201
|
de la Rubia Ortí JE, García-Pardo MP, Drehmer E, Sancho Cantus D, Julián Rochina M, Aguilar MA, Hu Yang I. Improvement of Main Cognitive Functions in Patients with Alzheimer's Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. J Alzheimers Dis 2019; 65:577-587. [PMID: 30056419 DOI: 10.3233/jad-180184] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder (mainly in women), and new therapies are needed. In this way, ketone bodies are a direct source of cellular energy and can be obtained from coconut oil, postulating that coconut oil could be a new non-pharmacological alternative in AD patients. OBJECTIVE The aim of this study is to detect changes in the main cognitive functions of patients with AD after following a coconut oil enriched Mediterranean diet, and to determine whether there are differences in function of stage or sex. METHODS A prospective, longitudinal, qualitative, analytic, experimental study was carried out in 44 patients with AD, who were randomly divided into two homogenous groups of 22 patients each: an experimental group of patients who followed a coconut oil enriched Mediterranean diet for 21 days and a control group. In order to determine the cognitive changes after the intervention, we carried out the 7 Minute Screen, which analyses temporal orientation, visuospatial and visuoconstructive abilities, and semantic and episodic memory. RESULTS After intervention with coconut oil, improvements in episodic, temporal orientation, and semantic memory were observed, and it seems that the positive effect is more evident in women with mild-moderate state, although other improvements in males and severe state were also shown. CONCLUSIONS The isocaloric coconut oil enriched Mediterranean diet seems to improve cognitive functions in patients with AD, with differences according to patient sex and degree of severity of the disease, although more studies in this line are needed.
Collapse
Affiliation(s)
| | - María Pilar García-Pardo
- Pre-Departmental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I. Castelló de la Plana, Castelló, Spain
| | - Eraci Drehmer
- Faculty of Nursing, Catholic University of Valencia, Valencia, Spain
| | | | | | | | - Iván Hu Yang
- Faculty of Nursing, Catholic University of Valencia, Valencia, Spain
| |
Collapse
|
202
|
Dietary Neuroketotherapeutics for Alzheimer's Disease: An Evidence Update and the Potential Role for Diet Quality. Nutrients 2019; 11:nu11081910. [PMID: 31443216 PMCID: PMC6722814 DOI: 10.3390/nu11081910] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/29/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease with growing prevalence as the global population ages. Currently available treatments for AD have minimal efficacy and there are no proven treatments for its prodrome, mild cognitive impairment (MCI). AD etiology is not well understood and various hypotheses of disease pathogenesis are currently under investigation. A consistent hallmark in patients with AD is reduced brain glucose utilization; however, evidence suggests that brain ketone metabolism remains unimpaired, thus, there is a great deal of increased interest in the potential value of ketone-inducing therapies for the treatment of AD (neuroketotherapeutics; NKT). The goal of this review was to discuss dietary NKT approaches and mechanisms by which they exert a possible therapeutic benefit, update the evidence available on NKTs in AD and consider a potential role of diet quality in the clinical use of dietary NKTs. Whether NKTs affect AD symptoms through the restoration of bioenergetics, the direct and indirect modulation of antioxidant and inflammation pathways, or both, preliminary positive evidence suggests that further study of dietary NKTs as a disease-modifying treatment in AD is warranted.
Collapse
|
203
|
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: 178] [Impact Index Per Article: 29.7] [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
|
204
|
Croteau E, Castellano CA, Richard MA, Fortier M, Nugent S, Lepage M, Duchesne S, Whittingstall K, Turcotte ÉE, Bocti C, Fülöp T, Cunnane SC. Ketogenic Medium Chain Triglycerides Increase Brain Energy Metabolism in Alzheimer's Disease. J Alzheimers Dis 2019; 64:551-561. [PMID: 29914035 DOI: 10.3233/jad-180202] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In Alzheimer's disease (AD), it is unknown whether the brain can utilize additional ketones as fuel when they are derived from a medium chain triglyceride (MCT) supplement. OBJECTIVE To assess whether brain ketone uptake in AD increases in response to MCT as it would in young healthy adults. METHODS Mild-moderate AD patients sequentially consumed 30 g/d of two different MCT supplements, both for one month: a mixture of caprylic (55%) and capric acids (35%) (n = 11), followed by a wash-out and then tricaprylin (95%; n = 6). Brain ketone (11C-acetoacetate) and glucose (FDG) uptake were quantified by PET before and after each MCT intervention. RESULTS Brain ketone consumption doubled on both types of MCT supplement. The slope of the relationship between plasma ketones and brain ketone uptake was the same as in healthy young adults. Both types of MCT increased total brain energy metabolism by increasing ketone supply without affecting brain glucose utilization. CONCLUSION Ketones from MCT compensate for the brain glucose deficit in AD in direct proportion to the level of plasma ketones achieved.
Collapse
Affiliation(s)
- Etienne Croteau
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Marie Anne Richard
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Fortier
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Scott Nugent
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, QC, Canada
| | - Martin Lepage
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Duchesne
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, QC, Canada.,Department of Radiology, Université Laval, Québec, QC, Canada
| | | | - Éric E Turcotte
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christian Bocti
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Tamàs Fülöp
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stephen C Cunnane
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
205
|
Yang H, Shan W, Zhu F, Wu J, Wang Q. Ketone Bodies in Neurological Diseases: Focus on Neuroprotection and Underlying Mechanisms. Front Neurol 2019; 10:585. [PMID: 31244753 PMCID: PMC6581710 DOI: 10.3389/fneur.2019.00585] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
There is growing evidence that ketone bodies, which are derived from fatty acid oxidation and usually produced in fasting state or on high-fat diets have broad neuroprotective effects. Although the mechanisms underlying the neuroprotective effects of ketone bodies have not yet been fully elucidated, studies in recent years provided abundant shreds of evidence that ketone bodies exert neuroprotective effects through possible mechanisms of anti-oxidative stress, maintaining energy supply, modulating the activity of deacetylation and inflammatory responses. Based on the neuroprotective effects, the ketogenic diet has been used in the treatment of several neurological diseases such as refractory epilepsy, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. The ketogenic diet has great potential clinically, which should be further explored in future studies. It is necessary to specify the roles of components in ketone bodies and their therapeutic targets and related pathways to optimize the strategy and efficacy of ketogenic diet therapy in the future.
Collapse
Affiliation(s)
- Huajun Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China
| | - Wei Shan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fei Zhu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China
| | - Jianping Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
206
|
Mediterranean and MIND Diets Containing Olive Biophenols Reduces the Prevalence of Alzheimer's Disease. Int J Mol Sci 2019; 20:ijms20112797. [PMID: 31181669 PMCID: PMC6600544 DOI: 10.3390/ijms20112797] [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: 05/18/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
The risk of Alzheimer’s disease (AD) increases with nonmodifiable conditions including age and lack of effective efficacious pharmacotherapy. During the past decades, the non-pharmacotherapy mode of treatment of dietary modification received extensive attention in AD research. In order to reduce the AD pathology and cognitive decline, various dietary patterns have been attempted including caloric restriction (CR), dietary approaches to stop hypertension (DASH), ketogenic diets (KD), Mediterranean diet (MedDi) and Mediterranean-DASH diet Intervention for Neurological Delay (MIND) diet. Higher adherence to the MedDi diet was associated with decreases in cardiovascular and neurological disorders including AD and related cognitive decline. However, another emerging healthy dietary pattern MIND diet has also been associated with slower rates of cognitive decline and significant reduction of AD rate. Olive serves as one of the building block components of MedDi and MIND diets and the exerted potential health beneficial might be suggested due to the presence of its bioactive constituents such as oleic acids and phenolic compounds (biophenols). A few trials using medical food showed an optimal result in presymptomatic or early stages of AD. The review supports the notion that MedDi and MIND diets display potential for maintaining the cognitive function as nonpharmacological agents against AD pathology and proposed preventative mechanism through the presence of olive biophenols and presents the gaps along with the future directions.
Collapse
|
207
|
Crosstalk between the Ketogenic Diet and Epilepsy: From the Perspective of Gut Microbiota. Mediators Inflamm 2019; 2019:8373060. [PMID: 31281229 PMCID: PMC6589192 DOI: 10.1155/2019/8373060] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022] Open
Abstract
Given the association between a range of neurological disorders and changes in the gut microbiota, interest in the gut microbiota has recently increased. In particular, the significant involvement of the autoimmune processes in the development of epilepsy, one of the most serious and widespread neurological diseases, has led to a suggested link with the gut microbiome. Because the constitution of the gut microbiome can be influenced by diet, dietary therapy has been shown to have a positive impact on a wide range of conditions via alteration of the gut microbiota. An example of one such diet is the ketogenic diet (KD), which promotes a diet that contains high levels of fat, adequate levels of protein, and low levels of carbohydrate. Due to the near-total elimination of carbohydrates from the individual's food in this ultra-high-fat diet, ketone bodies become an important source of energy. Although the ketogenic diet has proven successful in the treatment of refractory epilepsy and other illnesses, the underlying mechanisms of its neuroprotective effects have yet to be fully elucidated. Nevertheless, recent studies strongly indicate a role for the gut microbiota in the effective treatment of epilepsy with the ketogenic diet. The latest advances regarding the links between the ketogenic diet, gut microbiota, and epilepsy are reviewed in this article, with a particular focus on the role of the gut microbiota in the treatment outcome.
Collapse
|
208
|
Geary DC. The Spark of Life and the Unification of Intelligence, Health, and Aging. CURRENT DIRECTIONS IN PSYCHOLOGICAL SCIENCE 2019. [DOI: 10.1177/0963721419829719] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
General intelligence ( g) represents the factors that influence performance across all academic and cognitive domains. The search for these factors has been ongoing for more than a century and has focused on the brain and cognitive systems that support learning and problem solving. In recent decades, it has become clear that the factors that influence academic and cognitive performance extend to general health and to successful aging in adulthood. The implication is that there may be one or several fundamental processes that influence the functioning of all biological systems, not simply the brain. The functioning of mitochondria is well situated as one of the processes that might unify intelligence, health, and aging. These organelles are located within cells and are the primary producers of cellular energy, among other functions. Energy availability, in turn, is the lowest common denominator needed for the development, maintenance, and optimal functioning of all biological systems. Here, I review the relations among intelligence, health, and aging and outline how the efficiency of mitochondrial functioning can link them together.
Collapse
Affiliation(s)
- David C. Geary
- Department of Psychological Sciences, University of Missouri
| |
Collapse
|
209
|
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: 3.3] [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
|
210
|
O'Neill BV, Dodds CM, Miller SR, Gupta A, Lawrence P, Bullman J, Chen C, Dewit O, Kumar S, Dustagheer M, Price J, Shabbir S, Nathan PJ. The effects of GSK2981710, a medium-chain triglyceride, on cognitive function in healthy older participants: A randomised, placebo-controlled study. Hum Psychopharmacol 2019; 34:e2694. [PMID: 31124194 DOI: 10.1002/hup.2694] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This double-blind, randomised, placebo-controlled, two-part study assessed the impact of GSK2981710, a medium-chain triglyceride (MCT) that liberates ketone bodies, on cognitive function, safety, and tolerability in healthy older adults. METHODS Part 1 was a four-period dose-selection study (n = 8 complete). Part 2 was a two-period crossover study (n = 80 complete) assessing the acute (Day 1) and prolonged (Day 15) effects of GSK2981710 on cognition and memory-related neuronal activity. Safety and tolerability of MCT supplementation were monitored in both parts of the study. RESULTS The most common adverse event was diarrhoea (100% and 75% of participants in Parts 1 and 2, respectively). Most adverse events were mild to moderate, and 11% participants were withdrawn due to one or more adverse events. Although GSK2981710 (30 g/day) resulted in increased peak plasma β-hydroxybutyrate (BHB) concentrations, no significant improvements in cognitive function or memory-related neuronal activity were observed. CONCLUSION Over a duration of 14 days, increasing plasma BHB levels with daily administration of GSK2981710 had no effects on neuronal activity or cognitive function. This result indicates that modulating plasma ketone levels with GSK2981710 may be ineffective in improving cognitive function in healthy older adults, or the lack of observed effect could be related to several factors including study population, plasma BHB concentrations, MCT composition, or treatment duration.
Collapse
Affiliation(s)
- Barry V O'Neill
- GSK Nutrition, GSK Consumer Healthcare, Brentford, UK.,Respiratory Health, GSK Consumer Healthcare, Nyon, Switzerland
| | - Chris M Dodds
- Department of Psychology, University of Exeter, Exeter, UK
| | - Sam R Miller
- Department of Quantitative Sciences, GlaxoSmithKline, Stevenage, UK
| | - Ashutosh Gupta
- Department of Quantitative Sciences India, GlaxoSmithKline, Bangalore, India
| | | | - Jonathan Bullman
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Stevenage, UK
| | - Chao Chen
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, London, UK
| | - Odile Dewit
- Clinical Unit, GlaxoSmithKline, Cambridge, UK
| | | | | | | | - Shaila Shabbir
- Clinical Pharmacology Study Sciences and Operations, GlaxoSmithKline, Stevenage, UK
| | - Pradeep J Nathan
- Sosei Heptares, Cambridge, UK.,The School of Psychological Sciences, Monash University, Clayton, Australia.,Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
211
|
Rho JM, Shao LR, Stafstrom CE. 2-Deoxyglucose and Beta-Hydroxybutyrate: Metabolic Agents for Seizure Control. Front Cell Neurosci 2019; 13:172. [PMID: 31114484 PMCID: PMC6503754 DOI: 10.3389/fncel.2019.00172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 04/11/2019] [Indexed: 01/12/2023] Open
Abstract
Current anti-seizure drugs (ASDs) are believed to reduce neuronal excitability through modulation of ion channels and transporters that regulate excitability at the synaptic level. While most patients with epilepsy respond to ASDs, many remain refractory to medical treatment but respond favorably to a high-fat, low-carbohydrate metabolism-based therapy known as the ketogenic diet (KD). The clinical effectiveness of the KD has increasingly underscored the thesis that metabolic factors also play a crucial role in the dampening neuronal hyperexcitability that is a hallmark feature of epilepsy. This notion is further amplified by the clinical utility of other related metabolism-based diets such as the modified Atkins diet and the low-glycemic index treatment (LGIT). Traditional high-fat diets are characterized by enhanced fatty acid oxidation (which produces ketone bodies such as beta-hydroxybutyrate) and a reduction in glycolytic flux, whereas the LGIT is predicated mainly on the latter observation of reduced blood glucose levels. As dietary implementation is not without challenges regarding clinical administration and patient compliance, there is an inherent desire and need to determine whether specific metabolic substrates and/or enzymes might afford similar clinical benefits, hence validating the concept of a “diet in a pill.” Here, we discuss the evidence for one glycolytic inhibitor, 2-deoxyglucose (2DG) and one metabolic substrate, β-hydroxybutyrate (BHB) exerting direct effects on neuronal excitability, highlight their mechanistic differences, and provide the strengthening scientific rationale for their individual or possibly combined use in the clinical arena of seizure management.
Collapse
Affiliation(s)
- Jong M Rho
- Section of Pediatric Neurology, Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Li-Rong Shao
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
212
|
A ketogenic drink improves brain energy and some measures of cognition in mild cognitive impairment. Alzheimers Dement 2019; 15:625-634. [DOI: 10.1016/j.jalz.2018.12.017] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/31/2018] [Accepted: 12/29/2018] [Indexed: 11/23/2022]
|
213
|
Affiliation(s)
| | - Viswanathan Mohan
- Dr. Mohan's Diabetes Specialities Centre & Madras Diabetes Research Foundation, WHO Collaborating Centre for Noncommunicable Diseases Prevention & Control, IDF Centre of Excellence in Diabetes Care & ICMR Centre for Advanced Research on Diabetes, Chennai 600 086, Tamil Nadu, India
| |
Collapse
|
214
|
St-Pierre V, Vandenberghe C, Lowry CM, Fortier M, Castellano CA, Wagner R, Cunnane SC. Plasma Ketone and Medium Chain Fatty Acid Response in Humans Consuming Different Medium Chain Triglycerides During a Metabolic Study Day. Front Nutr 2019; 6:46. [PMID: 31058159 PMCID: PMC6481320 DOI: 10.3389/fnut.2019.00046] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/27/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Medium chain triglycerides (MCT) are ketogenic but the relationship between the change in plasma ketones and the change plasma medium chain fatty acids (MCFA)—octanoate, decanoate, or dodecanoate—after an oral dose of MCT is not well-known. An 8 h metabolic study day is a suitable model to assess the acute effects on plasma ketones and MCFA after a dose of tricaprylin (C8), tricaprin (C10), trilaurin (C12) or mixed MCT (C8C10). Objective: To assess in healthy humans the relationship between the change in plasma ketones, and octanoate, decanoate and dodecanoate in plasma total lipids during an 8 h metabolic study day in which a first 20 ml dose of the homogenized test oil is taken with breakfast and a second 20 ml dose is taken 4 h later without an accompanying meal. Results: The change in plasma acetoacetate, β-hydroxybutyrate and total ketones was highest after C8 (0.5 to 3 h post-dose) and was lower during tests in which octanoate was absent or was diluted by C10 in the test oil. The plasma ketone response was also about 2 fold higher without an accompanying meal (P = 0.012). However, except during the pure C10 test, the response of octanoate, decanoate or dodecanoate in plasma total lipids to the test oils was not affected by consuming an accompanying meal. Except with C12, the 4 h area-under-the-curve of plasma β-hydroxybutyrate/acetoacetate was 2–3 fold higher when no meal was consumed (P < 0.04). Conclusion: C8 was about three times more ketogenic than C10 and about six times more ketogenic than C12 under these acute metabolic test conditions, an effect related to the post-dose increase in octanoate in plasma total lipids.
Collapse
Affiliation(s)
| | | | - Carolyne-Mary Lowry
- Departments of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | - Richard Wagner
- Research Center on Aging, Sherbrooke, QC, Canada.,Departments of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Departments of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stephen C Cunnane
- Research Center on Aging, Sherbrooke, QC, Canada.,Departments of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,Departments of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
215
|
Gross EC, Klement RJ, Schoenen J, D'Agostino DP, Fischer D. Potential Protective Mechanisms of Ketone Bodies in Migraine Prevention. Nutrients 2019; 11:E811. [PMID: 30974836 PMCID: PMC6520671 DOI: 10.3390/nu11040811] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
An increasing amount of evidence suggests that migraines are a response to a cerebral energy deficiency or oxidative stress levels that exceed antioxidant capacity. The ketogenic diet (KD), a diet mimicking fasting that leads to the elevation of ketone bodies (KBs), is a therapeutic intervention targeting cerebral metabolism that has recently shown great promise in the prevention of migraines. KBs are an alternative fuel source for the brain, and are thus likely able to circumvent some of the abnormalities in glucose metabolism and transport found in migraines. Recent research has shown that KBs-D-β-hydroxybutyrate in particular-are more than metabolites. As signalling molecules, they have the potential to positively influence other pathways commonly believed to be part of migraine pathophysiology, namely: mitochondrial functioning, oxidative stress, cerebral excitability, inflammation and the gut microbiome. This review will describe the mechanisms by which the presence of KBs, D-BHB in particular, could influence those migraine pathophysiological mechanisms. To this end, common abnormalities in migraines are summarised with a particular focus on clinical data, including phenotypic, biochemical, genetic and therapeutic studies. Experimental animal data will be discussed to elaborate on the potential therapeutic mechanisms of elevated KBs in migraine pathophysiology, with a particular focus on the actions of D-BHB. In complex diseases such as migraines, a therapy that can target multiple possible pathogenic pathways seems advantageous. Further research is needed to establish whether the absence/restriction of dietary carbohydrates, the presence of KBs, or both, are of primary importance for the migraine protective effects of the KD.
Collapse
Affiliation(s)
- Elena C Gross
- Division of Paediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, 4056 Basel, Switzerland.
| | - Rainer J Klement
- Department of Radiation Oncology, Leopoldina Hospital Schweinfurt, 97422 Schweinfurt, Germany.
| | - Jean Schoenen
- Headache Research Unit, University of Liège, Dept of Neurology-Citadelle Hospital, 4000 Liège, Belgium.
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Metabolic Medicine Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA.
| | - Dirk Fischer
- Division of Paediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
216
|
The ketogenic diet as a potential treatment and prevention strategy for Alzheimer's disease. Nutrition 2019; 60:118-121. [DOI: 10.1016/j.nut.2018.10.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/12/2018] [Accepted: 10/07/2018] [Indexed: 01/01/2023]
|
217
|
Effects of 7-Day Ketone Ingestion and a Physiological Workload on Postural Stability, Cognitive, and Muscular Exertion Measures in Professional Firefighters. SAFETY 2019. [DOI: 10.3390/safety5010015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Postural stability and cognitive performance are challenged in firefighters. The purpose of this investigation was to examine the impact of 7-day ketone supplementation on postural stability, cognitive performance, and muscular activation before and after a physiological workload. Methods: Nine professional firefighters completed two experimental sessions (pre- and post-workload) in a counterbalanced, double-blind design. Participants ingested either a ketone salt (KS) or placebo (PLA) daily for seven days, and had an eighth ingestion 30 min prior to testing. Each experimental testing consisted of maximal voluntary contractions (MVIC) for four muscles (knee flexors—BF, extensor—VM, ankle dorsiflexors—TA, and plantar flexors—MG) using electromyography and postural stability testing (eyes open (EO), eyes closed (EC), and eyes open-dual-task using a FitLight™ system (EOT)), before (pre-workload) and after (post-workload) a simulated physiological workload. The workload consisted of 35 min steady state exercise at 60% of peak oxygen consumption wearing firefighter personal protective equipment (PPE). Results: Significant differences were limited to time effects (pre-workload vs. post-workload), with no differences between groups (KS vs. PLA). Significantly lower muscle activity in VM, TA, and MG during MVIC, greater postural sway and muscle activity in BF during EC and EOT, and slower response time during EOT were evident post-workload. Conclusions: A 7-day ketone supplementation does not impact postural stability, muscle activity, and cognitive tasks, but a fatiguing workload causes significant performance reduction.
Collapse
|
218
|
Lange KW, Guo J, Kanaya S, Lange KM, Nakamura Y, Li S. Medical foods in Alzheimer’s disease. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2019.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
219
|
Chen F, He X, Luan G, Li T. Role of DNA Methylation and Adenosine in Ketogenic Diet for Pharmacoresistant Epilepsy: Focus on Epileptogenesis and Associated Comorbidities. Front Neurol 2019; 10:119. [PMID: 30863356 PMCID: PMC6399128 DOI: 10.3389/fneur.2019.00119] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/29/2019] [Indexed: 01/02/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by a long term propensity to produce unprovoked seizures and by the associated comorbidities including neurological, cognitive, psychiatric, and impairment the quality of life. Despite the clinic availability of several novel antiepileptic drugs (AEDs) with different mechanisms of action, more than one-third of patients with epilepsy suffer with pharmacoresistant epilepsy. Until now, no AEDs have been proven to confer the efficacy in alteration of disease progression or inhibition of the development of epilepsy. The ketogenic diet, the high-fat, low-carbohydrate composition is an alternative metabolic therapy for epilepsy, especially for children with drug-resistant epilepsy. Recently clinical and experimental results demonstrate its efficacy in ameliorating both seizures and comorbidities associated with epilepsy, such as cognitive/psychiatric concerns for the patients with refractory epilepsy. Of importance, ketogenic diet demonstrates to be a promising disease-modifying or partial antiepileptogenesis therapy for epilepsy. The mechanisms of action of ketogenic diet in epilepsy have been revealed recently, such as epigenetic mechanism for increase the adenosine level in the brain and inhibition of DNA methylation. In the present review, we will focus on the mechanisms of ketogenic diet therapies underlying adenosine system in the prevention of epileptogenesis and disease modification. In addition, we will review the role of ketogenic diet therapy in comorbidities associated epilepsy and the underlying mechanisms of adenosine.
Collapse
Affiliation(s)
- Fan Chen
- Beijing Key Laboratory of Epilepsy Research, Department of Neurology, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Xinghui He
- Beijing Key Laboratory of Epilepsy Research, Department of Neurosurgery, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Guoming Luan
- Beijing Key Laboratory of Epilepsy Research, Department of Neurosurgery, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tianfu Li
- Beijing Key Laboratory of Epilepsy Research, Department of Neurology, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
220
|
Abstract
PURPOSE OF REVIEW Various groups have explored the effect of apolipoprotein E (APOE) on neurodegeneration through nutritional and metabolic alterations. In this review, we hope to summarize recent findings in humans as well as preclinical APOE models. RECENT FINDINGS Metabolic pathways including lipid metabolism appear to play a large role in the pathophysiology of Alzheimer's disease. Carrier status of the E4 variant of the APOE gene is the strongest genetic risk factor for Alzheimer's disease, and increasing evidence suggests that E4 carriers may respond differently to a host of dietary and metabolic-related treatments. A new appreciation is forming for the role of APOE in cerebral metabolism, and how nutritional factors may impact this role. SUMMARY Considering the role dietary factors play in APOE-associated cognitive decline will help us to understand how nutritional interventions may facilitate or mitigate disease progression.
Collapse
Affiliation(s)
- Brandon C. Farmer
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Lance A. Johnson
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Angela J. Hanson
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Healthcare System, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
221
|
Gross E, Putananickal N, Orsini AL, Schmidt S, Vogt DR, Cichon S, Sandor P, Fischer D. Efficacy and safety of exogenous ketone bodies for preventive treatment of migraine: A study protocol for a single-centred, randomised, placebo-controlled, double-blind crossover trial. Trials 2019; 20:61. [PMID: 30654835 PMCID: PMC6337840 DOI: 10.1186/s13063-018-3120-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/09/2018] [Indexed: 01/22/2023] Open
Abstract
Background Currently available prophylactic migraine treatment options are limited and are associated with many, often intolerable, side-effects. Various lines of research suggest that abnormalities in energy metabolism are likely to be part of migraine pathophysiology. Previously, a ketogenic diet (KD) has been reported to lead to a drastic reduction in migraine frequency. An alternative method to a strict KD is inducing a mild nutritional ketosis (0.4–2 mmol/l) with exogenous ketogenic substances. The aim of this randomised, placebo-controlled, double-blind, crossover, single-centre trial is to demonstrate safety and superiority of beta-hydroxybutyrate (βHB) in mineral salt form over placebo in migraine prevention. Methods/design Forty-five episodic migraineurs (5–14 migraine days/months), with or without aura, aged between 18 and 65 years, will be recruited at headache clinics in Switzerland, Germany and Austria and via Internet announcements. After a 4-week baseline period, patients will be randomly allocated to one of the two trial arms and receive either the βHB mineral salt or placebo for 12 weeks. This will be followed by a 4-week wash-out period, a subsequent second baseline period and, finally, another 12-week intervention with the alternative treatment. Co-medication with triptans (10 days per months) or analgesics (14 days per months) is permitted. The primary outcome is the mean change from baseline in the number of migraine days (meeting International Classification of Headache Disorders version 3 criteria) during the last 4 weeks of intervention compared to placebo. Secondary endpoints include mean changes in headache days of any severity, acute migraine medication use, migraine intensity and migraine and headache-related disability. Exploratory outcomes are (in addition to routine laboratory analysis) genetic profiling and expression analysis, oxidative and nitrosative stress, as well as serum cytokine analysis, and blood βHB and glucose analysis (pharmacokinetics). Discussion A crossover design was chosen as it greatly improves statistical power and participation rates, without increasing costs. To our knowledge this is the first RCT using βHB salts worldwide. If proven effective and safe, βHB might not only offer a new prophylactic treatment option for migraine patients, but might additionally pave the way for clinical trials assessing its use in related diseases. Trial registration ClinicalTrials.gov, NCT03132233. Registered on 27 April 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-3120-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Gross
- Division of Neuropaediatrics, University of Basel Children's Hospital, University of Basel, Spitalstrasse 33, Postfach, 4056, Basel, Switzerland
| | - Niveditha Putananickal
- Division of Neuropaediatrics, University of Basel Children's Hospital, University of Basel, Spitalstrasse 33, Postfach, 4056, Basel, Switzerland.
| | - Anna-Lena Orsini
- Division of Neuropaediatrics, University of Basel Children's Hospital, University of Basel, Spitalstrasse 33, Postfach, 4056, Basel, Switzerland
| | - Simone Schmidt
- Division of Neuropaediatrics, University of Basel Children's Hospital, University of Basel, Spitalstrasse 33, Postfach, 4056, Basel, Switzerland
| | - Deborah R Vogt
- Department of Clinical Research, Clinical Trial Unit, University of Basel Hospital, University of Basel, Basel, Switzerland
| | - Sven Cichon
- Department of Medical Genetics, University of Basel Hospital, University of Basel, Basel, Switzerland
| | | | - Dirk Fischer
- Division of Neuropaediatrics, University of Basel Children's Hospital, University of Basel, Spitalstrasse 33, Postfach, 4056, Basel, Switzerland
| |
Collapse
|
222
|
Włodarek D. Role of Ketogenic Diets in Neurodegenerative Diseases (Alzheimer's Disease and Parkinson's Disease). Nutrients 2019; 11:nu11010169. [PMID: 30650523 PMCID: PMC6356942 DOI: 10.3390/nu11010169] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/31/2018] [Accepted: 01/09/2019] [Indexed: 12/29/2022] Open
Abstract
The goal of this review was to assess the effectiveness of ketogenic diets on the therapy of neurodegenerative diseases. The ketogenic diet is a low-carbohydrate and fat-rich diet. Its implementation has a fasting-like effect, which brings the body into a state of ketosis. The ketogenic diet has, for almost 100 years, been used in the therapy of drug-resistant epilepsy, but current studies indicate possible neuroprotective effects. Thus far, only a few studies have evaluated the role of the ketogenic diet in the prevention of Parkinson’s disease (PD) and Alzheimer’s disease (AD). Single studies with human participants have demonstrated a reduction of disease symptoms after application. The application of the ketogenic diet to elderly people, however, raises certain concerns. Persons with neurodegenerative diseases are at risk of malnutrition, while food intake reduction is associated with disease symptoms. In turn, the ketogenic diet leads to a reduced appetite; it is not attractive from an organoleptic point of view, and may be accompanied by side effects of the gastrointestinal system. All this may lead to further lowering of consumed food portions by elderly persons with neurodegenerative diseases and, in consequence, to further reduction in the supply of nutrients provided by the diet. Neither data on the long-term application of the ketogenic diet in patients with neurodegenerative disease or data on its effects on disease symptoms are available. Further research is needed to evaluate the suitability of the ketogenic diet in the therapy of AD- or PD-affected persons.
Collapse
Affiliation(s)
- Dariusz Włodarek
- Department of Dietetics, Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159c Nowoursynowska Str., 02-776 Warsaw, Poland.
| |
Collapse
|
223
|
Dominguez LJ, Barbagallo M. Dietary Strategies and Supplements for the Prevention of Cognitive Decline and Alzheimer’s Disease. OMEGA FATTY ACIDS IN BRAIN AND NEUROLOGICAL HEALTH 2019:231-247. [DOI: 10.1016/b978-0-12-815238-6.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
224
|
Schloss J. Medical synopsis: New metabolic markers to help diagnosis and assess disease progression for Alzheimer's disease, normal pressure hydrocephalus and brain tumours. ADVANCES IN INTEGRATIVE MEDICINE 2018. [DOI: 10.1016/j.aimed.2018.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
225
|
Harun-Or-Rashid M, Inman DM. Reduced AMPK activation and increased HCAR activation drive anti-inflammatory response and neuroprotection in glaucoma. J Neuroinflammation 2018; 15:313. [PMID: 30424795 PMCID: PMC6234605 DOI: 10.1186/s12974-018-1346-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023] Open
Abstract
Background Glaucoma is a chronic degenerative disease for which inflammation is considered to play a pivotal role in the pathogenesis and progression. In this study, we examined the impact of a ketogenic diet on the inflammation evident in glaucoma as a follow-up to a recent set of experiments in which we determined that a ketogenic diet protected retinal ganglion cell structure and function. Methods Both sexes of DBA/2J (D2) mice were placed on a ketogenic diet (keto) or standard rodent chow (untreated) for 8 weeks beginning at 9 months of age. DBA/2J-Gpnmb+ (D2G) mice were also used as a non-pathological genetic control for the D2 mice. Retina and optic nerve (ON) tissues were micro-dissected and used for the analysis of microglia activation, expression of pro- and anti-inflammatory molecules, and lactate- or ketone-mediated anti-inflammatory signaling. Data were analyzed by immunohistochemistry, quantitative RT-PCR, ELISA, western blot, and capillary tube-based electrophoresis techniques. Results Microglia activation was observed in D2 retina and ON as documented by intense microglial-specific Iba1 immunolabeling of rounded-up and enlarged microglia. Ketogenic diet treatment reduced Iba1 expression and the activated microglial phenotype. We detected low energy-induced AMP-activated protein kinase (AMPK) phosphorylation in D2 retina and ON that triggered NF-κB p65 signaling through its nuclear translocation. NF-κB induced pro-inflammatory TNF-α, IL-6, and NOS2 expression in D2 retina and ON. However, treatment with the ketogenic diet reduced AMPK phosphorylation, NF-κB p65 nuclear translocation, and expression of pro-inflammatory molecules. The ketogenic diet also induced expression of anti-inflammatory agents Il-4 and Arginase-1 in D2 retina and ON. Increased expression of hydroxycarboxylic acid receptor 1 (HCAR1) after ketogenic diet treatment was observed. HCAR1 stimulation by lactate or ketones from the ketogenic diet reduced inflammasome formation, as shown by reduced mRNA and protein expression of NLRP3 and IL-1β. We also detected increased levels of Arrestin β-2 protein, an adapter protein required for HCAR1 signaling. Conclusion Our data demonstrate that the AMPK activation apparent in the glaucomatous retina and ON triggers NF-κB signaling and consequently induces a pro-inflammatory response. The ketogenic diet resolves energy demand and ameliorates the inflammation by inhibition of AMPK activation and stimulation of HCAR1-ARRB2 signaling that inhibits NLRP3 inflammasome-mediated inflammation. Thus, these findings depict a neuroprotective mechanism of the ketogenic diet in controlling inflammation and suggest potential therapeutic targets for inflammatory neurodegenerative diseases, including glaucoma.
Collapse
Affiliation(s)
- Mohammad Harun-Or-Rashid
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| |
Collapse
|
226
|
Solfrizzi V, Agosti P, Lozupone M, Custodero C, Schilardi A, Valiani V, Santamato A, Sardone R, Dibello V, Di Lena L, Stallone R, Ranieri M, Bellomo A, Greco A, Daniele A, Seripa D, Sabbà C, Logroscino G, Panza F. Nutritional interventions and cognitive-related outcomes in patients with late-life cognitive disorders: A systematic review. Neurosci Biobehav Rev 2018; 95:480-498. [PMID: 30395922 DOI: 10.1016/j.neubiorev.2018.10.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022]
Abstract
There have been a large number of observational studies on the impact of nutrition on neuroprotection, however, there was a lack of evidence from randomized clinical trials (RCTs). In the present systematic review, from the 32 included RCTs published in the last four years (2014-2017) in patients aged 60 years and older with different late-life cognitive disorders, nutritional intervention through medical food/nutraceutical supplementation and multidomain approach improved magnetic resonance imaging findings and other cognitive-related biomarkers, but without clear effect on cognition in mild Alzheimer's disease (AD) and mild cognitive impairment (MCI). Antioxidant-rich foods (nuts, grapes, cherries) and fatty acid supplementation, mainly n-3 polyunsaturated fatty acids (PUFA), improved specific cognitive domains and cognitive-related outcomes in MCI, mild-to-moderate dementia, and AD. Antioxidant vitamin and trace element supplementations improved only cognitive-related outcomes and biomarkers, high-dose B vitamin supplementation in AD and MCI patients improved cognitive outcomes in the subjects with a high baseline plasma n-3 PUFA, while folic acid supplementation had positive impact on specific cognitive domains in those with high homocysteine.
Collapse
Affiliation(s)
- Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy.
| | - Pasquale Agosti
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Carlo Custodero
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Andrea Schilardi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Vincenzo Valiani
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Andrea Santamato
- Physical Medicine and Rehabilitation Section, "OORR Hospital", University of Foggia, Foggia, Italy
| | - Rodolfo Sardone
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Vittorio Dibello
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luca Di Lena
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Roberta Stallone
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Maurizio Ranieri
- Physical Medicine and Rehabilitation Section, "OORR Hospital", University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Greco
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Davide Seripa
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Carlo Sabbà
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy; Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy.
| |
Collapse
|
227
|
Halikas A, Gibas KJ. AMPK induced memory improvements in the diabetic population: A case study. Diabetes Metab Syndr 2018; 12:1141-1146. [PMID: 29748034 DOI: 10.1016/j.dsx.2018.04.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
Abstract
Diabetics in mid-life carry a 1.5 times higher risk of developing Alzheimer's disease than those diagnosed with diabetes (T2D) later in life [1]. Recent research points to accelerated cognitive decline within a range of 20%-50% for middle-aged diabetics as compared to non-diabetic populations [2,3]. Metabolic syndrome (MetS), a type 2 diabetes (T2D) precursor, is also linked to MCI and AD pathologies via hypo-metabolic brain circuitry that inhibits glucose metabolism and attenuates cognitive function [4]. Dysregulation of intracellular and extracellular signaling as mediated by the mTOR and AMPK pathways is the result. These critical nutrient sensing pathways modulate epigenetic shifts in the genome by channeling fuel substrates either towards mitochondrial fatty acid oxidation (AMPK) or cytosolic glycolysis and substrate level phosphorylation (mTOR) [5]. This case study was designed to examine the link between peripheral insulin resistance and early stage memory loss in a type 2 diabetic male. Reactivating the AMPK pathway via induced and controlled nutritional ketosis combined with high intensity interval training (HIIT) (in order to inhibit mTOR signaling) were primary features of the 10 week intervention. Post intervention results revealed statistically significant reductions in HgA1c, fasting insulin and HOMA-IR (homeostatic model assessment of insulin resistance). Restoring peripheral and hypothalamic insulin sensitivity by way of AMPK activation may restore memory function, improve neuroplasticity, and normalize MetS biomarkers (Demetrius and Driver, 2014; [4,6]).
Collapse
Affiliation(s)
- Alicia Halikas
- Human Bioenergetics & Applied Health Science, Bethel University, Minnesota, USA.
| | - Kelly J Gibas
- Doctorate of Behavioral Health Sciences, Human Bioenergetics & Applied Health Science, Bethel University, Minnesota, USA.
| |
Collapse
|
228
|
Ota M, Matsuo J, Ishida I, Takano H, Yokoi Y, Hori H, Yoshida S, Ashida K, Nakamura K, Takahashi T, Kunugi H. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer's disease. Neurosci Lett 2018; 690:232-236. [PMID: 30367958 DOI: 10.1016/j.neulet.2018.10.048] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/11/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023]
Abstract
Clinical and animal studies suggested that a medium-chain triglyceride (MCT)-based ketogenic diet provides an alternative energy substrate to the brain and has neuroprotective effects, but the clinical evidence is still scarce. Here we examined the effect of an MCT-based ketogenic formula on cognitive function in patients with Alzheimer's disease (AD). The subjects were 20 Japanese patients with mild-to-moderate AD (11 males, nine females, mean age 73.4 ± 6.0 years) who, on separate days, underwent neurocognitive tests 120 min after consuming 50 g of a ketogenic formula (Ketonformula®) containing 20 g of MCTs or an isocaloric placebo formula without MCTs. The patients then took 50 g of the ketogenic formula daily for up to 12 weeks, and underwent neurocognitive tests monthly. In the first trial, although the patients' plasma levels of ketone bodies were successfully increased 120 min after the single intake of the ketogenic formula, there was no significant difference in any cognitive test results between the administrations of the ketogenic and placebo formulae. In the subsequent chronic intake trial of the ketogenic formula, 16 of the 20 patients completed the 12-week regimen. At 8 weeks after the trial's start, the patients showed significant improvement in their immediate and delayed logical memory tests compared to their baseline scores, and at 12 weeks they showed significant improvements in the digit-symbol coding test and immediate logical memory test compared to the baseline. The chronic consumption of the ketogenic formula was therefore suggested to have positive effects on verbal memory and processing speed in patients with AD.
Collapse
Affiliation(s)
- Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.
| | - Junko Matsuo
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Ikki Ishida
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Harumasa Takano
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Yuma Yokoi
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Hiroaki Hori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Sumiko Yoshida
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Kinya Ashida
- Food Science Research Laboratories, R & D Division, Meiji Co., Ltd., 1-29-1 Nanakuni, Hachiouji, Tokyo 192-0919, Japan
| | - Kentaro Nakamura
- Food Science Research Laboratories, R & D Division, Meiji Co., Ltd., 1-29-1 Nanakuni, Hachiouji, Tokyo 192-0919, Japan
| | - Takeshi Takahashi
- Food Science Research Laboratories, R & D Division, Meiji Co., Ltd., 1-29-1 Nanakuni, Hachiouji, Tokyo 192-0919, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| |
Collapse
|
229
|
Abstract
The current review highlights the evidence supporting the use of ketogenic diet therapies in the management of a growing number of neurological disorders in adults. An overview of the scientific literature supporting posited mechanisms of therapeutic efficacy is presented including effects on neurotransmission, oxidative stress, and neuro-inflammation. The clinical evidence supporting ketogenic diet use in the management of adult epilepsy, malignant glioma, Alzheimer's disease, migraine headache, motor neuron disease, and other neurologic disorders is highlighted and reviewed. Lastly, common adverse effects of ketogenic therapy in adults, including gastrointestinal symptoms, weight loss, and transient dyslipidemia are discussed.
Collapse
Affiliation(s)
- Tanya J W McDonald
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 2-147, Baltimore, Maryland, 21287, USA
| | - Mackenzie C Cervenka
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 2-147, Baltimore, Maryland, 21287, USA.
| |
Collapse
|
230
|
Dahlgren K, Gibas KJ. Ketogenic diet, high intensity interval training (HIIT) and memory training in the treatment of mild cognitive impairment: A case study. Diabetes Metab Syndr 2018; 12:819-822. [PMID: 29678606 DOI: 10.1016/j.dsx.2018.04.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 04/10/2018] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) deaths have increased by 89% since 2000. This alarming trajectory of neurological disease highlights the failure of current best practice. Deteriorating brain fuel supply is the nemesis of intact neurological health. Cerebral hypo-metabolism associated with AD occurs years before onset. Both the ketogenic diet and calorie restriction (fasting) lead to a compensatory rise in ketones to improve energy deficits in the brain derived from cerebral insulin resistance. Two forms of ketone bodies, β-hydroxybutyrate and acetoacetate, fuel the brain during starvation, fasting and strenuous exercise. Ketones are neuroprotective agents that shelter the aging brain from memory loss and neurodegeneration. Induced ketone production has been shown to ameliorate mitochondrial function, reduce the expression of apoptotic and inflammatory mediators and provide neuroprotection to cells (Lange et al., 2017). This case study highlights an innovative research design aimed at attenuating memory decline in a 57 year old female previously diagnosed with comorbid mild cognitive impairment (MCI) and metabolic syndrome (MetS). Mild cognitive impairment is a predementia syndrome known to precede AD (Michaud et al, 2017). The 12-week intervention included ketogenic nutrition protocol, high intensity interval training (HIIT) and memory training using the PEAK brain training app. Memory function was assessed via the MoCA (Montreal Cognitive Assessment) pre/post intervention. Physiological biomarkers for MetS including HOMA-IR(homeostatic model assessment of insulin resistance), triglyceride/HDL ratio, HgA1c, fasting triglycerides and HDL were measured pre/post intervention. MoCA baseline score was 22/30 (MCI); post intervention score: 30/30 (normal). MetS biomarker improvements also reflected statistical significance.
Collapse
Affiliation(s)
- Kaitlyn Dahlgren
- Human Bioenergetics & Applied Health Science, Bethel University, MN, USA.
| | - Kelly J Gibas
- Human Bioenergetics & Applied Health Science, Bethel University, MN, USA.
| |
Collapse
|
231
|
Zilberter T, Zilberter Y. Ketogenic Ratio Determines Metabolic Effects of Macronutrients and Prevents Interpretive Bias. Front Nutr 2018; 5:75. [PMID: 30214902 PMCID: PMC6125377 DOI: 10.3389/fnut.2018.00075] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
|
232
|
McDonald TJW, Cervenka MC. The Expanding Role of Ketogenic Diets in Adult Neurological Disorders. Brain Sci 2018; 8:E148. [PMID: 30096755 PMCID: PMC6119973 DOI: 10.3390/brainsci8080148] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/15/2022] Open
Abstract
The current review highlights the evidence supporting the use of ketogenic diet therapies in the management of adult epilepsy, adult malignant glioma and Alzheimer's disease. An overview of the scientific literature, both preclinical and clinical, in each area is presented and management strategies for addressing adverse effects and compliance are discussed.
Collapse
Affiliation(s)
- Tanya J W McDonald
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 2-147, Baltimore, MD 21287, USA.
| | - Mackenzie C Cervenka
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 2-147, Baltimore, MD 21287, USA.
| |
Collapse
|
233
|
Schoeler NE, Leu C, Balestrini S, Mudge JM, Steward CA, Frankish A, Leung M, Mackay M, Scheffer I, Williams R, Sander JW, Cross JH, Sisodiya SM. Genome-wide association study: Exploring the genetic basis for responsiveness to ketogenic dietary therapies for drug-resistant epilepsy. Epilepsia 2018; 59:1557-1566. [PMID: 30009487 PMCID: PMC6099477 DOI: 10.1111/epi.14516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/19/2018] [Indexed: 02/04/2023]
Abstract
OBJECTIVE With the exception of specific metabolic disorders, predictors of response to ketogenic dietary therapies (KDTs) are unknown. We aimed to determine whether common variation across the genome influences the response to KDT for epilepsy. METHODS We genotyped individuals who were negative for glucose transporter type 1 deficiency syndrome or other metabolic disorders, who received KDT for epilepsy. Genotyping was performed with the Infinium HumanOmniExpressExome Beadchip. Hospital records were used to obtain demographic and clinical data. KDT response (≥50% seizure reduction) at 3-month follow-up was used to dissect out nonresponders and responders. We then performed a genome-wide association study (GWAS) in nonresponders vs responders, using a linear mixed model and correcting for population stratification. Variants with minor allele frequency <0.05 and those that did not pass quality control filtering were excluded. RESULTS After quality control filtering, the GWAS of 112 nonresponders vs 123 responders revealed an association locus at 6p25.1, 61 kb upstream of CDYL (rs12204701, P = 3.83 × 10-8 , odds ratio [A] = 13.5, 95% confidence interval [CI] 4.07-44.8). Although analysis of regional linkage disequilibrium around rs12204701 did not strengthen the likelihood of CDYL being the candidate gene, additional bioinformatic analyses suggest it is the most likely candidate. SIGNIFICANCE CDYL deficiency has been shown to disrupt neuronal migration and to influence susceptibility to epilepsy in mice. Further exploration with a larger replication cohort is warranted to clarify whether CDYL is the causal gene underlying the association signal.
Collapse
Affiliation(s)
- Natasha E. Schoeler
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUK
- UCL Great Ormond Street Institute of Child HealthLondonUK
| | - Costin Leu
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUK
- NIHR University College London Hospitals Biomedical Research CentreUCL Institute of NeurologyLondonUK
| | - Simona Balestrini
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUK
- Chalfont Centre for EpilepsyChalfont St PeterUK
| | - Jonathan M. Mudge
- European Molecular Biology LaboratoryWellcome Genome CampusEuropean Bioinformatics InstituteCambridgeUK
| | | | - Adam Frankish
- European Molecular Biology LaboratoryWellcome Genome CampusEuropean Bioinformatics InstituteCambridgeUK
| | - Mary‐Anne Leung
- Children's Neurosciences CentreGuy's and St Thomas’ NHS Foundation TrustLondonUK
| | - Mark Mackay
- Department of PaediatricsThe University of MelbourneRoyal Children's HospitalMelbourneVic.Australia
- Murdoch Children's Research InstituteMelbourneVic.Australia
| | - Ingrid Scheffer
- Department of PaediatricsThe University of MelbourneRoyal Children's HospitalMelbourneVic.Australia
- Epilepsy Research CentreDepartment of MedicineThe University of MelbourneAustin HealthMelbourneVic.Australia
- Austin HealthFlorey Institute of Neurosciences and Mental HealthMelbourneVic.Australia
| | - Ruth Williams
- Children's Neurosciences CentreGuy's and St Thomas’ NHS Foundation TrustLondonUK
| | - Josemir W. Sander
- NIHR University College London Hospitals Biomedical Research CentreUCL Institute of NeurologyLondonUK
- Chalfont Centre for EpilepsyChalfont St PeterUK
- Stichting Epilepsie Instellingen Nederland (SEIN)HeemstedeThe Netherlands
| | - J. Helen Cross
- UCL Great Ormond Street Institute of Child HealthLondonUK
- Great Ormond Street Hospital for ChildrenLondonUK
- Young EpilepsyLingfieldUK
| | - Sanjay M. Sisodiya
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUK
- Chalfont Centre for EpilepsyChalfont St PeterUK
| |
Collapse
|
234
|
Wu L, Zhang X, Zhao L. Human ApoE Isoforms Differentially Modulate Brain Glucose and Ketone Body Metabolism: Implications for Alzheimer's Disease Risk Reduction and Early Intervention. J Neurosci 2018; 38:6665-6681. [PMID: 29967007 PMCID: PMC6067075 DOI: 10.1523/jneurosci.2262-17.2018] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 06/02/2018] [Accepted: 06/03/2018] [Indexed: 01/16/2023] Open
Abstract
Humans possess three genetic isoforms of apolipoprotein E (ApoE)-ApoE2, ApoE3, and ApoE4-that confer differential risk for Alzheimer's disease (AD); however, the underlying mechanisms are poorly understood. This study sought to investigate the impact of human ApoE isoforms on brain energy metabolism, an area significantly perturbed in preclinical AD. A TaqMan custom array was performed to examine the expression of a total of 43 genes involved in glucose and ketone body transport and metabolism, focusing on pathways leading to the generation of acetyl-CoA, in human ApoE gene-targeted replacement female mice. Consistent with our previous findings, brains expressing ApoE2 exhibited the most robust profile, whereas brains expressing ApoE4 displayed the most deficient profile on the uptake and metabolism of glucose, the primary fuel for the brain. Specifically, the three ApoE brains differed significantly in facilitated glucose transporters, which mediate the entry of glucose into neurons, and hexokinases, which act as the "gateway enzyme" in glucose metabolism. Interestingly, on the uptake and metabolism of ketone bodies, the secondary energy source for the brain, ApoE2 and ApoE4 brains showed a similar level of robustness, whereas ApoE3 brains presented a relatively deficient profile. Further, ingenuity pathway analysis indicated that the PPAR-γ/PGC-1α signaling pathway could be activated in the ApoE2 brain and inhibited in the ApoE4 brain. Notably, PGC-1α overexpression ameliorated ApoE4-induced deficits in glycolysis and mitochondrial respiration. Overall, our data provide additional evidence that human ApoE isoforms differentially modulate brain bioenergetic metabolism, which could serve as a potential mechanism contributing to their discrete risk impact in AD.SIGNIFICANCE STATEMENT We uncovered hexokinase as a key cytosolic point in the glucose metabolism that is differentially modulated by the three ApoE genotypes. The differences in hexokinase expression and activity exhibited in the three ApoE brains may underlie their distinct impact on brain glucose utilization and further susceptibility to AD. Therefore, a therapeutic approach that could circumvent the deficiencies in the cytosolic metabolism of glucose by providing glucose metabolizing intermediates, e.g., pyruvate, may hold benefits for ApoE4 carriers, who are at high risk for AD. The bioenergetic robustness may translate into enhanced synaptic activity and, ultimately, reduces the risk of developing AD and/or delays the onset of clinical manifestation.
Collapse
Affiliation(s)
- Long Wu
- Department of Pharmacology and Toxicology, School of Pharmacy and
| | - Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy and
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy and
- Neuroscience Graduate Program, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
235
|
Torosyan N, Sethanandha C, Grill JD, Dilley ML, Lee J, Cummings JL, Ossinalde C, Silverman DH. Changes in regional cerebral blood flow associated with a 45 day course of the ketogenic agent, caprylidene, in patients with mild to moderate Alzheimer's disease: Results of a randomized, double-blinded, pilot study. Exp Gerontol 2018; 111:118-121. [PMID: 30006299 DOI: 10.1016/j.exger.2018.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/14/2018] [Accepted: 07/10/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Caprylidene is a ketogen that, when metabolized, produces the ketones beta-hydroxybutyrate and acetoacetate, which can cross the blood brain barrier. It has been hypothesized that ketone bodies can be used as an alternate energy source by neurons with impaired glucose utilization. Caprylidene has been shown to improve cognition in patients with mild-to-moderate Alzheimer's disease (AD) who lacked an AD-predisposing allele (ɛ4) of the gene for apolipoprotein E. In this pilot study, we examined the effects of caprylidene on regional cerebral blood flow (rCBF) in patients with mild to moderate AD. METHODS Sixteen subjects with mild-to-moderate AD, based on NINCDS-ADRDA criteria, were enrolled in a double-blinded, placebo-controlled, randomized clinical trial. Fourteen subjects received treatment with caprylidene, and 2 subjects were given placebo. Subjects received 4 15O-water PET scans over the course of the study to assess rCBF: once before receiving a standard caprylidene or placebo dose and 90 min after the dose, on the first day and after 45 days of daily caprylidene or placebo consumption. The scans were examined by standardized volumes of interest (sVOI) and voxel-based statistical parametric mapping (spm) methods of analysis. RESULTS Subjects lacking an ɛ4 allele had significantly elevated rCBF in the left superior lateral temporal cortex by sVOI analysis after adopting a caprylidene diet for 45 days (p = 0.04), which was further corroborated by spm. The anterior cerebellum, left inferior temporal cortex, and hypothalamus were also found by spm to be regions of long-term increase in rCBF in these subjects. In contrast, patients who possessed the ɛ4 allele did not display these changes in rCBF. CONCLUSION Daily ingestion of caprylidene over 45 days was associated with increased blood flow in specific brain regions in patients lacking an apolipoprotein ɛ4 allele.
Collapse
Affiliation(s)
- Nare Torosyan
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA; Institute for Memory Impairments and Neurological Disorders, Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, University of California, Irvine, 3206 Biological Sciences III, Irvine, CA 92697, USA
| | - Chakmeedaj Sethanandha
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA; Mahidol University, 999 Phutthamonthon Sai 4 Rd, Chang Wat Nakhon Pathom 73170, Thailand
| | - Joshua D Grill
- Institute for Memory Impairments and Neurological Disorders, Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, University of California, Irvine, 3206 Biological Sciences III, Irvine, CA 92697, USA
| | - Michelle L Dilley
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA
| | - Jooyeon Lee
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA
| | - Jeffrey L Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W Bonneville Ave, Las Vegas, NV 89106, USA
| | - Celine Ossinalde
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA
| | - Daniel H Silverman
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California Los Angeles, 200 Medical Plaza, Los Angeles, CA 90095, USA.
| |
Collapse
|
236
|
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. [PMID: 29471055 DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [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]
|
237
|
Dominguez LJ, Barbagallo M. Nutritional prevention of cognitive decline and dementia. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:276-290. [PMID: 29957766 PMCID: PMC6179018 DOI: 10.23750/abm.v89i2.7401] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 04/28/2018] [Indexed: 12/28/2022]
Abstract
Cognitive impairment results from a complex interplay of many factors. The most important independent predictor of cognitive decline is age but other contributing factors include demographic, genetic, socio-economic, and environmental parameters, including nutrition. The number of persons with cognitive decline and dementia will increase in the next decades in parallel with aging of the world population. Effective pharmaceutical treatments for age-related cognitive decline are lacking, emphasizing the importance of prevention strategies. There is extensive evidence supporting a relationship between diet and cognitive functions. Thus, nutritional approaches to prevent or slow cognitive decline could have a remarkable public health impact. Several dietary components and supplements have been examined in relation to their association with the development of cognitive decline. A number of studies have examined the role of dietary patterns on late-life cognition, with accumulating evidence that combinations of foods and nutrients may act synergistically to provide stronger benefit than those conferred by individual dietary components. Higher adherence to the Mediterranean dietary pattern has been associated with decreased cognitive decline and incident AD. Another dietary pattern with neuroprotective actions is the Dietary Approach to Stop Hypertension (DASH). The combination of these two dietary patterns has been associated with slower rates of cognitive decline and significant reduction in incident AD. This review evaluates the evidence for the effects of some dietary components, supplements, and dietary patterns as neuroprotective, with potential to delay cognitive decline and the onset of dementia.
Collapse
Affiliation(s)
- Ligia J Dominguez
- Geriatric Unit, Dept. of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy.
| | - Mario Barbagallo
- Geriatric Unit, Dept. of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy.
| |
Collapse
|
238
|
Structural and Functional Rescue of Chronic Metabolically Stressed Optic Nerves through Respiration. J Neurosci 2018; 38:5122-5139. [PMID: 29760184 DOI: 10.1523/jneurosci.3652-17.2018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/27/2018] [Accepted: 04/24/2018] [Indexed: 01/05/2023] Open
Abstract
Axon degeneration can arise from metabolic stress, potentially a result of mitochondrial dysfunction or lack of appropriate substrate input. In this study, we investigated whether the metabolic vulnerability observed during optic neuropathy in the DBA/2J (D2) model of glaucoma is due to dysfunctional mitochondria or impaired substrate delivery to axons, the latter based on our observation of significantly decreased glucose and monocarboxylate transporters in D2 optic nerve (ON), human ON, and mice subjected to acute glaucoma injury. We placed both sexes of D2 mice destined to develop glaucoma and mice of a control strain, the DBA/2J-Gpnmb+, on a ketogenic diet to encourage mitochondrial function. Eight weeks of the diet generated mitochondria, improved energy availability by reversing monocarboxylate transporter decline, reduced glial hypertrophy, protected retinal ganglion cells and their axons from degeneration, and maintained physiological signaling to the brain. A robust antioxidant response also accompanied the response to the diet. These results suggest that energy compromise and subsequent axon degeneration in the D2 is due to low substrate availability secondary to transporter downregulation.SIGNIFICANCE STATEMENT We show axons in glaucomatous optic nerve are energy depleted and exhibit chronic metabolic stress. Underlying the metabolic stress are low levels of glucose and monocarboxylate transporters that compromise axon metabolism by limiting substrate availability. Axonal metabolic decline was reversed by upregulating monocarboxylate transporters as a result of placing the animals on a ketogenic diet. Optic nerve mitochondria responded capably to the oxidative phosphorylation necessitated by the diet and showed increased number. These findings indicate that the source of metabolic challenge can occur upstream of mitochondrial dysfunction. Importantly, the intervention was successful despite the animals being on the cusp of significant glaucoma progression.
Collapse
|
239
|
Pinto A, Bonucci A, Maggi E, Corsi M, Businaro R. Anti-Oxidant and Anti-Inflammatory Activity of Ketogenic Diet: New Perspectives for Neuroprotection in Alzheimer's Disease. Antioxidants (Basel) 2018; 7:E63. [PMID: 29710809 PMCID: PMC5981249 DOI: 10.3390/antiox7050063] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022] Open
Abstract
The ketogenic diet, originally developed for the treatment of epilepsy in non-responder children, is spreading to be used in the treatment of many diseases, including Alzheimer’s disease. The main activity of the ketogenic diet has been related to improved mitochondrial function and decreased oxidative stress. B-Hydroxybutyrate, the most studied ketone body, has been shown to reduce the production of reactive oxygen species (ROS), improving mitochondrial respiration: it stimulates the cellular endogenous antioxidant system with the activation of nuclear factor erythroid-derived 2-related factor 2 (Nrf2), it modulates the ratio between the oxidized and reduced forms of nicotinamide adenine dinucleotide (NAD⁺/NADH) and it increases the efficiency of electron transport chain through the expression of uncoupling proteins. Furthermore, the ketogenic diet performs anti-inflammatory activity by inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) activation and nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome as well as inhibiting histone deacetylases (HDACs), improving memory encoding. The underlying mechanisms and the perspectives for the treatment of Alzheimer’s disease are discussed.
Collapse
Affiliation(s)
- Alessandro Pinto
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy.
| | - Alessio Bonucci
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| | - Elisa Maggi
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| | - Mariangela Corsi
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| |
Collapse
|
240
|
Potential Synergies of β-Hydroxybutyrate and Butyrate on the Modulation of Metabolism, Inflammation, Cognition, and General Health. J Nutr Metab 2018; 2018:7195760. [PMID: 29805804 PMCID: PMC5902005 DOI: 10.1155/2018/7195760] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/06/2018] [Accepted: 02/28/2018] [Indexed: 12/31/2022] Open
Abstract
The low-carbohydrate high-fat diet (LCHFD), also known as the ketogenic diet, has cycled in and out of popularity for decades as a therapeutic program to treat metabolic syndrome, weight mismanagement, and drug-resistant disorders as complex as epilepsy, cancer, dementia, and depression. Despite the benefits of this diet, health care professionals still question its safety due to the elevated serum ketones it induces and the limited dietary fiber. To compound the controversy, patient compliance with the program is poor due to the restrictive nature of the diet and symptoms related to energy deficit and gastrointestinal adversity during the introductory and energy substrate transition phase of the diet. The studies presented here demonstrate safety and efficacy of the diet including the scientific support and rationale for the administration of exogenous ketone bodies and ketone sources as a complement to the restrictive dietary protocol or as an alternative to the diet. This review also highlights the synergy provided by exogenous ketone, β-hydroxybutyrate (BHB), accompanied by the short chain fatty acid, butyrate (BA) in the context of cellular and physiological outcomes. More work is needed to unveil the molecular mechanisms by which this program provides health benefits.
Collapse
|
241
|
Schloss J, Rainforest J. Medical synopsis: New metabolic markers to help diagnosis and assess disease progression for Alzheimer’s disease, normal pressure hydrocephalus and brain tumours. ADVANCES IN INTEGRATIVE MEDICINE 2018. [DOI: 10.1016/j.aimed.2018.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
242
|
Barbanti P, Fofi L, Aurilia C, Egeo G, Caprio M. Ketogenic diet in migraine: rationale, findings and perspectives. Neurol Sci 2018; 38:111-115. [PMID: 28527061 DOI: 10.1007/s10072-017-2889-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ketogenic diet (KD) is an established treatment for refractory pediatric epilepsy and a promising therapy for diverse neurological diseases. Clinical data on KD in migraine-obtained from 150 patients investigated in case reports and prospective studies-suggest that KD may be a rapid onset effective prophylaxis for episodic and chronic migraine. KD would contribute to restore brain excitability and metabolism and to counteract neuroinflammation in migraine, although its precise mechanism is still unclear. Randomized controlled studies are needed to confirm the usefulness of KD in migraine and to investigate its optimal duration, repeatability, feasibility in normal weight subjects, efficacy in pediatric population and association to conventional migraine prophylaxis.
Collapse
Affiliation(s)
- Piero Barbanti
- Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana, via Della Pisana 235, Rome, 00163, Italy.
| | - Luisa Fofi
- Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana, via Della Pisana 235, Rome, 00163, Italy
| | - Cinzia Aurilia
- Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana, via Della Pisana 235, Rome, 00163, Italy
| | - Gabriella Egeo
- Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana, via Della Pisana 235, Rome, 00163, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
243
|
Ibhazehiebo K, Gavrilovici C, de la Hoz CL, Ma SC, Rehak R, Kaushik G, Meza Santoscoy PL, Scott L, Nath N, Kim DY, Rho JM, Kurrasch DM. A novel metabolism-based phenotypic drug discovery platform in zebrafish uncovers HDACs 1 and 3 as a potential combined anti-seizure drug target. Brain 2018; 141:744-761. [PMID: 29373639 PMCID: PMC5837409 DOI: 10.1093/brain/awx364] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 10/29/2017] [Accepted: 11/05/2017] [Indexed: 01/01/2023] Open
Abstract
Despite the development of newer anti-seizure medications over the past 50 years, 30-40% of patients with epilepsy remain refractory to treatment. One explanation for this lack of progress is that the current screening process is largely biased towards transmembrane channels and receptors, and ignores intracellular proteins and enzymes that might serve as efficacious molecular targets. Here, we report the development of a novel drug screening platform that harnesses the power of zebrafish genetics and combines it with in vivo bioenergetics screening assays to uncover therapeutic agents that improve mitochondrial health in diseased animals. By screening commercially available chemical libraries of approved drugs, for which the molecular targets and pathways are well characterized, we were able to reverse-identify the proteins targeted by efficacious compounds and confirm the physiological roles that they play by utilizing other pharmacological ligands. Indeed, using an 870-compound screen in kcna1-morpholino epileptic zebrafish larvae, we uncovered vorinostat (Zolinza™; suberanilohydroxamic acid, SAHA) as a potent anti-seizure agent. We further demonstrated that vorinostat decreased average daily seizures by ∼60% in epileptic Kcna1-null mice using video-EEG recordings. Given that vorinostat is a broad histone deacetylase (HDAC) inhibitor, we then delineated a specific subset of HDACs, namely HDACs 1 and 3, as potential drug targets for future screening. In summary, we have developed a novel phenotypic, metabolism-based experimental therapeutics platform that can be used to identify new molecular targets for future drug discovery in epilepsy.
Collapse
Affiliation(s)
- Kingsley Ibhazehiebo
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Cezar Gavrilovici
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Cristiane L de la Hoz
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Shun-Chieh Ma
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Renata Rehak
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Gaurav Kaushik
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Paola L Meza Santoscoy
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Lucas Scott
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Nandan Nath
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Do-Young Kim
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Jong M Rho
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| |
Collapse
|
244
|
Gardener SL, Rainey-Smith SR, Sohrabi HR, Weinborn M, Verdile G, Fernando WMADB, Lim YY, Harrington K, Burnham S, Taddei K, Masters CL, Macaulay SL, Rowe CC, Ames D, Maruff P, Martins RN. Increased Carbohydrate Intake is Associated with Poorer Performance in Verbal Memory and Attention in an APOE Genotype-Dependent Manner. J Alzheimers Dis 2018; 58:193-201. [PMID: 28387666 DOI: 10.3233/jad-161158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Evidence suggests that a diet low in carbohydrates can impact on cognitive performance among those with Alzheimer's disease (AD). However, there is a lack of data assessing this relationship among cognitively normal (CN) older adults at increased future risk of developing AD due to carriage of the apolipoprotein E (APOE) ɛ4 allele. We assessed the cross-sectional association between carbohydrate intake, cognitive performance, and cerebral amyloid-β (Aβ) load in CN older adults, genotyped for APOEɛ4 allele carrier status. Greater carbohydrate intake was associated with poorer performance in verbal memory in APOEɛ4 allele non-carriers, and poorer performance in attention in APOEɛ4 allele carriers. There were no associations between carbohydrate intake and cerebral Aβ load. These results provide support to the idea that decreasing carbohydrate intake may offer neurocognitive benefits, with specific cognitive domains affected in an APOE genotype-dependent manner. These findings warrant further investigation utilizing a longitudinal study design.
Collapse
Affiliation(s)
- Samantha L Gardener
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia
| | - Hamid R Sohrabi
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia.,Department of Biomedical Sciences, Macquarie University, NSW, Australia
| | - Michael Weinborn
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia.,School of Psychology, University of Western Australia, Crawley, WA, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia.,School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University of Technology, Bentley, WA, Australia
| | - W M A D Binosha Fernando
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia
| | - Yen Ying Lim
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Karra Harrington
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Cooperative Research Centre (CRC) for Mental Health, Carlton South, VIC, Australia
| | - Samantha Burnham
- CSIRO Computational Informatics, Preventative Health Flagship, Floreat, WA, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | | | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - David Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Australia
| | | | - Ralph N Martins
- Centre of Excellence for Alzheimer's disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), Perth, WA, Australia.,Department of Biomedical Sciences, Macquarie University, NSW, Australia
| | | |
Collapse
|
245
|
Perkins M, Wolf AB, Chavira B, Shonebarger D, Meckel JP, Leung L, Ballina L, Ly S, Saini A, Jones TB, Vallejo J, Jentarra G, Valla J. Altered Energy Metabolism Pathways in the Posterior Cingulate in Young Adult Apolipoprotein E ɛ4 Carriers. J Alzheimers Dis 2018; 53:95-106. [PMID: 27128370 PMCID: PMC4942726 DOI: 10.3233/jad-151205] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The APOE gene, encoding apolipoprotein E, is the primary genetic risk factor for late-onset Alzheimer’s disease (AD). Apolipoprotein E ɛ4 allele (APOE4) carriers have alterations in brain structure and function (as measured by brain imaging) even as young adults. Examination of this population is valuable in further identifying details of these functional changes and their association with vulnerability to AD decades later. Previous work demonstrates functional declines in mitochondrial activity in the posterior cingulate cortex, a key region in the default mode network, which appears to be strongly associated with functional changes relevant to AD risk. Here, we demonstrate alterations in the pathways underlying glucose, ketone, and mitochondrial energy metabolism. Young adult APOE4 carriers displayed upregulation of specific glucose (GLUT1 & GLUT3) and monocarboxylate (MCT2) transporters, the glucose metabolism enzyme hexokinase, the SCOT & AACS enzymes involved in ketone metabolism, and complexes I, II, and IV of the mitochondrial electron transport chain. The monocarboxylate transporter (MCT4) was found to be downregulated in APOE4 carriers. These data suggest that widespread dysregulation of energy metabolism in this at-risk population, even decades before possible disease onset. Therefore, these findings support the idea that alterations in brain energy metabolism may contribute significantly to the risk that APOE4 confers for AD.
Collapse
Affiliation(s)
- Michelle Perkins
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Andrew B Wolf
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Bernardo Chavira
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | | | - J P Meckel
- Midwestern University, Glendale, AZ, USA
| | - Lana Leung
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | | | - Sarah Ly
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aman Saini
- Midwestern University, Glendale, AZ, USA
| | | | - Johana Vallejo
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Garilyn Jentarra
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Jon Valla
- Midwestern University, Glendale, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| |
Collapse
|
246
|
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: 41] [Impact Index Per Article: 5.9] [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
|
247
|
Griffith CM, Eid T, Rose GM, Patrylo PR. Evidence for altered insulin receptor signaling in Alzheimer's disease. Neuropharmacology 2018; 136:202-215. [PMID: 29353052 DOI: 10.1016/j.neuropharm.2018.01.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022]
Abstract
Epidemiological data have shown that metabolic disease can increase the propensity for developing cognitive decline and dementia, particularly Alzheimer's disease (AD). While this interaction is not completely understood, clinical studies suggest that both hyper- and hypoinsulinemia are associated with an increased risk for developing AD. Indeed, insulin signaling is altered in post-mortem brain tissue from AD patients and treatments known to enhance insulin signaling can improve cognitive function. Further, clinical evidence has shown that AD patients and mouse models of AD often display alterations in peripheral metabolism. Since insulin is primarily derived from the periphery, it is likely that changes in peripheral insulin levels lead to alterations in central nervous system (CNS) insulin signaling and could contribute to cognitive decline and pathogenesis. Developing a better understanding of the relationship between alterations in peripheral metabolism and cognitive function might provide a foundation for the development of better treatment options for patients with AD. In this article we will begin to piece together the present data defining this relationship by briefly discussing insulin signaling in the periphery and CNS, its role in cognitive function, insulin's relationship to AD, peripheral metabolic alterations in mouse models of AD and how information from these models helps understand the mechanisms through which these changes potentially lead to impairments in insulin signaling in the CNS, and potential ways to target insulin signaling that could improve cognitive function in AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Chelsea M Griffith
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gregory M Rose
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA
| | - Peter R Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, IL 62901, USA.
| |
Collapse
|
248
|
Orešič M, Anderson G, Mattila I, Manoucheri M, Soininen H, Hyötyläinen T, Basignani C. Targeted Serum Metabolite Profiling Identifies Metabolic Signatures in Patients with Alzheimer's Disease, Normal Pressure Hydrocephalus and Brain Tumor. Front Neurosci 2018; 11:747. [PMID: 29375291 PMCID: PMC5767271 DOI: 10.3389/fnins.2017.00747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/21/2017] [Indexed: 01/21/2023] Open
Abstract
Progression to AD is preceded by elevated levels of 2,4-dihydroxybutanoic acid (2,4-DHB), implicating hypoxia in early pathogenesis. Since hypoxia may play a role in multiple CNS disorders, we investigated serum metabolite profiles across three disorders, AD, Normal Pressure Hydrocephalus (NPH) and brain tumors (BT). Blood samples were collected from 27 NPH and 20 BT patients. The profiles of 21 metabolites were examined. Additionally, data from 37 AD patients and 46 controls from a previous study were analyzed together with the newly acquired data. No differences in 2,4-DHB were found across AD, NPH and BT samples. In the BT group, the fatty acids were increased as compared to HC and NPH groups, while the ketone body 3-hydroxybutyrate was increased as compared to AD. Glutamic acid was increased in AD as compared to the HC group. In the AD group, 3-hydroxybutyrate tended to be decreased with respect to all other groups (mean values −30% or more), but the differences were not statistically significant. Serine was increased in NPH as compared to BT. In conclusion, AD, NPH and BT have different metabolic profiles. This preliminary study may help in identifying the blood based markers that are specific to these three CNS diseases.
Collapse
Affiliation(s)
- Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Gabriella Anderson
- Florida Hospital Orlando, Neuroscience Research Institute, Orlando, FL, United States
| | - Ismo Mattila
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Manoucher Manoucheri
- Florida Hospital Orlando, Neuroscience Research Institute, Orlando, FL, United States
| | - Hilkka Soininen
- Department of Neurology, Neuro Center, Kuopio University Hospital, Kuopio, Finland.,Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tuulia Hyötyläinen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Chemistry, Örebro University, Örebro, Sweden
| | - Cherlynn Basignani
- Florida Hospital Orlando, Neuroscience Research Institute, Orlando, FL, United States
| |
Collapse
|
249
|
Botchway BO, Moore MK, Akinleye FO, Iyer IC, Fang M. Nutrition: Review on the Possible Treatment for Alzheimer’s Disease. J Alzheimers Dis 2018; 61:867-883. [DOI: 10.3233/jad-170874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Benson O.A. Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Masania K. Moore
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Faith O. Akinleye
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Ishwari C. Iyer
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
250
|
Veiga S, Wahrheit J, Rodríguez-Martín A, Sonntag D. Quantitative Metabolomics in Alzheimer's Disease: Technical Considerations for Improved Reproducibility. Methods Mol Biol 2018; 1779:463-470. [PMID: 29886550 DOI: 10.1007/978-1-4939-7816-8_28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Metabolomics is the comprehensive analysis of small molecules (metabolites) that are intermediates or endpoints of metabolism. Since metabolites change more rapidly to both external and internal stimuli than genes and proteins, metabolomics provides a more sensitive tool to study physiological changes to a wide range of factors such age, medication, or disease status. Therefore, metabolomics is being increasingly used for the study of several pathological states, including complex diseases like Alzheimer's disease (AD).Both untargeted and targeted metabolomics have been applied for AD and both have provided diagnostic algorithms that accurately discriminate healthy patients from patients with AD by combining different metabolites. However, none of these algorithms have been replicated in larger, different cohorts, and a consensus in methodology has been claimed by the scientific community. The AbsoluteIDQ® p180 Kit (Biocrates, Life Science AG, Innsbruck, Austria) is to date the only commercially available, validated, and standardized assay that measures up to 188 metabolites in biological samples. This kit unifies methodology in a common user manual and provides quantitative measurements of metabolites, thus facilitating an easier comparison among studies and reducing the technical variability that might contribute to replication failures. Nevertheless, recent studies showed no replication even when using this kit, suggesting that additional measures should be taken to achieve replication of metabolite-based discriminative algorithms. The aim of this chapter is to provide technical guidance on how to apply quantitative metabolomic data to the definition of discriminative algorithms for the diagnosis of neurodegenerative diseases such as AD. This chapter will provide an overview of technical aspects on the whole process, from blood sampling to raw data handling, and will highlight several technical aspects in the process that could hamper replication attempts even when using validated and standardized assays, such as the AbsoluteIDQ® p180 Kit.
Collapse
|