex/deficiency , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/pathology
17.
Neurochem Int ; 96: 32-45, 2016 06.
Article En | MEDLINE | ID: mdl-26923918

Brain activities of the mitochondrial enzyme α-ketoglutarate dehydrogenase complex (KGDHC) are reduced in Alzheimer's disease and other age-related neurodegenerative disorders. The goal of the present study was to test the consequences of mild impairment of KGDHC on the structure, protein signaling and dynamics (mitophagy, fusion, fission, biogenesis) of the mitochondria. Inhibition of KGDHC reduced its in situ activity by 23-53% in human neuroblastoma SH-SY5Y cells, but neither altered the mitochondrial membrane potential nor the ATP levels at any tested time-points. The attenuated KGDHC activity increased translocation of dynamin-related protein-1 (Drp1) and microtubule-associated protein 1A/1B-light chain 3 (LC3) from the cytosol to the mitochondria, and promoted mitochondrial cytochrome c release. Inhibition of KGDHC also increased the negative surface charges (anionic phospholipids as assessed by Annexin V binding) on the mitochondria. Morphological assessments of the mitochondria revealed increased fission and mitophagy. Taken together, our results suggest the existence of the regulation of the mitochondrial dynamism including fission and fusion by the mitochondrial KGDHC activity via the involvement of the cytosolic and mitochondrial protein signaling molecules. A better understanding of the link among mild impairment of metabolism, induction of mitophagy/autophagy and altered protein signaling will help to identify new mechanisms of neurodegeneration and reveal potential new therapeutic approaches.


Alzheimer Disease/enzymology , Autophagy/physiology , Intracellular Fluid/enzymology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/enzymology , Alzheimer Disease/pathology , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Humans , Intracellular Fluid/drug effects , Mitochondria/drug effects , Organophosphonates/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Succinates/pharmacology
18.
Ann N Y Acad Sci ; 1367(1): 21-30, 2016 03.
Article En | MEDLINE | ID: mdl-26971083

The earliest and perhaps best example of an interaction between nutrition and dementia is related to thiamine (vitamin B1). Throughout the last century, research showed that thiamine deficiency is associated with neurological problems, including cognitive deficits and encephalopathy. Multiple similarities exist between classical thiamine deficiency and Alzheimer's disease (AD) in that both are associated with cognitive deficits and reductions in brain glucose metabolism. Thiamine-dependent enzymes are critical components of glucose metabolism that are reduced in the brains of AD patients and by thiamine decline, and a decrease in their levels could account for the reduction in glucose metabolism. In preclinical models, reduced thiamine can drive AD-like abnormalities, including memory deficits, neuritic plaques, and hyperphosphorylation of tau. Furthermore, excess thiamine diminishes AD-like pathologies. In addition to dietary deficits, drugs or other manipulations that interfere with thiamine absorption can cause thiamine deficiency. Elucidating the reasons why the brains of AD patients are functionally thiamine deficient and determining the effects of thiamine restoration may provide critical information to help treat patients with AD.


Dementia/drug therapy , Dementia/metabolism , Thiamine Deficiency/drug therapy , Thiamine Deficiency/metabolism , Thiamine/administration & dosage , Thiamine/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Dementia/diagnosis , Glucose/metabolism , Humans , Randomized Controlled Trials as Topic/methods , Thiamine Deficiency/diagnosis
19.
Neurochem Res ; 40(12): 2557-69, 2015 Dec.
Article En | MEDLINE | ID: mdl-26077923

Diminished glucose metabolism accompanies many neurodegenerative diseases including Alzheimer's disease. An understanding of the relation of these metabolic changes to the disease will enable development of novel therapeutic strategies. Following a metabolic challenge, cells generally conserve energy to preserve viability. This requires activation of many cellular repair/regenerative processes such as mitophagy/autophagy and fusion/fission. These responses may diminish cell function in the long term. Prolonged fission induces mitophagy/autophagy which promotes repair but if prolonged progresses to mitochondrial degradation. Abnormal glucose metabolism alters protein signaling including the release of proteins from the mitochondria or migration of proteins from the cytosol to the mitochondria or nucleus. This overview provides an insight into the different mechanisms of autophagy/mitophagy and mitochondrial dynamics in response to the diminished metabolism that occurs with diseases, especially neurodegenerative diseases such as Alzheimer's disease. The review discusses multiple aspects of mitochondrial responses including different signaling proteins and pathways of mitophagy and mitochondrial biogenesis. Improving cellular bioenergetics and mitochondrial dynamics will alter protein signaling and improve cellular/mitochondrial repair and regeneration. An understanding of these changes will suggest new therapeutic strategies.


Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Autophagy , Glucose/metabolism , Mitophagy , Alzheimer Disease/pathology , Animals , Humans , Mitochondria/metabolism
20.
J Neuropathol Exp Neurol ; 74(6): 527-37, 2015 Jun.
Article En | MEDLINE | ID: mdl-25978848

Glucose metabolism is reduced in the brains of patients with Huntington disease (HD). The mechanisms underlying this deficit, its link to the pathology of the disease, and the vulnerability of the striatum in HD remain unknown. Abnormalities in some of the key mitochondrial enzymes involved in glucose metabolism, including the pyruvate dehydrogenase complex (PDHC) and the tricarboxylic acid (TCA) cycle, may contribute to these deficits. Here, activities for these enzymes and select protein levels were measured in human postmortem cortex and in striatum and cortex of an HD mouse model (Q175); mRNA levels encoding for these enzymes were also measured in the Q175 mouse cortex. The activities of PDHC and nearly all of the TCA cycle enzymes were dramatically lower (-50% to 90%) in humans than in mice. The activity of succinate dehydrogenase increased with HD in human (35%) and mouse (23%) cortex. No other changes were detected in the human HD cortex or mouse striatum. In Q175 cortex, there were increased activities of PDHC (+12%) and aconitase (+32%). Increased mRNA levels for succinyl thiokinase (+88%) and isocitrate dehydrogenase (+64%) suggested an upregulation of the TCA cycle. These patterns of change differ from those reported in other diseases, which may offer unique metabolic therapeutic opportunities for HD patients.


Cerebral Cortex/enzymology , Citric Acid Cycle/physiology , Corpus Striatum/enzymology , Huntington Disease , Ketoglutarate Dehydrogenase Complex/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Aged , Aged, 80 and over , Animals , Dihydrolipoamide Dehydrogenase/genetics , Dihydrolipoamide Dehydrogenase/metabolism , Disease Models, Animal , Female , Humans , Huntington Disease/enzymology , Huntington Disease/pathology , Huntington Disease/physiopathology , Ketoglutarate Dehydrogenase Complex/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Mutation/genetics , Postmortem Changes , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , Retrospective Studies
...