51
|
Avgerinos KI, Egan JM, Mattson MP, Kapogiannis D. Medium Chain Triglycerides induce mild ketosis and may improve cognition in Alzheimer's disease. A systematic review and meta-analysis of human studies. Ageing Res Rev 2020; 58:101001. [PMID: 31870908 DOI: 10.1016/j.arr.2019.101001] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 11/15/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION/AIM The brain in Alzheimer's disease shows glucose hypometabolism but may utilize ketones for energy production. Ketone levels can potentially be boosted through oral intake of Medium Chain Triglycerides (MCTs). The aim of this meta-analysis is to investigate the effect of MCTs on peripheral ketone levels and cognitive performance in patients with mild cognitive impairment and Alzheimer's disease. METHODS Medline, Scopus and Web of Science were searched for literature up to March 1, 2019. Meta-analyses were performed by implementing continuous random-effects models and outcomes were reported as weighted Mean Differences (MDs) or Standardized Mean Differences (SMDs). RESULTS Twelve records (422 participants) were included. Meta-analysis of RCTs showed that, compared with placebo, MCTs elevated beta-hydroxybutyrate [MD = 0.355; 95 % CI (0.286, 0.424), I2 = 0 %], showed a trend towards cognitive improvement on ADAS-Cog [MD = -0.539; 95% CI (-1.239, -0.161), I2 = 0 %], and significantly improved cognition on a combined measure (ADAS-Cog with MMSE) [SMD = -0.289; 95 % CI (-0.551, -0.027), I2 = 0 %]. CONCLUSIONS In this meta-analysis, we demonstrated that MCTs can induce mild ketosis and may improve cognition in patients with mild cognitive impairment and Alzheimer's disease. However, risk of bias of existing studies necessitates future trials.
Collapse
|
52
|
Morris G, Puri BK, Carvalho A, Maes M, Berk M, Ruusunen A, Olive L. Induced Ketosis as a Treatment for Neuroprogressive Disorders: Food for Thought? Int J Neuropsychopharmacol 2020; 23:366-384. [PMID: 32034911 PMCID: PMC7311648 DOI: 10.1093/ijnp/pyaa008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/05/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Induced ketosis (or ketone body ingestion) can ameliorate several changes associated with neuroprogressive disorders, including schizophrenia, bipolar disorder, and major depressive disorder. Thus, the effects of glucose hypometabolism can be bypassed through the entry of beta-hydroxybutyrate, providing an alternative source of energy to glucose. The weight of evidence suggests that induced ketosis reduces levels of oxidative stress, mitochondrial dysfunction, and inflammation-core features of the above disorders. There are also data to suggest that induced ketosis may be able to target other molecules and signaling pathways whose levels and/or activity are also known to be abnormal in at least some patients suffering from these illnesses such as peroxisome proliferator-activated receptors, increased activity of the Kelch-like ECH-associated protein/nuclear factor erythroid 2-related factor 2, Sirtuin-1 nuclear factor-κB p65, and nicotinamide adenine dinucleotide (NAD). This review explains the mechanisms by which induced ketosis might reduce mitochondrial dysfunction, inflammation, and oxidative stress in neuropsychiatric disorders and ameliorate abnormal levels of molecules and signaling pathways that also appear to contribute to the pathophysiology of these illnesses. This review also examines safety data relating to induced ketosis over the long term and discusses the design of future studies.
Collapse
Affiliation(s)
- Gerwyn Morris
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| | - Basant K Puri
- C.A.R., Cambridge, United Kingdom,Hammersmith Hospital, London, United Kingdom
| | - Andre Carvalho
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Maes
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia,Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia,Correspondence: Michael Berk, PO Box 281 Geelong, Victoria 3220 Australia ()
| | - Anu Ruusunen
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| | - Lisa Olive
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| |
Collapse
|
53
|
A Dietary Ketone Ester Normalizes Abnormal Behavior in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21031044. [PMID: 32033248 PMCID: PMC7036949 DOI: 10.3390/ijms21031044] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/22/2020] [Accepted: 01/29/2020] [Indexed: 12/02/2022] Open
Abstract
Because of a decreased sensitivity toward insulin, a key regulator of pyruvate dehydrogenase (PDH), Alzheimer’s patients have lower brain glucose utilization with reductions in Tricarboxylic Acid (TCA) cycle metabolites such as citrate, a precursor to n-acetyl-aspartate. In the 3xTgAd mouse model of Alzheimer’s disease (AD), aging mice also demonstrate low brain glucose metabolism. Ketone metabolism can overcome PDH inhibition and restore TCA cycle metabolites, thereby enhancing amino acid biosynthesis. A ketone ester of d-β-hydroxybutyrate was incorporated into a diet (Ket) and fed to 3xTgAd mice. A control group was fed a calorically matched diet (Cho). At 15 months of age, the exploratory and avoidance-related behavior patterns of the mice were evaluated. At 16.5 months of age, the animals were euthanized, and their hippocampi were analyzed for citrate, α-ketoglutarate, and amino acids. In the hippocampi of the Ket-fed mice, there were higher concentrations of citrate and α-ketoglutarate as well as higher concentrations of glutamate, aspartate and n-acetyl-aspartate compared with controls. There were positive associations between (1) concentrations of aspartate and n-acetyl-aspartate (n = 14, R = 0.9327), and (2) α-ketoglutarate and glutamate (n = 14, R = 0.8521) in animals maintained on either diet. Hippocampal n-acetyl-aspartate predicted the outcome of several exploratory and avoidance-related behaviors. Ketosis restored citrate and α-ketoglutarate in the hippocampi of aging mice. Higher concentrations of n-acetyl-aspartate corresponded with greater exploratory activity and reduced avoidance-related behavior.
Collapse
|
54
|
Versele R, Corsi M, Fuso A, Sevin E, Businaro R, Gosselet F, Fenart L, Candela P. Ketone Bodies Promote Amyloid-β 1-40 Clearance in a Human in Vitro Blood-Brain Barrier Model. Int J Mol Sci 2020; 21:E934. [PMID: 32023814 PMCID: PMC7037612 DOI: 10.3390/ijms21030934] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the abnormal accumulation of amyloid-β (Aβ) peptides in the brain. The pathological process has not yet been clarified, although dysfunctional transport of Aβ across the blood-brain barrier (BBB) appears to be integral to disease development. At present, no effective therapeutic treatment against AD exists, and the adoption of a ketogenic diet (KD) or ketone body (KB) supplements have been investigated as potential new therapeutic approaches. Despite experimental evidence supporting the hypothesis that KBs reduce the Aβ load in the AD brain, little information is available about the effect of KBs on BBB and their effect on Aβ transport. Therefore, we used a human in vitro BBB model, brain-like endothelial cells (BLECs), to investigate the effect of KBs on the BBB and on Aβ transport. Our results show that KBs do not modify BBB integrity and do not cause toxicity to BLECs. Furthermore, the presence of KBs in the culture media was combined with higher MCT1 and GLUT1 protein levels in BLECs. In addition, KBs significantly enhanced the protein levels of LRP1, P-gp, and PICALM, described to be involved in Aβ clearance. Finally, the combined use of KBs promotes Aβ efflux across the BBB. Inhibition experiments demonstrated the involvement of LRP1 and P-gp in the efflux. This work provides evidence that KBs promote Aβ clearance from the brain to blood in addition to exciting perspectives for studying the use of KBs in therapeutic approaches.
Collapse
Affiliation(s)
- Romain Versele
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Mariangela Corsi
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Dip. di Chirurgia “P. Valdoni”, Via A. Scarpa 16, 00161 Rome, Italy;
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Laurence Fenart
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Pietra Candela
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| |
Collapse
|
55
|
Kapogiannis D, Avgerinos KI. Brain glucose and ketone utilization in brain aging and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:79-110. [PMID: 32739015 PMCID: PMC9989941 DOI: 10.1016/bs.irn.2020.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To meet its high energy demands, the brain mostly utilizes glucose. However, the brain has evolved to exploit additional fuels, such as ketones, especially during prolonged fasting. With aging and neurodegenerative diseases (NDDs), the brain becomes inefficient at utilizing glucose due to changes in glia and neurons that involve glucose transport, glycolytic and Krebs cycle enzyme activities, and insulin signaling. Positron emission tomography and magnetic resonance spectroscopy studies have identified glucose metabolism abnormalities in aging, Alzheimer's disease (AD) and other NDDs in vivo. Despite glucose hypometabolism, brain cells can utilize ketones efficiently, thereby providing a rationale for the development of therapeutic ketogenic interventions in AD and other NDDs. This review compares available ketogenic interventions and discusses the potential of the potent oral Ketone Ester for future therapeutic use in AD and other NDDs characterized by inefficient glucose utilization.
Collapse
Affiliation(s)
- Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.
| | - Konstantinos I Avgerinos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
56
|
Oligosaccharides from Morinda officinalis Slow the Progress of Aging Mice by Regulating the Key Microbiota-Metabolite Pairs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9306834. [PMID: 31929824 PMCID: PMC6942866 DOI: 10.1155/2019/9306834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/26/2019] [Accepted: 11/27/2019] [Indexed: 11/17/2022]
Abstract
The gut microbiota is considered an important factor in the progression of Alzheimer's disease (AD). Active research on the association between the metabolome and the gut microbiome is ongoing and can provide a large amount of beneficial information about the interactions between the microbiome and the metabolome. Previous studies have shown that the oligosaccharides from Morinda officinalis (OMO) can delay the progress of AD in model animals by regulating the diversity of the gut microbiome and metabolic components, and the correlation between the gut microbiome and metabolic components still needs to be further verified. This study applied a new two-level strategy to investigate and ensure the accuracy and consistency of the results. This strategy can be used to determine the association between the gut microbiome and serum metabolome in APP/PS1 transgenic mice and C57BL/6J male mice. The “4C0d-2 spp.-Cholesterol,” “CW040 spp.-L-valine,” “CW040 spp.-L-acetylcarnitine,” “RF39 spp.-L-valine,” “TM7-3 spp.-L-valine,” and “TM7-3 spp.-L-acetylcarnitine” associations among specific “microbiota-metabolite” pairs were further identified based on univariate and multivariate correlation analyses and functional analyses. The key relevant pairs were verified by an independent oligosaccharide intervention study, and the gut microbiome and serum metabolome of the OMO intervention group were similar to those of the normal group. The results indicate that OMO can significantly suppress Alzheimer's disease by regulating the key microbiota-metabolite pairs. Therefore, this two-level strategy is effective in identifying the principal correlations in large datasets obtained from combinations of multiomic studies and further enhancing our understanding of the correlation between the brain and gut in patients with AD.
Collapse
|
57
|
Camberos-Luna L, Massieu L. Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases. Neurochem Int 2019; 133:104614. [PMID: 31785349 DOI: 10.1016/j.neuint.2019.104614] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
The therapeutic use of ketone bodies (KB) against acute brain injury and neurodegenerative disorders has lately been suggested by many studies. Several mechanisms responsible for the protective action of KB have been described, including metabolic, anti-inflammatory and epigenetic. However, it is still not clear whether a specific mechanism of action can be associated with a particular neurological disorder. Different strategies to induce ketosis including the ketogenic diet (KD), caloric restriction (CR), intermittent fasting (IF), as well as the administration of medium chain triglycerides (MCTs), exogenous ketones or KB derivatives, have been used in animal models of brain injury and in humans. They have shown different degrees of success to prevent neuronal damage, motor alterations and cognitive decline. However, more investigation is needed in order to establish safe protocols for clinical application. Throughout the present review, we describe the different approaches that have been used to elevate blood KB and discuss their effectiveness considering their advantages and limitations, as tested in models of brain injury, neurodegeneration and clinical research. We also describe the mechanisms of action of KB in non-pathologic conditions and in association with their protective effect against neuronal damage in acute neurological disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucy Camberos-Luna
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| | - Lourdes Massieu
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| |
Collapse
|
58
|
Abstract
NAD+ is a pivotal metabolite involved in cellular bioenergetics, genomic stability, mitochondrial homeostasis, adaptive stress responses, and cell survival. Multiple NAD+-dependent enzymes are involved in synaptic plasticity and neuronal stress resistance. Here, we review emerging findings that reveal key roles for NAD+ and related metabolites in the adaptation of neurons to a wide range of physiological stressors and in counteracting processes in neurodegenerative diseases, such as those occurring in Alzheimer's, Parkinson's, and Huntington diseases, and amyotrophic lateral sclerosis. Advances in understanding the molecular and cellular mechanisms of NAD+-based neuronal resilience will lead to novel approaches for facilitating healthy brain aging and for the treatment of a range of neurological disorders.
Collapse
Affiliation(s)
- Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - David A Sinclair
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway; The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway.
| |
Collapse
|
59
|
Cenini G, Voos W. Mitochondria as Potential Targets in Alzheimer Disease Therapy: An Update. Front Pharmacol 2019; 10:902. [PMID: 31507410 PMCID: PMC6716473 DOI: 10.3389/fphar.2019.00902] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer disease (AD) is a progressive and deleterious neurodegenerative disorder that affects mostly the elderly population. At the moment, no effective treatments are available in the market, making the whole situation a compelling challenge for societies worldwide. Recently, novel mechanisms have been proposed to explain the etiology of this disease leading to the new concept that AD is a multifactor pathology. Among others, the function of mitochondria has been considered as one of the intracellular processes severely compromised in AD since the early stages and likely represents a common feature of many neurodegenerative diseases. Many mitochondrial parameters decline already during the aging, reaching an extensive functional failure concomitant with the onset of neurodegenerative conditions, although the exact timeline of these events is still unclear. Thereby, it is not surprising that mitochondria have been already considered as therapeutic targets in neurodegenerative diseases including AD. Together with an overview of the role of mitochondrial dysfunction, this review examines the pros and cons of the tested therapeutic approaches targeting mitochondria in the context of AD. Since mitochondrial therapies in AD have shown different degrees of progress, it is imperative to perform a detailed analysis of the significance of mitochondrial deterioration in AD and of a pharmacological treatment at this level. This step would be very important for the field, as an effective drug treatment in AD is still missing and new therapeutic concepts are urgently needed.
Collapse
Affiliation(s)
- Giovanna Cenini
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Wolfgang Voos
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
60
|
Ketogenic Diet in Alzheimer's Disease. Int J Mol Sci 2019; 20:ijms20163892. [PMID: 31405021 PMCID: PMC6720297 DOI: 10.3390/ijms20163892] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 01/18/2023] Open
Abstract
At present, the prevalence of Alzheimer's disease, a devastating neurodegenerative disorder, is increasing. Although the mechanism of the underlying pathology is not fully uncovered, in the last years, there has been significant progress in its understanding. This includes: Progressive deposition of amyloid β-peptides in amyloid plaques and hyperphosphorylated tau protein in intracellular as neurofibrillary tangles; neuronal loss; and impaired glucose metabolism. Due to a lack of effective prevention and treatment strategy, emerging evidence suggests that dietary and metabolic interventions could potentially target these issues. The ketogenic diet is a very high-fat, low-carbohydrate diet, which has a fasting-like effect bringing the body into a state of ketosis. The presence of ketone bodies has a neuroprotective impact on aging brain cells. Moreover, their production may enhance mitochondrial function, reduce the expression of inflammatory and apoptotic mediators. Thus, it has gained interest as a potential therapy for neurodegenerative disorders like Alzheimer's disease. This review aims to examine the role of the ketogenic diet in Alzheimer's disease progression and to outline specific aspects of the nutritional profile providing a rationale for the implementation of dietary interventions as a therapeutic strategy for Alzheimer's disease.
Collapse
|
61
|
Wood TR, Stubbs BJ, Juul SE. Exogenous Ketone Bodies as Promising Neuroprotective Agents for Developmental Brain Injury. Dev Neurosci 2019; 40:451-462. [PMID: 31085911 DOI: 10.1159/000499563] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/12/2019] [Indexed: 11/19/2022] Open
Abstract
Ketone bodies are a promising area of neuroprotection research that may be ideally suited to the injured newborn. During normal development, the human infant is in significant ketosis for at least the first week of life. Ketone uptake and metabolism is upregulated in the both the fetus and neonate, with ketone bodies providing at least 10% of cerebral metabolic energy requirements, as well as being the preferred precursors for the synthesis of fatty acids and cholesterol. At the same time, ketone bodies have been shown to have multiple neuroprotective effects, including being anticonvulsant, decreasing oxidative stress and inflammation, and epigenetically upregulating the production of neurotrophic factors. While ketogenic diets and exogenous ketosis are largely being investigated in the setting of adult brain injury, the adaptation of the neonate to ketosis suggests that developmental brain injury may be the area most suited to the use of ketones for neuroprotection. Here, we describe the mechanisms by which ketone bodies exert their neuroprotective effects, and how these may translate to benefits within each of the phases of neonatal asphyxial brain injury.
Collapse
Affiliation(s)
- Thomas R Wood
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA, .,Institute for Human and Machine Cognition, Pensacola, Florida, USA,
| | - Brianna J Stubbs
- HVMN Inc., San Francisco, California, USA.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
62
|
Frost PS, Barros-Aragão F, da Silva RT, Venancio A, Matias I, Lyra E Silva NM, Kincheski GC, Pimentel-Coelho PM, De Felice FG, Gomes FCA, Ferreira ST, Figueiredo CP, Clarke JR. Neonatal infection leads to increased susceptibility to Aβ oligomer-induced brain inflammation, synapse loss and cognitive impairment in mice. Cell Death Dis 2019; 10:323. [PMID: 30975983 PMCID: PMC6459845 DOI: 10.1038/s41419-019-1529-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/22/2022]
Abstract
Harmful environmental stimuli during critical stages of development can profoundly affect behavior and susceptibility to diseases. Alzheimer disease (AD) is the most frequent neurodegenerative disease, and evidence suggest that inflammatory conditions act cumulatively, contributing to disease onset. Here we investigated whether infection early in life can contribute to synapse damage and cognitive impairment induced by amyloid-β oligomers (AβOs), neurotoxins found in AD brains. To this end, wild-type mice were subjected to neonatal (post-natal day 4) infection by Escherichia coli (1 × 104 CFU/g), the main cause of infection in low-birth-weight premature infants in the US. E. coli infection caused a transient inflammatory response in the mouse brain starting shortly after infection. Although infected mice performed normally in behavioral tasks in adulthood, they showed increased susceptibility to synapse damage and memory impairment induced by low doses of AβOs (1 pmol; intracerebroventricular) in the novel object recognition paradigm. Using in vitro and in vivo approaches, we show that microglial cells from E. coli-infected mice undergo exacerbated activation when exposed to low doses of AβOs. In addition, treatment of infected pups with minocycline, an antibiotic that inhibits microglial pro-inflammatory polarization, normalized microglial response to AβOs and restored normal susceptibility of mice to oligomer-induced cognitive impairment. Interestingly, mice infected with by E. coli (1 × 104 CFU/g) during adolescence (post-natal day 21) or adulthood (post-natal day 60) showed normal cognitive performance even in the presence of AβOs (1 pmol), suggesting that only infections at critical stages of development may lead to increased susceptibility to amyloid-β-induced toxicity. Altogether, our findings suggest that neonatal infections can modulate microglial response to AβOs into adulthood, thus contributing to amyloid-β-induced synapse damage and cognitive impairment.
Collapse
Affiliation(s)
- Paula S Frost
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Fernanda Barros-Aragão
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Rachel T da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Aline Venancio
- Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Isadora Matias
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Natalia M Lyra E Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Grasielle C Kincheski
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Pedro M Pimentel-Coelho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.,Department of Psychiatry, Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Flávia C A Gomes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil.
| |
Collapse
|
63
|
Cytoplasmic and Mitochondrial NADPH-Coupled Redox Systems in the Regulation of Aging. Nutrients 2019; 11:nu11030504. [PMID: 30818813 PMCID: PMC6471790 DOI: 10.3390/nu11030504] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/20/2022] Open
Abstract
The reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) protects against redox stress by providing reducing equivalents to antioxidants such as glutathione and thioredoxin. NADPH levels decline with aging in several tissues, but whether this is a major driving force for the aging process has not been well established. Global or neural overexpression of several cytoplasmic enzymes that synthesize NADPH have been shown to extend lifespan in model organisms such as Drosophila suggesting a positive relationship between cytoplasmic NADPH levels and longevity. Mitochondrial NADPH plays an important role in the protection against redox stress and cell death and mitochondrial NADPH-utilizing thioredoxin reductase 2 levels correlate with species longevity in cells from rodents and primates. Mitochondrial NADPH shuttles allow for some NADPH flux between the cytoplasm and mitochondria. Since a decline of nicotinamide adenine dinucleotide (NAD+) is linked with aging and because NADP+ is exclusively synthesized from NAD+ by cytoplasmic and mitochondrial NAD+ kinases, a decline in the cytoplasmic or mitochondrial NADPH pool may also contribute to the aging process. Therefore pro-longevity therapies should aim to maintain the levels of both NAD+ and NADPH in aging tissues.
Collapse
|
64
|
Jackson TC, Kochanek PM. A New Vision for Therapeutic Hypothermia in the Era of Targeted Temperature Management: A Speculative Synthesis. Ther Hypothermia Temp Manag 2019; 9:13-47. [PMID: 30802174 PMCID: PMC6434603 DOI: 10.1089/ther.2019.0001] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three decades of animal studies have reproducibly shown that hypothermia is profoundly cerebroprotective during or after a central nervous system (CNS) insult. The success of hypothermia in preclinical acute brain injury has not only fostered continued interest in research on the classic secondary injury mechanisms that are prevented or blunted by hypothermia but has also sparked a surge of new interest in elucidating beneficial signaling molecules that are increased by cooling. Ironically, while research into cold-induced neuroprotection is enjoying newfound interest in chronic neurodegenerative disease, conversely, the scope of the utility of therapeutic hypothermia (TH) across the field of acute brain injury is somewhat controversial and remains to be fully defined. This has led to the era of Targeted Temperature Management, which emphasizes a wider range of temperatures (33–36°C) showing benefit in acute brain injury. In this comprehensive review, we focus on our current understandings of the novel neuroprotective mechanisms activated by TH, and discuss the critical importance of developmental age germane to its clinical efficacy. We review emerging data on four cold stress hormones and three cold shock proteins that have generated new interest in hypothermia in the field of CNS injury, to create a framework for new frontiers in TH research. We make the case that further elucidation of novel cold responsive pathways might lead to major breakthroughs in the treatment of acute brain injury, chronic neurological diseases, and have broad potential implications for medicines of the distant future, including scenarios such as the prevention of adverse effects of long-duration spaceflight, among others. Finally, we introduce several new phrases that readily summarize the essence of the major concepts outlined by this review—namely, Ultramild Hypothermia, the “Responsivity of Cold Stress Pathways,” and “Hypothermia in a Syringe.”
Collapse
Affiliation(s)
- Travis C Jackson
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
65
|
Abstract
Glucose is the long-established, obligatory fuel for brain that fulfills many critical functions, including ATP production, oxidative stress management, and synthesis of neurotransmitters, neuromodulators, and structural components. Neuronal glucose oxidation exceeds that in astrocytes, but both rates increase in direct proportion to excitatory neurotransmission; signaling and metabolism are closely coupled at the local level. Exact details of neuron-astrocyte glutamate-glutamine cycling remain to be established, and the specific roles of glucose and lactate in the cellular energetics of these processes are debated. Glycolysis is preferentially upregulated during brain activation even though oxygen availability is sufficient (aerobic glycolysis). Three major pathways, glycolysis, pentose phosphate shunt, and glycogen turnover, contribute to utilization of glucose in excess of oxygen, and adrenergic regulation of aerobic glycolysis draws attention to astrocytic metabolism, particularly glycogen turnover, which has a high impact on the oxygen-carbohydrate mismatch. Aerobic glycolysis is proposed to be predominant in young children and specific brain regions, but re-evaluation of data is necessary. Shuttling of glucose- and glycogen-derived lactate from astrocytes to neurons during activation, neurotransmission, and memory consolidation are controversial topics for which alternative mechanisms are proposed. Nutritional therapy and vagus nerve stimulation are translational bridges from metabolism to clinical treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas ; and Department of Cell Biology and Physiology, University of New Mexico , Albuquerque, New Mexico
| |
Collapse
|
66
|
Veech RL, Todd King M, Pawlosky R, Kashiwaya Y, Bradshaw PC, Curtis W. The "great" controlling nucleotide coenzymes. IUBMB Life 2019; 71:565-579. [PMID: 30624851 PMCID: PMC6850382 DOI: 10.1002/iub.1997] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
Nucleotide coenzymes dot the map of metabolic pathways providing energy to drive the reactions of the pathway and play an important role in regulating and controlling energy metabolism through their shared potential energy, which is widely unobserved due to the paradox that the energy in the coenzyme pools cannot be determined from the concentration of the coenzyme couples. The potential energy of the nucleotide couples in the mitochondria or the cytoplasm is expressed in the enzyme reactions in which they take part. The energy in these couples, [NAD+]/[NADH], [NADP+]/[NADPH], [acetyl CoA]/[CoA], and [ATP]/[ADP]x[Pi], regulates energy metabolism. The energy contained in the couples can be altered by suppling energy equivalents in the form of ketones, such as, D-β-hydroxybutyrate to overcome insulin resistance, to restore antioxidants capacity, to form potential treatments for Alzheimer's and Parkinson's diseases, to enhance life span, and to increase physiological performance. © 2019 IUBMB Life, 71(5):565-579, 2019.
Collapse
Affiliation(s)
- Richard L Veech
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Michael Todd King
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Robert Pawlosky
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | | | - Patrick C Bradshaw
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - William Curtis
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| |
Collapse
|
67
|
Titov VN, Ivanov GA, Antonov AM. [Laurine fatty acids, medium fatty acids and triglycerides, hyperlipidemia, resistance to insulin, prevention of atherosclerosis and ateromatosis.]. Klin Lab Diagn 2019; 64:68-77. [PMID: 30917246 DOI: 10.18821/0869-2084-2019-64-2-68-77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/25/2018] [Indexed: 06/09/2023]
Abstract
Although the biochemistry of the positive effects of medium-chain fatty acids (FA) and triglycerides (TG) of the same name in vivo is not fully understood, food enriched with medium-chain LC and the same TG is effective in patients with type I diabetes, insulin resistance syndrome and in neurodegenerative pathology. Lauric C12 LC is half the FA in coconut oil. Residents of southeast Asia with constant use of coconut oil, have a low level of diseases of the cardiovascular system in the population. With a regulatory intake with food C12:0 laurin FA formed moderate ketosis and neuroprotective effect. Unlike long-chain LC, medium-chain TG cells are not deposited either in visceral fat cells, or in insulin-dependent adipocytes. Medium-chain fatty acids rapidly oxidize mitochondria; the formation of acetyl-CoA cells is used to form ketone bodies, activating thermogenesis in orange and brown adipocytes. Experiments with animals and observations in the clinic showed that taking medium-chain TG with food is more physiological than long-chain oils. This significantly increases the level of cholesterol in high-density lipoproteins. Food enriched with medium chain TG is optimal for increasing the ketone content in blood plasma, cerebrospinal fluid without limiting the carbohydrate content in food. The formation of excess ketone bodies by cells can be achieved by activating the metabolic transformations of medium-chain FAs, without fasting and preserving carbohydrates in food. Coconut oil has a positive effect on the cardiovascular system, preventing the formation of atherosclerosis and atheromatosis. Effective in the prevention of the pathology of the cardiovascular system is a decrease in food amounts of palmitic acid, an increase in oleic acid, polyene FA with a simultaneous increase in the proportion of medium-chain FA.
Collapse
Affiliation(s)
- V N Titov
- FGBU National Medical Research Center for Cardiology of the Ministry of Health of Russia, 121552, Moscow
| | - G A Ivanov
- OOO «Biolaboratory», Moscow, territory of the innovation center «Skolkovo»
| | - A M Antonov
- FGAOU VO "Northern (Arctic) Federal University, M.V. Lomonosov", Arkhangelsk
| |
Collapse
|
68
|
Kraeuter AK, Guest PC, Sarnyai Z. The Therapeutic Potential of Ketogenic Diet Throughout Life: Focus on Metabolic, Neurodevelopmental and Neurodegenerative Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:77-101. [PMID: 31493223 DOI: 10.1007/978-3-030-25650-0_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter reviews the efficacy of the ketogenic diet in a variety of neurodegenerative, neurodevelopmental and metabolic conditions throughout different stages of life. It describes conditions affecting children, metabolic disorders in adults and disorderrs affecting the elderly. We have focused on application of the ketogenic diet in clinical studies and in preclinical models and discuss the benefits and negative aspects of the diet. Finally, we highlight the need for further research in this area with a view of discovering novel mechanistic targets of the ketogenic diet, as a means of maximising the potential benefits/risks ratio.
Collapse
Affiliation(s)
- Ann-Katrin Kraeuter
- Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.,Discipline of Biomedicine, College of Public Health, Medicine and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Zoltan Sarnyai
- Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia. .,Discipline of Biomedicine, College of Public Health, Medicine and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.
| |
Collapse
|
69
|
Elamin M, Ruskin DN, Masino SA, Sacchetti P. Ketogenic Diet Modulates NAD +-Dependent Enzymes and Reduces DNA Damage in Hippocampus. Front Cell Neurosci 2018; 12:263. [PMID: 30214397 PMCID: PMC6125375 DOI: 10.3389/fncel.2018.00263] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022] Open
Abstract
The ketogenic diet's (KD) anti-seizure effects have long been documented. Recently, its therapeutic potential in multiple neurodegenerative and neurodevelopmental disorders has emerged. Yet experimental evidence for a fundamental mechanism underlying beneficial effects across numerous diseases remains lacking. We previously showed that feeding rats a KD produced an early (within 2 days) and persistent elevation of hippocampal nicotinamide adenine dinucleotide+ (NAD+), an essential metabolic coenzyme and signaling molecule. NAD+ is a marker of cellular health and a substrate for enzymes implicated in longevity and DNA damage repair such as sirtuins and poly-ADP ribose polymerase-1 (PARP-1). As a result, activation of NAD+-dependent enzymes' downstream pathways could be the origin of KD's broad beneficial effects. Here rats were fed ad libitum regular chow or KD for 2 days or 3 weeks and the levels of hippocampal sirtuins, PARP-1, and the oxidative DNA damage marker 8-hydroxy-2'-deoxyguanosine were quantified. We found a significant immediate and persistent increase in the collective activity of nuclear sirtuin enzymes, and a significant augmentation of Sirt1 mRNA at 2 days. Levels of PARP-1 and 8-hydroxy-2'-deoxyguanosine decreased after 2 days of treatment and further declined at 3 weeks. Our data show that a KD can rapidly modulate energy metabolism by acting on NAD+-dependent enzymes and their downstream pathways. Thus, therapy with a KD can potentially enhance brain health and increase overall healthspan via NAD+-related mechanisms that render cells more resilient against DNA damage and a host of metabolic, epileptic, neurodegenerative, or neurodevelopmental insults.
Collapse
Affiliation(s)
- Marwa Elamin
- Graduate Program in Neuroscience, Department of Biology, University of Hartford, West Hartford, CT, United States
| | - David N Ruskin
- Neuroscience Program and Psychology Department, Trinity College, Hartford, CT, United States
| | - Susan A Masino
- Neuroscience Program and Psychology Department, Trinity College, Hartford, CT, United States
| | - Paola Sacchetti
- Graduate Program in Neuroscience, Department of Biology, University of Hartford, West Hartford, CT, United States
| |
Collapse
|
70
|
Zhang W, Hu X, Zhou W, Tam KY. Liquid Chromatography-Tandem Mass Spectrometry Method Revealed that Lung Cancer Cells Exhibited Distinct Metabolite Profiles upon the Treatment with Different Pyruvate Dehydrogenase Kinase Inhibitors. J Proteome Res 2018; 17:3012-3021. [PMID: 30028142 DOI: 10.1021/acs.jproteome.8b00184] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pyruvate dehydrogenase kinases (PDKs) dominate the critical switch between mitochondria-based respiration and cytoplasm-based glycolysis by controlling pyruvate dehydrogenase (PDH) activity. Up-regulated PDKs play a great role in the Warburg effect in cancer cells and accordingly present a therapeutic target. Dichloroacetate (DCA) and AZD7545 are the two most-well-known PDK inhibitors exhibiting distinct pharmacological profiles. DCA showed anticancer effects in various preclinical models and clinical studies, while the primary preclinical indication of AZD7545 was on the improvement of glucose control in type II diabetes. Little, if any, study has been undertaken the elucidation of the effects of PDK inhibition on the metabolites in the tricarboxylic acid (TCA) cycle. Herein, the metabolite alterations of lung cancer cells (A549) upon the treatment with PDK inhibitors were studied using a reliable liquid-chromatography-based tandem mass spectrometry method. The developed method was validated for quantification of all common glycolysis and TCA cycle catabolites with good sensitivity and reproducibility, including glucose, pyruvate, lactate, acetyl coenzyme A, citrate, α-ketoglutarate, fumarate, succinate, malate, and oxaloacetate. Our results suggested that A549 cells exhibited distinct metabolite profiles following the treatment with DCA or AZD7545, which may reflect the different pharmacological indications of these two drugs.
Collapse
Affiliation(s)
- Wen Zhang
- Faculty of Health Sciences , University of Macau , Taipa , Macau, China
| | - Xiaohui Hu
- Faculty of Health Sciences , University of Macau , Taipa , Macau, China
| | - Wei Zhou
- Faculty of Health Sciences , University of Macau , Taipa , Macau, China
| | - Kin Yip Tam
- Faculty of Health Sciences , University of Macau , Taipa , Macau, China
| |
Collapse
|
71
|
Ronowska A, Szutowicz A, Bielarczyk H, Gul-Hinc S, Klimaszewska-Łata J, Dyś A, Zyśk M, Jankowska-Kulawy A. The Regulatory Effects of Acetyl-CoA Distribution in the Healthy and Diseased Brain. Front Cell Neurosci 2018; 12:169. [PMID: 30050410 PMCID: PMC6052899 DOI: 10.3389/fncel.2018.00169] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/31/2018] [Indexed: 12/25/2022] Open
Abstract
Brain neurons, to support their neurotransmitter functions, require a several times higher supply of glucose than non-excitable cells. Pyruvate, the end product of glycolysis, through pyruvate dehydrogenase complex reaction, is a principal source of acetyl-CoA, which is a direct energy substrate in all brain cells. Several neurodegenerative conditions result in the inhibition of pyruvate dehydrogenase and decrease of acetyl-CoA synthesis in mitochondria. This attenuates metabolic flux through TCA in the mitochondria, yielding energy deficits and inhibition of diverse synthetic acetylation reactions in all neuronal sub-compartments. The acetyl-CoA concentrations in neuronal mitochondrial and cytoplasmic compartments are in the range of 10 and 7 μmol/L, respectively. They appear to be from 2 to 20 times lower than acetyl-CoA Km values for carnitine acetyltransferase, acetyl-CoA carboxylase, aspartate acetyltransferase, choline acetyltransferase, sphingosine kinase 1 acetyltransferase, acetyl-CoA hydrolase, and acetyl-CoA acetyltransferase, respectively. Therefore, alterations in acetyl-CoA levels alone may significantly change the rates of metabolic fluxes through multiple acetylation reactions in brain cells in different physiologic and pathologic conditions. Such substrate-dependent alterations in cytoplasmic, endoplasmic reticulum or nuclear acetylations may directly affect ACh synthesis, protein acetylations, and gene expression. Thereby, acetyl-CoA may regulate the functional and adaptative properties of neuronal and non-neuronal brain cells. The excitotoxicity-evoked intracellular zinc excess hits several intracellular targets, yielding the collapse of energy balance and impairment of the functional and structural integrity of postsynaptic cholinergic neurons. Acute disruption of brain energy homeostasis activates slow accumulation of amyloid-β1-42 (Aβ). Extra and intracellular oligomeric deposits of Aβ affect diverse transporting and signaling pathways in neuronal cells. It may combine with multiple neurotoxic signals, aggravating their detrimental effects on neuronal cells. This review presents evidences that changes of intraneuronal levels and compartmentation of acetyl-CoA may contribute significantly to neurotoxic pathomechanisms of different neurodegenerative brain disorders.
Collapse
Affiliation(s)
- Anna Ronowska
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej Szutowicz
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Hanna Bielarczyk
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Sylwia Gul-Hinc
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Joanna Klimaszewska-Łata
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Dyś
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Marlena Zyśk
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
72
|
Duarte A, Santos M, Oliveira C, Moreira P. Brain insulin signalling, glucose metabolism and females' reproductive aging: A dangerous triad in Alzheimer's disease. Neuropharmacology 2018; 136:223-242. [DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
73
|
Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice. Sci Rep 2018; 8:6670. [PMID: 29703936 PMCID: PMC5923270 DOI: 10.1038/s41598-018-25190-5] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Neurovascular integrity, including cerebral blood flow (CBF) and blood-brain barrier (BBB) function, plays a major role in determining cognitive capability. Recent studies suggest that neurovascular integrity could be regulated by the gut microbiome. The purpose of the study was to identify if ketogenic diet (KD) intervention would alter gut microbiome and enhance neurovascular functions, and thus reduce risk for neurodegeneration in young healthy mice (12–14 weeks old). Here we show that with 16 weeks of KD, mice had significant increases in CBF and P-glycoprotein transports on BBB to facilitate clearance of amyloid-beta, a hallmark of Alzheimer’s disease (AD). These neurovascular enhancements were associated with reduced mechanistic target of rapamycin (mTOR) and increased endothelial nitric oxide synthase (eNOS) protein expressions. KD also increased the relative abundance of putatively beneficial gut microbiota (Akkermansia muciniphila and Lactobacillus), and reduced that of putatively pro-inflammatory taxa (Desulfovibrio and Turicibacter). We also observed that KD reduced blood glucose levels and body weight, and increased blood ketone levels, which might be associated with gut microbiome alteration. Our findings suggest that KD intervention started in the early stage may enhance brain vascular function, increase beneficial gut microbiota, improve metabolic profile, and reduce risk for AD.
Collapse
|
74
|
Veyrat-Durebex C, Reynier P, Procaccio V, Hergesheimer R, Corcia P, Andres CR, Blasco H. How Can a Ketogenic Diet Improve Motor Function? Front Mol Neurosci 2018; 11:15. [PMID: 29434537 PMCID: PMC5790787 DOI: 10.3389/fnmol.2018.00015] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022] Open
Abstract
A ketogenic diet (KD) is a normocaloric diet composed by high fat (80-90%), low carbohydrate, and low protein consumption that induces fasting-like effects. KD increases ketone body (KBs) production and its concentration in the blood, providing the brain an alternative energy supply that enhances oxidative mitochondrial metabolism. In addition to its profound impact on neuro-metabolism and bioenergetics, the neuroprotective effect of specific polyunsaturated fatty acids and KBs involves pleiotropic mechanisms, such as the modulation of neuronal membrane excitability, inflammation, or reactive oxygen species production. KD is a therapy that has been used for almost a century to treat medically intractable epilepsy and has been increasingly explored in a number of neurological diseases. Motor function has also been shown to be improved by KD and/or medium-chain triglyceride diets in rodent models of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and spinal cord injury. These studies have proposed that KD may induce a modification in synaptic morphology and function, involving ionic channels, glutamatergic transmission, or synaptic vesicular cycling machinery. However, little is understood about the molecular mechanisms underlying the impact of KD on motor function and the perspectives of its use to acquire the neuromuscular effects. The aim of this review is to explore the conditions through which KD might improve motor function. First, we will describe the main consequences of KD exposure in tissues involved in motor function. Second, we will report and discuss the relevance of KD in pre-clinical and clinical trials in the major diseases presenting motor dysfunction.
Collapse
Affiliation(s)
- Charlotte Veyrat-Durebex
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | - Pascal Reynier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | - Vincent Procaccio
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | | | - Philippe Corcia
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Service de Neurologie, Centre Hospitalier Universitaire de Tours, Tours, France
| | - Christian R. Andres
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire de Tours, Tours, France
| | - Hélène Blasco
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire de Tours, Tours, France
| |
Collapse
|
75
|
Tang BL. Brain activity-induced neuronal glucose uptake/glycolysis: Is the lactate shuttle not required? Brain Res Bull 2017; 137:225-228. [PMID: 29273209 DOI: 10.1016/j.brainresbull.2017.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/09/2017] [Accepted: 12/15/2017] [Indexed: 01/12/2023]
Abstract
The astrocyte-neuron lactate shuttle (ANLS) hypothesis posits that during neuronal activation, astrocytic glycolysis consumes glucose and generates lactate, with the latter then imported by neurons as a preferred fuel. The hypothesis has been controversial, with multiple theoretical postulates for and against, and with empirical evidence that were either supportive or otherwise. Recent findings using direct in vivo imaging of lactate and glucose uptake as well as associated metabolic changes in neurons have now placed important constraints on the hypothesis. Here, I review these recent findings and discuss their implications on neuronal energetics.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore,.
| |
Collapse
|
76
|
Nakamura T, Lipton SA. 'SNO'-Storms Compromise Protein Activity and Mitochondrial Metabolism in Neurodegenerative Disorders. Trends Endocrinol Metab 2017; 28:879-892. [PMID: 29097102 PMCID: PMC5701818 DOI: 10.1016/j.tem.2017.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 01/07/2023]
Abstract
The prevalence of neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), is currently a major public health concern due to the lack of efficient disease-modifying therapeutic options. Recent evidence suggests that mitochondrial dysfunction and nitrosative/oxidative stress are key common mediators of pathogenesis. In this review, we highlight molecular mechanisms linking NO-dependent post-translational modifications, such as cysteine S-nitrosylation and tyrosine nitration, to abnormal mitochondrial metabolism. We further discuss the hypothesis that pathological levels of NO compromise brain energy metabolism via aberrant S-nitrosylation of key enzymes in the tricarboxylic acid (TCA) cycle and oxidative phosphorylation, contributing to neurodegenerative conditions. A better understanding of these pathophysiological events may provide a potential pathway for designing novel therapeutics to ameliorate neurodegenerative disorders.
Collapse
Affiliation(s)
- Tomohiro Nakamura
- Neuroscience Translational Center, and Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA.
| | - Stuart A Lipton
- Neuroscience Translational Center, and Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA; Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
77
|
Elamin M, Ruskin DN, Masino SA, Sacchetti P. Ketone-Based Metabolic Therapy: Is Increased NAD + a Primary Mechanism? Front Mol Neurosci 2017; 10:377. [PMID: 29184484 PMCID: PMC5694488 DOI: 10.3389/fnmol.2017.00377] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
The ketogenic diet’s (KD) anticonvulsant effects have been well-documented for nearly a century, including in randomized controlled trials. Some patients become seizure-free and some remain so after diet cessation. Many recent studies have explored its expanded therapeutic potential in diverse neurological disorders, yet no mechanism(s) of action have been established. The diet’s high fat, low carbohydrate composition reduces glucose utilization and promotes the production of ketone bodies. Ketone bodies are a more efficient energy source than glucose and improve mitochondrial function and biogenesis. Cellular energy production depends on the metabolic coenzyme nicotinamide adenine dinucleotide (NAD), a marker for mitochondrial and cellular health. Furthermore, NAD activates downstream signaling pathways (such as the sirtuin enzymes) associated with major benefits such as longevity and reduced inflammation; thus, increasing NAD is a coveted therapeutic endpoint. Based on differential NAD+ utilization during glucose- vs. ketone body-based acetyl-CoA generation for entry into the tricarboxylic cycle, we propose that a KD will increase the NAD+/NADH ratio. When rats were fed ad libitum KD, significant increases in hippocampal NAD+/NADH ratio and blood ketone bodies were detected already at 2 days and remained elevated at 3 weeks, indicating an early and persistent metabolic shift. Based on diverse published literature and these initial data we suggest that increased NAD during ketolytic metabolism may be a primary mechanism behind the beneficial effects of this metabolic therapy in a variety of brain disorders and in promoting health and longevity.
Collapse
Affiliation(s)
- Marwa Elamin
- Neuroscience Program, Department of Biology, University of Hartford, West Hartford, CT, United States
| | - David N Ruskin
- Neuroscience Program and Psychology Department, Trinity College, Hartford, CT, United States
| | - Susan A Masino
- Neuroscience Program and Psychology Department, Trinity College, Hartford, CT, United States
| | - Paola Sacchetti
- Neuroscience Program, Department of Biology, University of Hartford, West Hartford, CT, United States
| |
Collapse
|
78
|
D'Cunha NM, McKune AJ, Panagiotakos DB, Georgousopoulou EN, Thomas J, Mellor DD, Naumovski N. Evaluation of dietary and lifestyle changes as modifiers of S100β levels in Alzheimer's disease. Nutr Neurosci 2017; 22:1-18. [PMID: 28696163 DOI: 10.1080/1028415x.2017.1349032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is a significant body of research undertaken in order to elucidate the mechanisms underlying the pathology of Alzheimer's disease (AD), as well as to discover early detection biomarkers and potential therapeutic strategies. One such proposed biomarker is the calcium binding protein S100β, which, depending on its local concentration, is known to exhibit both neurotrophic and neuroinflammatory properties in the central nervous system. At present, relatively little is known regarding the effect of chronic S100β disruption in AD. Dietary intake has been identified as a modifiable risk factor for AD. Preliminary in vitro and animal studies have demonstrated an association between S100β expression and dietary intake which links to AD pathophysiology. This review describes the association of S100β to fatty acids, ketone bodies, insulin, and botanicals as well as the potential impact of physical activity as a lifestyle factor. We also discuss the prospective implications of these findings, including support of the use of a Mediterranean dietary pattern and/or the ketogenic diet as an approach to modify AD risk.
Collapse
Affiliation(s)
- Nathan M D'Cunha
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Andrew J McKune
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,c University of Canberra, Research Institute for Sport and Exercise , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,d Discipline of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences , University of KwaZulu-Natal , Durban 4041 , South Africa
| | - Demosthenes B Panagiotakos
- e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Ekavi N Georgousopoulou
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Jackson Thomas
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Duane D Mellor
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Nenad Naumovski
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| |
Collapse
|
79
|
Puchowicz MA, Seyfried TN. Novel ketone body therapy for managing Alzheimer's disease: An Editorial Highlight for Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease. J Neurochem 2017; 141:162-164. [PMID: 28299805 DOI: 10.1111/jnc.13979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 11/29/2022]
Abstract
Read the highlighted article 'Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease' on page 195.
Collapse
Affiliation(s)
- Michelle A Puchowicz
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas N Seyfried
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|