1
|
Wei DX, Chen Z. Current situation and challenge of exogenous 3-hydroxybutyrate derived from polyhydroxyalkanoates for elderly health: A review. Int J Biol Macromol 2025; 285:138328. [PMID: 39631604 DOI: 10.1016/j.ijbiomac.2024.138328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
3-Hydroxybutyrate (3HB), predominantly found in the liver, muscles, and brain, is the most important endogenous ketone body in humans. During prolonged fasting and starvation, 3HB can partially replace glucose to meet some of the body's energy needs. In recent years, the resurgence of the ketogenic diet (KD) and systematic exploration of the favorable biocompatibility of polyhydroxyalkanoates (PHAs), the precursor polymer to 3HB, have led to numerous reports indicating that the benefits of exogenous 3HB supplementation outweigh the drawbacks, particularly for middle-aged and elderly individuals. In this review, the physicochemical properties, physiological functions, and biosafety of 3HB in the elderly population are introduced. The effects of exogenous 3HB supplements, including KD, ketone esters, 1,3-butanediol, and ketone salts, on the elderly are compared. It is proposed that exogenous 3HB derived from PHAs is optimal for this population. Exogenous 3HB supplementation for elderly health maintenance and disease treatment is summarized, with an outline of four challenges related to the mechanistic and clinical research of exogenous 3HB supplementation for elderly health. This aims to explore its novel therapeutic potential as a small molecule in the context of aging.
Collapse
Affiliation(s)
- Dai-Xu Wei
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China.
| | - Zongcun Chen
- Department of Endocrinology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, China.
| |
Collapse
|
2
|
Rühling MR, Hartmann H, Das AM. Simplification of Dietary Treatment in Pharmacoresistant Epilepsy: Impact of C8 and C10 Fatty Acids on Sirtuins of Neuronal Cells In Vitro. Nutrients 2024; 16:1678. [PMID: 38892612 PMCID: PMC11174688 DOI: 10.3390/nu16111678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pharmacotherapy is the therapeutic mainstay in epilepsy; however, in about 30% of patients, epileptic seizures are drug-resistant. A ketogenic diet (KD) is an alternative therapeutic option. The mechanisms underlying the anti-seizure effect of a KD are not fully understood. Epileptic seizures lead to an increased energy demand of neurons. An improvement in energy provisions may have a protective effect. C8 and C10 fatty acids have been previously shown to activate mitochondrial function in vitro. This could involve sirtuins (SIRTs) as regulatory elements of energy metabolism. The aim of the present study was to investigate whether ß-hydroxybutyrate (ßHB), C8 fatty acids, C10 fatty acids, or a combination of C8 and C10 (250/250 µM) fatty acids, which all increase under a KD, could up-regulate SIRT1, -3, -4, and -5 in HT22 hippocampal murine neurons in vitro. Cells were incubated for 1 week in the presence of these metabolites. The sirtuins were measured at the enzyme (fluorometrically), protein (Western blot), and gene expression (PCR) levels. In hippocampal cells, the C8, C10, and C8 and C10 incubations led to increases in the sirtuin levels, which were not inferior to a ßHB incubation as the 'gold standard'. This may indicate that both C8 and C10 fatty acids are important for the antiepileptic effect of a KD. A KD may be replaced by nutritional supplements of C8 and C10 fatty acids, which could facilitate the diet.
Collapse
|
3
|
Putananickal N, Gross EC, Orsini AL, Schmidt S, Hafner P, Gocheva V, Nagy S, Henzi BC, Rubino D, Schädelin S, Sandor P, Fischer D. Metabolic markers of short and long-term exogenous DL-beta-hydroxybutyrate supplementation in episodic migraine patients: an exploratory analysis of a randomized-controlled-trial. Front Pharmacol 2023; 14:1172483. [PMID: 37214431 PMCID: PMC10192563 DOI: 10.3389/fphar.2023.1172483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Background: Emerging findings propose that the pathophysiology of migraine may be associated with dysfunctional metabolic mechanisms. Recent findings suggest that migraine attacks are a response to the cerebral energy deficit, and ingestion of ketone bodies stabilizes the generation of a migraine attack. Based on these findings, ketone body supplementation is postulated as a prophylactic treatment approach to restore cerebral metabolism deficiency. Metabolic markers are unexplored after exogenous ketone body supplementation in episodic migraineurs. Therefore, the present single-arm uncontrolled explorative analysis evaluated blood ketone body and glucose concentration after short and long-term 6 g exogenous DL-Mg-Ca-beta-hydroxybutyrate (DL-βHB) supplementation. Methods: The presented data are part of the MigraKet randomized-control cross-over clinical trial of 41 episodic migraineurs (Number NCT03132233). Patients were given a single dose of 6 g DL-βHB. Ketone body and glucose blood concentration were assessed before intake, 20, and 40 min after DL-βHB intake. Ketone body, glucose concentration and glycated hemoglobin values were evaluated after 12 weeks of 18 g DL-βHB ingestion (total dose), taken three times daily (6g/dose; 3x/day). Linear models explored the association between the ketone body and glucose levels. Results: Ketone body concentration increased within-group to a mean of 0.46 (0.30) mmol/L after 40 min post- DL-βHB supplementation [estimate = 0.24 mmol/L, CI = (0.20.0.27), p < 0.01]. This within-group increase of ketone body concentration did not change after repeated daily intake of DL-βHB supplementation over 12 weeks [estimate = 0.00 mmol/L, CI = (-0.03.0.04), p = 0.794]. DL-βHB intake significantly reduced blood glucose concentration within-group from a mean baseline of 4.91 (0.42) mmol/L to 4.75 (0.47) mmol/L 40 min post-DL-βHB supplementation [estimate = -0.16 mmol/L, CI = (-0.15, 0.03), p < 0.01]. Repeated DL-βHB supplementation for 12 weeks showed no change within-group in acute ketone bodies concentration [estimate = 0.00 mmol/L, CI = (-0.03.0.04), p = 0.794] and in the HbA1c value [estimate = 0.02, CI = (-0.07.0.11), p = 0.69]. Conclusion: A single dose of 6 g DL-βHB significantly elevated blood ketone bodies and decreased blood glucose concentration within-group in episodic migraineurs. Long-term DL-βHB supplementation for 12 weeks showed no effect within-group on acute ketone body concentration and had not impact on HbA1c. The elevation of the ketone body concentration was moderate, indicating that nutritional ketosis was not reached. Therefore, a dose higher than 6 g of DL-βHB is required to reach the nutritional level of ketosis. ClinicalTrials.gov Identifier: NCT03132233.
Collapse
Affiliation(s)
- Niveditha Putananickal
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Elena C. Gross
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Anna-Lena Orsini
- Neurology, University of Basel Hospital, University of Basel, Basel, Switzerland
| | - Simone Schmidt
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Patricia Hafner
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Vanya Gocheva
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Sara Nagy
- Neurology, University of Basel Hospital, University of Basel, Basel, Switzerland
| | - Bettina C. Henzi
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Daniela Rubino
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| | - Sabine Schädelin
- Department of Clinical Research, Clinical Trial Unit, University of Basel Hospital, University of Basel, Basel, Switzerland
| | | | - Dirk Fischer
- Division of Neuropaediatrics, University of Basel Children’s Hospital, University of Basel, Basel, Switzerland
| |
Collapse
|
4
|
Lopaschuk GD, Dyck JRB. Ketones and the cardiovascular system. NATURE CARDIOVASCULAR RESEARCH 2023; 2:425-437. [PMID: 39196044 DOI: 10.1038/s44161-023-00259-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/28/2023] [Indexed: 08/29/2024]
Abstract
Ketone bodies, the main one being β-hydroxybutyrate, have emerged as important regulators of the cardiovascular system. In healthy individuals, as well as in individuals with heart failure or post-myocardial infarction, ketones provide a supplemental energy source for both the heart and the vasculature. In the failing heart, this additional energy may contribute to improved cardiac performance, whereas increasing ketone oxidation in vascular smooth muscle and endothelial cells enhances cell proliferation and prevents blood vessel rarefication. Ketones also have important actions in signaling pathways, posttranslational modification pathways and gene transcription; many of which modify cell proliferation, inflammation, oxidative stress, endothelial function and cardiac remodeling. Attempts to therapeutically increase ketone delivery to the cardiovascular system are numerous and have shown mixed results in terms of effectiveness. Here we review the bioenergetic and signaling effects of ketones on the cardiovascular system, and we discuss how ketones can potentially be used to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Falkenhain K, Daraei A, Forbes SC, Little JP. Effects of Exogenous Ketone Supplementation on Blood Glucose: A Systematic Review and Meta-analysis. Adv Nutr 2022; 13:1697-1714. [PMID: 35380602 PMCID: PMC9526861 DOI: 10.1093/advances/nmac036] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 01/28/2023] Open
Abstract
Recently developed ketone (monoester or salt) supplements acutely elevate blood β-hydroxybutyrate (BHB) exogenously without prolonged periods of fasting or carbohydrate restriction. Previous (small-scale) studies have found a blood glucose-lowering effect of exogenous ketones. This study aimed to systematically review available evidence and conduct meta-analyses of studies reporting on exogenous ketones and blood glucose. We searched 6 electronic databases on 13 December 2021 for randomized and nonrandomized trials of any length that reported on the use of exogenous ketones. We calculated raw mean differences (MDs) in blood BHB and glucose in 2 main analyses: 1) after compared with before acute ingestion of exogenous ketones and 2) following acute ingestion of exogenous ketones compared with a comparator supplement. We pooled effect sizes using random-effects models and performed prespecified subgroup analyses to examine the effect of potential explanatory factors, including study population, exercise, blood BHB, and supplement type, dosing, and timing. Risk of bias was examined using Cochrane's risk-of-bias tools. Studies that could not be meta-analyzed were summarized narratively. Forty-three trials including 586 participants are summarized in this review. Following ingestion, exogenous ketones increased blood BHB (MD = 1.73 mM; 95% CI: 1.26, 2.21 mM; P < 0.001) and decreased mean blood glucose (MD = -0.54 mM; 95% CI: -0.68, -0.40 mM; P < 0.001). Similarly, when compared with placebo, blood BHB increased (MD = 1.98 mM; 95% CI: 1.52, 2.45 mM; P < 0.001) and blood glucose decreased (MD = -0.47 mM; 95% CI: -0.57, -0.36 mM; P < 0.001). Across both analyses, significantly greater effects were seen with ketone monoesters compared with salts (P < 0.001). The available evidence indicates that acute ingestion of exogenous ketones leads to increased blood BHB and decreased blood glucose. Limited evidence on prolonged ketone supplementation was found.
Collapse
Affiliation(s)
| | - Ali Daraei
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Scott C Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, Manitoba, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
6
|
Exogenous Ketone Supplements in Athletic Contexts: Past, Present, and Future. Sports Med 2022; 52:25-67. [PMID: 36214993 PMCID: PMC9734240 DOI: 10.1007/s40279-022-01756-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 12/15/2022]
Abstract
The ketone bodies acetoacetate (AcAc) and β-hydroxybutyrate (βHB) have pleiotropic effects in multiple organs including brain, heart, and skeletal muscle by serving as an alternative substrate for energy provision, and by modulating inflammation, oxidative stress, catabolic processes, and gene expression. Of particular relevance to athletes are the metabolic actions of ketone bodies to alter substrate utilisation through attenuating glucose utilisation in peripheral tissues, anti-lipolytic effects on adipose tissue, and attenuation of proteolysis in skeletal muscle. There has been long-standing interest in the development of ingestible forms of ketone bodies that has recently resulted in the commercial availability of exogenous ketone supplements (EKS). These supplements in the form of ketone salts and ketone esters, in addition to ketogenic compounds such as 1,3-butanediol and medium chain triglycerides, facilitate an acute transient increase in circulating AcAc and βHB concentrations, which has been termed 'acute nutritional ketosis' or 'intermittent exogenous ketosis'. Some studies have suggested beneficial effects of EKS to endurance performance, recovery, and overreaching, although many studies have failed to observe benefits of acute nutritional ketosis on performance or recovery. The present review explores the rationale and historical development of EKS, the mechanistic basis for their proposed effects, both positive and negative, and evidence to date for their effects on exercise performance and recovery outcomes before concluding with a discussion of methodological considerations and future directions in this field.
Collapse
|
7
|
Kolb H, Kempf K, Röhling M, Lenzen-Schulte M, Schloot NC, Martin S. Ketone bodies: from enemy to friend and guardian angel. BMC Med 2021; 19:313. [PMID: 34879839 PMCID: PMC8656040 DOI: 10.1186/s12916-021-02185-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
During starvation, fasting, or a diet containing little digestible carbohydrates, the circulating insulin levels are decreased. This promotes lipolysis, and the breakdown of fat becomes the major source of energy. The hepatic energy metabolism is regulated so that under these circumstances, ketone bodies are generated from β-oxidation of fatty acids and secreted as ancillary fuel, in addition to gluconeogenesis. Increased plasma levels of ketone bodies thus indicate a dietary shortage of carbohydrates. Ketone bodies not only serve as fuel but also promote resistance to oxidative and inflammatory stress, and there is a decrease in anabolic insulin-dependent energy expenditure. It has been suggested that the beneficial non-metabolic actions of ketone bodies on organ functions are mediated by them acting as a ligand to specific cellular targets. We propose here a major role of a different pathway initiated by the induction of oxidative stress in the mitochondria during increased ketolysis. Oxidative stress induced by ketone body metabolism is beneficial in the long term because it initiates an adaptive (hormetic) response characterized by the activation of the master regulators of cell-protective mechanism, nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, and AMP-activated kinase. This results in resolving oxidative stress, by the upregulation of anti-oxidative and anti-inflammatory activities, improved mitochondrial function and growth, DNA repair, and autophagy. In the heart, the adaptive response to enhanced ketolysis improves resistance to damage after ischemic insults or to cardiotoxic actions of doxorubicin. Sodium-dependent glucose co-transporter 2 (SGLT2) inhibitors may also exert their cardioprotective action via increasing ketone body levels and ketolysis. We conclude that the increased synthesis and use of ketone bodies as ancillary fuel during periods of deficient food supply and low insulin levels causes oxidative stress in the mitochondria and that the latter initiates a protective (hormetic) response which allows cells to cope with increased oxidative stress and lower energy availability. KEYWORDS: Ketogenic diet, Ketone bodies, Beta hydroxybutyrate, Insulin, Obesity, Type 2 diabetes, Inflammation, Oxidative stress, Cardiovascular disease, SGLT2, Hormesis.
Collapse
Affiliation(s)
- Hubert Kolb
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.,West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| | - Kerstin Kempf
- West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany.
| | - Martin Röhling
- West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| | | | - Nanette C Schloot
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Stephan Martin
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.,West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| |
Collapse
|
8
|
Daines SA. The Therapeutic Potential and Limitations of Ketones in Traumatic Brain Injury. Front Neurol 2021; 12:723148. [PMID: 34777197 PMCID: PMC8579274 DOI: 10.3389/fneur.2021.723148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) represents a significant health crisis. To date, no FDA approved pharmacotherapies are available to prevent the neurological deficits caused by TBI. As an alternative to pharmacotherapy treatment of TBI, ketones could be used as a metabolically based therapeutic strategy. Ketones can help combat post-traumatic cerebral energy deficits while also reducing inflammation, oxidative stress, and neurodegeneration. Experimental models of TBI suggest that administering ketones to TBI patients may provide significant benefits to improve recovery. However, studies evaluating the effectiveness of ketones in human TBI are limited. Unanswered questions remain about age- and sex-dependent factors, the optimal timing and duration of ketone supplementation, and the optimal levels of circulating and cerebral ketones. Further research and improvements in metabolic monitoring technology are also needed to determine if ketone supplementation can improve TBI recovery outcomes in humans.
Collapse
Affiliation(s)
- Savannah Anne Daines
- Department of Biology, Utah State University, Logan, UT, United States
- Department of Kinesiology and Health Science, Utah State University, Logan, UT, United States
| |
Collapse
|
9
|
Sridharan B, Lee MJ. Ketogenic diet: A promising neuroprotective composition for managing Alzheimer's diseases and its pathological mechanisms. Curr Mol Med 2021; 22:640-656. [PMID: 34607541 DOI: 10.2174/1566524021666211004104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/22/2022]
Abstract
Ketogenic diet and ketone bodies gained significant attention in recent years due to their ability to influence the specific energy metabolism and restoration of mitochondrial homeostasis that can help in hindering the progression of many metabolic diseases including diabetes and neurodegenerative diseases. Ketogenic diet consists of high fat and low carbohydrate contents which makes the body glucose deprived and rely on alternative sources (ketone bodies) for energy. It has been initially designed and supplemented for the treatment of epilepsy and later its influence on many energy-deriving biochemical pathways made it a highly sorted food supplement for many metabolic diseases and even by healthy individuals for body building and calorie restriction. Among the reported therapeutic action over a range of diseases, neurodegenerative disorders especially Alzheimer's disease gained the attention of many researchers and clinicians because of its potency and its easier supplementation as a food additive. Complex pathology and multiple influencing factors of Alzheimer's disease make exploration of its therapeutic strategies a demanding task. It was a common phenomenon that energy deprivation in neurological disorders including Alzheimer's disease, to progress rapidly. The ability of ketone bodies to stabilize the mitochondrial energy metabolism makes it a suitable intervening agent. In this review, we will discuss various research progress made with regards to ketone bodies/ketogenic diet for management of Alzheimer's disease and elaborate in detail about the mechanisms that are influenced during their therapeutic action.
Collapse
Affiliation(s)
- Badrinathan Sridharan
- Department of Applied Chemistry, Chaoyang University of Technology, 168 Jifeng East Road, Taichung. Taiwan
| | - Meng-Jen Lee
- Department of Applied Chemistry, Chaoyang University of Technology, 168 Jifeng East Road, Taichung. Taiwan
| |
Collapse
|
10
|
Yao A, Li Z, Lyu J, Yu L, Wei S, Xue L, Wang H, Chen GQ. On the nutritional and therapeutic effects of ketone body D-β-hydroxybutyrate. Appl Microbiol Biotechnol 2021; 105:6229-6243. [PMID: 34415393 PMCID: PMC8377336 DOI: 10.1007/s00253-021-11482-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022]
Abstract
Abstract d-β-hydroxybutyrate (d-3HB), a monomer of microbial polyhydroxybutyrate (PHB), is also a natural ketone body produced during carbohydrate deprivation to provide energy to the body cells, heart, and brain. In recent years, increasing evidence demonstrates that d-3HB can induce pleiotropic effects on the human body which are highly beneficial for improving physical and metabolic health. Conventional ketogenic diet (KD) or exogenous ketone salts (KS) and esters (KE) have been used to increase serum d-3HB level. However, strict adaptation to the KD was often associated with poor patient compliance, while the ingestion of KS caused gastrointestinal distresses due to excessive consumption of minerals. As for ingestion of KE, subsequent degradation is required before releasing d-3HB for absorption, making these methods somewhat inferior. This review provides novel insights into a biologically synthesized d-3HB (d-3-hydroxybutyric acid) which can induce a faster increase in plasma d-3HB compared to the use of KD, KS, or KE. It also emphasizes on the most recent applications of d-3HB in different fields, including its use in improving exercise performance and in treating metabolic or age-related diseases. Ketones may become a fourth micro-nutrient that is necessary to the human body along with carbohydrates, proteins, and fats. Indeed, d-3HB being a small molecule with multiple signaling pathways within the body exhibits paramount importance in mitigating metabolic and age-related diseases. Nevertheless, specific dose–response relationships and safety margins of using d-3HB remain to be elucidated with more research. Key points • d-3HB induces pleiotropic effects on physical and metabolic health. • Exogenous ketone supplements are more effective than ketogenic diet. • d-3HB as a ketone supplement has long-term healthy impact.
Collapse
Affiliation(s)
- Aliya Yao
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China
| | - Zihua Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Jinyan Lyu
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China
| | - Liusong Yu
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China
| | - Situ Wei
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China
| | - Lingyun Xue
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China
| | - Hui Wang
- Department of Colorectal Surgery, Guangdong Province Biomedical Material Conversion and Evaluation Engineering Technology Center, Institute of Biomedical Innovation, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, Guangdong Province, China
| | - Guo-Qiang Chen
- MedPHA Bioscience Co. Ltd., Traditional Chinese Medicine Science and Technology Industrial Park of Co-Operation Between Guangdong and Macau, Building No.103, 36 Doukou Rd, Hengqin District, Zhuhai, 519030, Guangdong Province, China. .,School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China. .,School of Life Sciences and Dept Chemical Engineering, Center for Synthetic and Systems Biology (CSSB), Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
11
|
Fang Y, Chen B, Gong AY, Malhotra D, Gupta R, Dworkin LD, Gong R. The ketone body β-hydroxybutyrate mitigates the senescence response of glomerular podocytes to diabetic insults. Kidney Int 2021; 100:1037-1053. [PMID: 34246657 DOI: 10.1016/j.kint.2021.06.031] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 01/23/2023]
Abstract
Diabetic kidney disease (DKD) is one of the most common complications of diabetes and clinically featured by progressive albuminuria, consequent to glomerular destruction that involves podocyte senescence. Burgeoning evidence suggests that ketosis, in particular β-hydroxybutyrate, exerts a beneficial effect on aging and on myriad metabolic or chronic diseases, including obesity, diabetes and chronic kidney diseases. Its effect on DKD is largely unknown. In vitro in podocytes exposed to a diabetic milieu, β-hydroxybutyrate treatment substantially mitigated cellular senescence and injury, as evidenced by reduced formation of γH2AX foci, reduced staining for senescence-associated-β-galactosidase activity, diminished expression of key mediators of senescence signaling like p16INK4A and p21, and preserved expression of synaptopodin. This beneficial action of β-hydroxybutyrate coincided with a reinforced transcription factor Nrf2 antioxidant response. Mechanistically, β-hydroxybutyrate inhibition of glycogen synthase kinase 3β (GSK3β), a convergent point for myriad signaling pathways regulating Nrf2 activity, seems to contribute. Indeed, trigonelline, a selective inhibitor of Nrf2, or ectopic expression of constitutively active mutant GSK3β abolished, whereas selective activation of Nrf2 was sufficient for the anti-senescent and podocyte protective effects of β-hydroxybutyrate. Moreover, molecular modeling and docking analysis revealed that β-hydroxybutyrate is able to directly target the ATP-binding pocket of GSK3β and thereby block its kinase activity. In murine models of streptozotocin-elicited DKD, β-hydroxybutyrate therapy inhibited GSK3β and reinforced Nrf2 activation in glomerular podocytes, resulting in lessened podocyte senescence and injury and improved diabetic glomerulopathy and albuminuria. Thus, our findings may pave the way for developing a β-hydroxybutyrate-based novel approach of therapeutic ketosis for treating DKD.
Collapse
Affiliation(s)
- Yudong Fang
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
| | - Bohan Chen
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio;; Division of Kidney Disease and Hypertension, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island
| | - Athena Y Gong
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
| | - Deepak Malhotra
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
| | - Rajesh Gupta
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio
| | - Lance D Dworkin
- Division of Kidney Disease and Hypertension, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island;; Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio
| | - Rujun Gong
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio;; Division of Kidney Disease and Hypertension, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island;; Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio; Deaprtment of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio.
| |
Collapse
|
12
|
Chen O, Blonquist TM, Mah E, Sanoshy K, Beckman D, Nieman KM, Winters BL, Anthony JC, Verdin E, Newman JC, Stubbs BJ. Tolerability and Safety of a Novel Ketogenic Ester, Bis-Hexanoyl (R)-1,3-Butanediol: A Randomized Controlled Trial in Healthy Adults. Nutrients 2021; 13:2066. [PMID: 34208742 PMCID: PMC8234448 DOI: 10.3390/nu13062066] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Nutritional ketosis is a state of mildly elevated blood ketone concentrations resulting from dietary changes (e.g., fasting or reduced carbohydrate intake) or exogenous ketone consumption. In this study, we determined the tolerability and safety of a novel exogenous ketone diester, bis-hexanoyl-(R)-1,3-butanediol (BH-BD), in a 28-day, randomized, double-blind, placebo-controlled, parallel trial (NCT04707989). Healthy adults (n = 59, mean (SD), age: 42.8 (13.4) y, body mass index: 27.8 (3.9) kg/m2) were randomized to consume a beverage containing 12.5 g (Days 0-7) and 25 g (Days 7-28) of BH-BD or a taste-matched placebo daily with breakfast. Tolerability, stimulation, and sedation were assessed daily by standardized questionnaires, and blood and urine samples were collected at Days 0, 7, 14, and 28 for safety assessment. There were no differences in at-home composite systemic and gastrointestinal tolerability scores between BH-BD and placebo at any time in the study, or in acute tolerability measured 1-h post-consumption in-clinic. Weekly at-home composite tolerability scores did not change when BH-BD servings were doubled. At-home scores for stimulation and sedation did not differ between groups. BH-BD significantly increased blood ketone concentrations 1-h post-consumption. No clinically meaningful changes in safety measures including vital signs and clinical laboratory measurements were detected within or between groups. These results support the overall tolerability and safety of consumption of up to 25 g/day BH-BD.
Collapse
Affiliation(s)
- Oliver Chen
- Biofortis Research, Addison, IL 60101, USA; (O.C.); (T.M.B.); (E.M.); (K.S.); (D.B.)
| | - Traci M. Blonquist
- Biofortis Research, Addison, IL 60101, USA; (O.C.); (T.M.B.); (E.M.); (K.S.); (D.B.)
| | - Eunice Mah
- Biofortis Research, Addison, IL 60101, USA; (O.C.); (T.M.B.); (E.M.); (K.S.); (D.B.)
| | - Kristen Sanoshy
- Biofortis Research, Addison, IL 60101, USA; (O.C.); (T.M.B.); (E.M.); (K.S.); (D.B.)
| | - Dawn Beckman
- Biofortis Research, Addison, IL 60101, USA; (O.C.); (T.M.B.); (E.M.); (K.S.); (D.B.)
| | | | | | - Joshua C. Anthony
- Nlumn LLC, Princeton, NJ 08543, USA; or
- Juvenescence Ltd., Princeton, NJ 08540, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA; (E.V.); (J.C.N.)
| | - John C. Newman
- Buck Institute for Research on Aging, Novato, CA 94945, USA; (E.V.); (J.C.N.)
- Division of Geriatrics, UCSF, San Francisco, CA 94143, USA
| | - Brianna J. Stubbs
- Buck Institute for Research on Aging, Novato, CA 94945, USA; (E.V.); (J.C.N.)
| |
Collapse
|
13
|
The Chemistry of the Ketogenic Diet: Updates and Opportunities in Organic Synthesis. Int J Mol Sci 2021; 22:ijms22105230. [PMID: 34063366 PMCID: PMC8157195 DOI: 10.3390/ijms22105230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023] Open
Abstract
The high-fat, low-carbohydrate (ketogenic) diet has grown in popularity in the last decade as a weight loss tool. Research into the diet’s effects on the body have revealed a variety of other health benefits. The use of exogenous ketone supplements to confer the benefits of the diet without strict adherence to it represents an exciting new area of focus. Synthetic ketogenic compounds are of particular interest that has received very little emphasis and is an untapped area of focus for chemical synthesis. In this review, we summarize the chemical basis for ketogenicity and opportunities for further advancement of the field.
Collapse
|
14
|
Takahara S, Soni S, Maayah ZH, Ferdaoussi M, Dyck JRB. Ketone Therapy for Heart Failure: Current Evidence for Clinical Use. Cardiovasc Res 2021; 118:977-987. [PMID: 33705533 DOI: 10.1093/cvr/cvab068] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/09/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
During conditions that result in depleted circulating glucose levels, ketone bodies synthesized in the liver are necessary fuel substrates for the brain. In other organs such as the heart, the reliance on ketones for generating energy is less life threatening as the heart can utilize alternative fuel sources such as fatty acids. However, during pathophysiological conditions such as heart failure, cardiac defects in metabolic processes that normally allow for sufficient energy production from fatty acids and carbohydrates contribute to a decline in contractile function. As such, it has been proposed that the failing heart relies more on ketone bodies as an energy source than previously appreciated. Furthermore, it has been suggested that ketone bodies may function as signaling molecules that can suppress systemic and cardiac inflammation. Thus, it is possible that intentionally elevating circulating ketones may be beneficial as an adjunct treatment for heart failure. Although many approaches can be used for 'ketone therapy', each of these has their own advantages and disadvantages in the treatment of heart failure. Thus, we summarize current preclinical and clinical studies involving various types of ketone therapy in cardiac disease and discuss the advantages and disadvantages of each modality as possible treatments for heart failure.
Collapse
Affiliation(s)
- Shingo Takahara
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shubham Soni
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zaid H Maayah
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mourad Ferdaoussi
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Karwi QG, Lopaschuk GD. CrossTalk proposal: Ketone bodies are an important metabolic fuel for the heart. J Physiol 2021; 600:1001-1004. [PMID: 33645632 DOI: 10.1113/jp281004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada.,Department of Pharmacology, College of Medicine, University of Diyala, Diyala, Iraq
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
16
|
Golonka RM, Xiao X, Abokor AA, Joe B, Vijay-Kumar M. Altered nutrient status reprograms host inflammation and metabolic health via gut microbiota. J Nutr Biochem 2020; 80:108360. [PMID: 32163821 PMCID: PMC7242157 DOI: 10.1016/j.jnutbio.2020.108360] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/07/2023]
Abstract
The metabolism of macro- and micronutrients is a complex and highly regulated biological process. An imbalance in the metabolites and their signaling networks can lead to nonresolving inflammation and consequently to the development of chronic inflammatory-associated diseases. Therefore, identifying the accumulated metabolites and altered pathways during inflammatory disorders would not only serve as "real-time" markers but also help in the development of nutritional therapeutics. In this review, we explore recent research that has delved into elucidating the effects of carbohydrate/calorie restriction, protein malnutrition, lipid emulsions and micronutrient deficiencies on metabolic health and inflammation. Moreover, we describe the integrated stress response in terms of amino acid starvation and lipemia and how this modulates new age diseases such as inflammatory bowel disease and atherosclerosis. Lastly, we explain the latest research on metaflammation and inflammaging. This review focuses on multiple signaling pathways, including, but not limited to, the FGF21-β-hydroxybutryate-NLRP3 axis, the GCN2-eIF2α-ATF4 pathway, the von Hippel-Lindau/hypoxia-inducible transcription factor pathway and the TMAO-PERK-FoxO1 axis. Additionally, throughout the review, we explain how the gut microbiota responds to altered nutrient status and also how antimicrobial peptides generated from nutrient-based signaling pathways can modulate the gut microbiota. Collectively, it must be emphasized that metabolic starvation and inflammation are strongly regulated by both environmental (i.e., nutrition, gut microbiome) and nonenvironmental (i.e., genetics) factors, which can influence the susceptibility to inflammatory disorders.
Collapse
Affiliation(s)
- Rachel M Golonka
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Xia Xiao
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ahmed A Abokor
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Bina Joe
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614.
| |
Collapse
|
17
|
In vitro human colonic microbiota utilises D-β-hydroxybutyrate to increase butyrogenesis. Sci Rep 2020; 10:8516. [PMID: 32444846 PMCID: PMC7244492 DOI: 10.1038/s41598-020-65561-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/05/2020] [Indexed: 12/31/2022] Open
Abstract
The ketone body D-β-hydroxybutyrate (DBHB) has gained attention owing to its cellular signalling function; however, its effect on the human colonic microbiota remains unclear. Here, DBHB dynamics in the human colon were investigated using an in vitro colonic microbiota model, which maintained most of the operational taxonomic units detected in the original faeces. Over 54% of 0.41% (w/v) DBHB was metabolised by microbiota models originating from seven faecal samples after 30 h of fermentation (regarded as DBHB utilisers); however, <19% of DBHB was metabolised by microbiota models from five faecal samples (regarded as non-utilisers of DBHB). In utilisers, DBHB administration increased the relative abundance of the genus Coprococcus, correlated with increased butyrogenesis. Increased butyrogenesis was not observed in DBHB non-utilisers. Based on PICRUSt analysis, the relative abundance of β-hydroxybutyrate dehydrogenase was maintained in microbiota models from DBHB utilisers following DBHB administration; however, it decreased in microbiota models from non-utilisers. After 21 h of fermentation, the intracellular glutamate concentration, which is indicative of growth, showed a positive correlation with DBHB utilisation (R2 = 0.70). Human colonic microbiotas with high growth activity demonstrate efficient utilisation of DBHB for increased butyrate production, which affords health benefits.
Collapse
|
18
|
Cuenoud B, Hartweg M, Godin JP, Croteau E, Maltais M, Castellano CA, Carpentier AC, Cunnane SC. Metabolism of Exogenous D-Beta-Hydroxybutyrate, an Energy Substrate Avidly Consumed by the Heart and Kidney. Front Nutr 2020; 7:13. [PMID: 32140471 PMCID: PMC7042179 DOI: 10.3389/fnut.2020.00013] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in the metabolism of ketones owing to their reported benefits in neurological and more recently in cardiovascular and renal diseases. As an alternative to a very high fat ketogenic diet, ketones precursors for oral intake are being developed to achieve ketosis without the need for dietary carbohydrate restriction. Here we report that an oral D-beta-hydroxybutyrate (D-BHB) supplement is rapidly absorbed and metabolized in humans and increases blood ketones to millimolar levels. At the same dose, D-BHB is significantly more ketogenic and provides fewer calories than a racemic mixture of BHB or medium chain triglyceride. In a whole body ketone positron emission tomography pilot study, we observed that after D-BHB consumption, the ketone tracer 11C-acetoacetate is rapidly metabolized, mostly by the heart and the kidneys. Beyond brain energy rescue, this opens additional opportunities for therapeutic exploration of D-BHB supplements as a "super fuel" in cardiac and chronic kidney diseases.
Collapse
Affiliation(s)
- Bernard Cuenoud
- Nestlé Health Science, Translation Research, Epalinges, Switzerland
| | - Mickaël Hartweg
- Nestlé Research, Clinical Development Unit, Lausanne, Switzerland
| | - Jean-Philippe Godin
- Nestlé Research, Institute of Food Safety and Analytical Sciences, Lausanne, Switzerland
| | | | - Mathieu Maltais
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,Research Center on Aging, Sherbrooke, QC, Canada
| | | | - André C Carpentier
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,CHUS Research Center, Sherbrooke, QC, Canada.,Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stephen C Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,Research Center on Aging, Sherbrooke, QC, Canada.,Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
19
|
Bhattacharya K, Matar W, Tolun AA, Devanapalli B, Thompson S, Dalkeith T, Lichkus K, Tchan M. The use of sodium DL-3-Hydroxybutyrate in severe acute neuro-metabolic compromise in patients with inherited ketone body synthetic disorders. Orphanet J Rare Dis 2020; 15:53. [PMID: 32070364 PMCID: PMC7029565 DOI: 10.1186/s13023-020-1316-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background Ketone bodies form a vital energy source for end organs in a variety of physiological circumstances. At different times, the heart, brain and skeletal muscle in particular can use ketones as a primary substrate. Failure to generate ketones in such circumstances leads to compromised energy delivery, critical end-organ dysfunction and potentially death. There are a range of inborn errors of metabolism (IEM) affecting ketone body production that can present in this way, including disorders of carnitine transport into the mitochondrion, mitochondrial fatty acid oxidation deficiencies (MFAOD) and ketone body synthesis. In situations of acute energy deficit, management of IEM typically entails circumventing the enzyme deficiency with replenishment of energy requirements. Due to profound multi-organ failure it is often difficult to provide optimal enteral therapy in such situations and rescue with sodium DL-3-hydroxybutyrate (S DL-3-OHB) has been attempted in these conditions as documented in this paper. Results We present 3 cases of metabolic decompensation, one with carnitine-acyl-carnitine translocase deficiency (CACTD) another with 3-hydroxyl, 3-methyl, glutaryl CoA lyase deficiency (HMGCLD) and a third with carnitine palmitoyl transferase II deficiency (CPT2D). All of these disorders are frequently associated with death in circumstance where catastrophic acute metabolic deterioration occurs. Intensive therapy with adjunctive S DL-3OHB led to rapid and sustained recovery in all. Alternative therapies are scarce in these situations. Conclusion S DL-3-OHB has been utilised in multiple acyl co A dehydrogenase deficiency (MADD) in cases with acute neurological and cardiac compromise with long-term data awaiting publication. The use of S DL-3-OHB is novel in non-MADD fat oxidation disorders and contribute to the argument for more widespread use.
Collapse
Affiliation(s)
- Kaustuv Bhattacharya
- Disciplines of Genetic Medicine and Child and Adolescent Health, University of Sydney, Sydney, Australia. .,Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia.
| | - Walid Matar
- Department of Neurology, St George Hospital, Kogarah, NSW, Australia
| | | | | | - Sue Thompson
- Disciplines of Genetic Medicine and Child and Adolescent Health, University of Sydney, Sydney, Australia.,Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
| | - Troy Dalkeith
- Disciplines of Genetic Medicine and Child and Adolescent Health, University of Sydney, Sydney, Australia.,Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
| | - Kate Lichkus
- Disciplines of Genetic Medicine and Child and Adolescent Health, University of Sydney, Sydney, Australia.,Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
| | - Michel Tchan
- Disciplines of Genetic Medicine and Child and Adolescent Health, University of Sydney, Sydney, Australia.,Westmead Hospital, University of Sydney, Westmead, Australia
| |
Collapse
|
20
|
Kapogiannis D, Avgerinos KI. Brain glucose and ketone utilization in brain aging and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:79-110. [PMID: 32739015 PMCID: PMC9989941 DOI: 10.1016/bs.irn.2020.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To meet its high energy demands, the brain mostly utilizes glucose. However, the brain has evolved to exploit additional fuels, such as ketones, especially during prolonged fasting. With aging and neurodegenerative diseases (NDDs), the brain becomes inefficient at utilizing glucose due to changes in glia and neurons that involve glucose transport, glycolytic and Krebs cycle enzyme activities, and insulin signaling. Positron emission tomography and magnetic resonance spectroscopy studies have identified glucose metabolism abnormalities in aging, Alzheimer's disease (AD) and other NDDs in vivo. Despite glucose hypometabolism, brain cells can utilize ketones efficiently, thereby providing a rationale for the development of therapeutic ketogenic interventions in AD and other NDDs. This review compares available ketogenic interventions and discusses the potential of the potent oral Ketone Ester for future therapeutic use in AD and other NDDs characterized by inefficient glucose utilization.
Collapse
Affiliation(s)
- Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.
| | - Konstantinos I Avgerinos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
21
|
Poff AM, Rho JM, D'Agostino DP. Ketone Administration for Seizure Disorders: History and Rationale for Ketone Esters and Metabolic Alternatives. Front Neurosci 2019; 13:1041. [PMID: 31680801 PMCID: PMC6803688 DOI: 10.3389/fnins.2019.01041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 09/13/2019] [Indexed: 12/31/2022] Open
Abstract
The ketogenic diet (KD) is a high-fat, low-carbohydrate treatment for medically intractable epilepsy. One of the hallmark features of the KD is the production of ketone bodies which have long been believed, but not yet proven, to exert direct anti-seizure effects. The prevailing view has been that ketosis is an epiphenomenon during KD treatment, mostly due to clinical observations that blood ketone levels do not correlate well with seizure control. Nevertheless, there is increasing experimental evidence that ketone bodies alone can exert anti-seizure properties through a multiplicity of mechanisms, including but not limited to: (1) activation of inhibitory adenosine and ATP-sensitive potassium channels; (2) enhancement of mitochondrial function and reduction in oxidative stress; (3) attenuation of excitatory neurotransmission; and (4) enhancement of central γ-aminobutyric acid (GABA) synthesis. Other novel actions more recently reported include inhibition of inflammasome assembly and activation of peripheral immune cells, and epigenetic effects by decreasing the activity of histone deacetylases (HDACs). Collectively, the preclinical evidence to date suggests that ketone administration alone might afford anti-seizure benefits for patients with epilepsy. There are, however, pragmatic challenges in administering ketone bodies in humans, but prior concerns may largely be mitigated through the use of ketone esters or balanced ketone electrolyte formulations that can be given orally and induce elevated and sustained hyperketonemia to achieve therapeutic effects.
Collapse
Affiliation(s)
- Angela M Poff
- Laboratory of Metabolic Medicine, Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jong M Rho
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Division of Pediatric Neurology, Rady Children's Hospital-San Diego, University of California, San Diego, San Diego, CA, United States
| | - Dominic P D'Agostino
- Laboratory of Metabolic Medicine, Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Ocala, FL, United States
| |
Collapse
|
22
|
Fischer T, Elpers C, Och U, Fobker M, Marquardt T. Ketone body therapy with D/L-β-hydroxybutyric acid solution in severe MADD. Mol Genet Metab Rep 2019; 20:100491. [PMID: 31312603 PMCID: PMC6610240 DOI: 10.1016/j.ymgmr.2019.100491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/31/2022] Open
Abstract
Objectives Multiple acyl-CoA dehydrogenase deficiency (MADD) is a severe inborn disorder of mitochondrial fatty acid oxidation. The only treatment option for MADD is the use of exogenous ketone bodies, like sodium β-hydroxybutyrate (NaβHB). However, the use of ketone body salts leads to a high intake of accompanying minerals, which can lead to additional side effects. The use of mineral-free formulations could improve tolerability. Methods In this report, the use of a βHB acid (βHBA) in a patient with MADD is described. The production of D/L-βHBA was carried out using ion exchange chromatography (IEX) and using a precipitation method. During two inpatient treatment intervals, the tolerability as well as clinical and metabolic effects were monitored. D-βHB in serum, blood gas analysis, and standard blood measurements (like minerals) were used as control parameters. Results Production of D/L-βHBA using the precipitation method was more effective than using IEX. The tube feed solution used had a minimum pH of 3.5. Capillary D-βHB measurements were between 0.1 and 0.4 mmol/L and venous were at 0.1 mmol/L or below. Minerals and serum pH were within the normal range. During application of D/L-βHBA, gastrointestinal discomfort occurred and no clinical improvement was observed. Conclusions The use of D/L-βHBA in the therapy of severe MADD could be a good addition to the use of classical ketone body salts. The observed gastrointestinal side effects were of a mild nature and could not be specifically attributed to the D/L-βHBA treatment. In short-term application, no clinical benefit and no substantial increase of D-βHB in serum were noted. No tendency towards acidosis or alkalosis was observed during the entire period of treatment.
Collapse
Affiliation(s)
- Tobias Fischer
- University Hospital Muenster, Department of Pediatrics, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Christiane Elpers
- University Hospital Muenster, Department of Pediatrics, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Ulrike Och
- University Hospital Muenster, Department of Pediatrics, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Manfred Fobker
- University Hospital Muenster, Center of laboratory medicine, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Thorsten Marquardt
- University Hospital Muenster, Department of Pediatrics, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| |
Collapse
|
23
|
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
|
24
|
Da Costa D, Bangalee V, Subban K, Naidoo R. Ketone body supplement label claims: what supplement has been supplemented? SOUTH AFRICAN JOURNAL OF SPORTS MEDICINE 2019; 31:v31i1a6369. [PMID: 36818000 PMCID: PMC9924521 DOI: 10.17159/2078-516x/2019/v31i1a6369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background There is a keen interest in performance-enhancing supplementation and the associated benefits, despite reports of incorrect label claims made by manufacturers and the questionable efficacy of the supplements. The use of ketone body supplements as a source of fuel during exercise and sporting performance, in particular, is of interest to sportspeople. By increasing blood ketone body levels, with an accompanying decrease in blood glucose, may indicate a state of nutritional ketosis, whereby the body no longer relies on glucose metabolism but rather the metabolism of ketone bodies. This could be beneficial for long, slow steady-state endurance exercise. Discussion There are numerous ketone body supplements on the market manufactured in South Africa and internationally. However, unlike medicines, the sports supplementation industry is poorly regulated. Furthermore, ketone body supplementation with regard to its effects on improving exercise and athletic performance is still unconvincing. Conclusion Within the ever-changing sports supplementation industry, ketone body supplements are being used despite controversies regarding the accuracy and scientific merit of label claims. The ingredients and their quantities, as well as the performance benefits, need to be objectively validated.
Collapse
Affiliation(s)
- D Da Costa
- Discipline of Biokinetics, Exercise and Leisure Sciences, University of KwaZulu-Natal, College of Health Sciences,
South Africa
| | - V Bangalee
- Discipline of Biokinetics, Exercise and Leisure Sciences, University of KwaZulu-Natal, College of Health Sciences,
South Africa
| | - K Subban
- Prime Human Performance Institute, 44 Isaiah Ntshangase Road, Durban,
South Africa
| | - R Naidoo
- Discipline of Biokinetics, Exercise and Leisure Sciences, University of KwaZulu-Natal, College of Health Sciences,
South Africa
| |
Collapse
|