1
|
Volek JS, Kackley ML, Buga A. Nutritional Considerations During Major Weight Loss Therapy: Focus on Optimal Protein and a Low-Carbohydrate Dietary Pattern. Curr Nutr Rep 2024; 13:422-443. [PMID: 38814519 PMCID: PMC11327213 DOI: 10.1007/s13668-024-00548-6] [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] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Considering the high prevalence of obesity and related metabolic impairments in the population, the unique role nutrition has in weight loss, reversing metabolic disorders, and maintaining health cannot be overstated. Normal weight and well-being are compatible with varying dietary patterns, but for the last half century there has been a strong emphasis on low-fat, low-saturated fat, high-carbohydrate based approaches. Whereas low-fat dietary patterns can be effective for a subset of individuals, we now have a population where the vast majority of adults have excess adiposity and some degree of metabolic impairment. We are also entering a new era with greater access to bariatric surgery and approval of anti-obesity medications (glucagon-like peptide-1 analogues) that produce substantial weight loss for many people, but there are concerns about disproportionate loss of lean mass and nutritional deficiencies. RECENT FINDINGS No matter the approach used to achieve major weight loss, careful attention to nutritional considerations is necessary. Here, we examine the recent findings regarding the importance of adequate protein to maintain lean mass, the rationale and evidence supporting low-carbohydrate and ketogenic dietary patterns, and the potential benefits of including exercise training in the context of major weight loss. While losing and sustaining weight loss has proven challenging, we are optimistic that application of emerging nutrition science, particularly personalized well-formulated low-carbohydrate dietary patterns that contain adequate protein (1.2 to 2.0 g per kilogram reference weight) and achieve the beneficial metabolic state of euketonemia (circulating ketones 0.5 to 5 mM), is a promising path for many individuals with excess adiposity.
Collapse
Affiliation(s)
- Jeff S Volek
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA.
| | - Madison L Kackley
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| | - Alex Buga
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| |
Collapse
|
2
|
Shahtaghi NR, Bigdelitabar S, Thakur S, Kaur M, Singh H, Saini M, Singh M, Singh K, Kumar Jain S. Oral beta-hydroxybutyrate alleviates COVID-19 related acute respiratory distress syndrome: A randomized, single-blind, placebo-controlled trial. Res Social Adm Pharm 2024; 20:760-767. [PMID: 38734512 DOI: 10.1016/j.sapharm.2024.04.015] [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: 12/15/2023] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a lung complication of COVID-19 that requires intensive care and ventilation. Beta-hydroxybutyrate (BHB) is a ketone body that can modulate metabolism and inflammation in immune cells and lung tissues. We hypothesized that oral BHB could alleviate COVID-19 related ARDS by reducing pro-inflammatory cytokines and increasing anti-inflammatory cytokines. METHODS We randomized 75 patients with mild (as per Berlin criteria) ARDS symptoms to receive oral 25 g twice daily or placebo for five days. The primary outcome was the change in pro-inflammatory cytokines (Interleukin-1β, Interleukin-6, interleukin-18, tumour necrosis factor-alpha) and anti-inflammatory cytokine (interleukin-10) from baseline to day 5. The secondary outcomes were the change in BHB levels from baseline to day 5, the number of hospitalization days, and the occurrence of adverse events. RESULTS Treatment with formulated BHB resulted in a significant decrease in pro-inflammatory cytokines; Interleukin-1β (p = 0.0204), Interleukin-6 (p = 0.0309), interleukin-18 (p = 0.0116), tumour necrosis factor-alpha (p = 0.0489) and increase in interleukin-10 (p = 0.0246) compared treatment with placebo. Importantly, higher BHB levels (p = 0.0001) were observed after supplementation; additionally, patients who underwent this approach were hospitalized for fewer days. No serious adverse events were reported. CONCLUSION Beta-hydroxybutyrate, an oral adjunct therapy, has shown promising results in ameliorating symptoms of ARDS. This includes reduced inflammation, oxidative stress, and decreased patient fatigue levels. Further study with a large sample size is warranted to assess the potential of BHB therapy's effectiveness in reducing the development of severe illness. CLINICAL TRIAL REGISTRATION (http://ctri.nic.in/CTRI/2021/03/031790).
Collapse
Affiliation(s)
- Navid Reza Shahtaghi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| | - Samira Bigdelitabar
- Department of Microbiology, Government Medical College, Amritsar, 143005, Punjab, India.
| | - Subham Thakur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| | - Manjot Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| | - Harjeet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| | - Muskaan Saini
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| | - Manjinder Singh
- Department of Pharmacy, Government Polytechnic College, Amritsar, 143105, Punjab, India.
| | - Kanwardeep Singh
- Department of Microbiology, Government Medical College, Amritsar, 143005, Punjab, India.
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India; Centre for Basic Translational Research in Health Sciences (CBTHRS), Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
3
|
Ma Q, Jiang L, You Y, Ni H, Ma L, Lin X, Wang Z, Yan W, Xiao X, Li X, Li J. Ketogenic diet ameliorates high-fat diet-induced insulin resistance in mouse skeletal muscle by alleviating endoplasmic reticulum stress. Biochem Biophys Res Commun 2024; 702:149559. [PMID: 38341923 DOI: 10.1016/j.bbrc.2024.149559] [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: 12/31/2023] [Revised: 01/07/2024] [Accepted: 01/20/2024] [Indexed: 02/13/2024]
Abstract
OBJECTIVE Ketogenic diets (KD) have been shown to alleviate insulin resistance (IR) by exerting anti-lipogenic and insulin sensitizing effects in the liver through a variety of pathways. The present study sought to investigate whether a ketogenic diet also improves insulin sensitization in skeletal muscle cells through alleviating endoplasmic reticulum stress. METHODS High-fat diet-induced IR mice were allowed to a 2-week ketogenic diet. Insulin resistance and glucose tolerance were evaluated through GTT, ITT, and HOMA-IR. The C2C12 myoblasts exposed to palmitic acid were used to evaluate the insulin sensitization effects of β-hydroxybutyric acid (β-OHB). Molecular mechanisms concerning ER stress signaling activation and glucose uptake were assessed. RESULTS The AKT/GSK3β pathway was inhibited, ER stress signaling associated with IRE1, PERK, and BIP was activated, and the number of Glut4 proteins translocated to membrane decreased in the muscle of HFD mice. However, all these changes were reversed after 2 weeks of feeding on a ketogenic diet. Consistently in C2C12 myoblasts, the AKT/GSK3β pathway was inhibited by palmitic acid (PA) treatment. The endoplasmic reticulum stress-related proteins, IRE1, and BIP were increased, and the number of Glut4 proteins on the cell membrane decreased. However, β-OHB treatment alleviated ER stress and improved the glucose uptake of C2C12 cells. CONCLUSION Our data reveal that KD ameliorated HFD-induced insulin resistance in skeletal muscle, which was partially mediated by inhibiting endoplasmic reticulum stress. The insulin sensitization effect of β-OHB is associated with up regulation of AKT/GSK3β pathway and the increase in the number of Glut4 proteins on the cell membrane.
Collapse
Affiliation(s)
- Qin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lincheng Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuehua You
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongbing Ni
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li Ma
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuyun Wang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weiyan Yan
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoqiu Xiao
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Li
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Jibin Li
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
4
|
Williams AS, Crown SB, Lyons SP, Koves TR, Wilson RJ, Johnson JM, Slentz DH, Kelly DP, Grimsrud PA, Zhang GF, Muoio DM. Ketone flux through BDH1 supports metabolic remodeling of skeletal and cardiac muscles in response to intermittent time-restricted feeding. Cell Metab 2024; 36:422-437.e8. [PMID: 38325337 PMCID: PMC10961007 DOI: 10.1016/j.cmet.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Time-restricted feeding (TRF) has gained attention as a dietary regimen that promotes metabolic health. This study questioned if the health benefits of an intermittent TRF (iTRF) schedule require ketone flux specifically in skeletal and cardiac muscles. Notably, we found that the ketolytic enzyme beta-hydroxybutyrate dehydrogenase 1 (BDH1) is uniquely enriched in isolated mitochondria derived from heart and red/oxidative skeletal muscles, which also have high capacity for fatty acid oxidation (FAO). Using mice with BDH1 deficiency in striated muscles, we discover that this enzyme optimizes FAO efficiency and exercise tolerance during acute fasting. Additionally, iTRF leads to robust molecular remodeling of muscle tissues, and muscle BDH1 flux does indeed play an essential role in conferring the full adaptive benefits of this regimen, including increased lean mass, mitochondrial hormesis, and metabolic rerouting of pyruvate. In sum, ketone flux enhances mitochondrial bioenergetics and supports iTRF-induced remodeling of skeletal muscle and heart.
Collapse
Affiliation(s)
- Ashley S Williams
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott P Lyons
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca J Wilson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Jordan M Johnson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Dorothy H Slentz
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Daniel P Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
5
|
Paoli A, Tinsley GM, Mattson MP, De Vivo I, Dhawan R, Moro T. Common and divergent molecular mechanisms of fasting and ketogenic diets. Trends Endocrinol Metab 2024; 35:125-141. [PMID: 38577754 DOI: 10.1016/j.tem.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 04/06/2024]
Abstract
Intermittent short-term fasting (ISTF) and ketogenic diets (KDs) exert overlapping but not identical effects on cell metabolism, function, and resilience. Whereas health benefits of KD are largely mediated by the ketone bodies (KBs), ISTF engages additional adaptive physiological responses. KDs act mainly through inhibition of histone deacetylases (HDACs), reduction of oxidative stress, improvement of mitochondria efficiency, and control of inflammation. Mechanisms of action of ISTF include stimulation of autophagy, increased insulin and leptin sensitivity, activation of AMP-activated protein kinase (AMPK), inhibition of the mechanistic target of rapamycin (mTOR) pathway, bolstering mitochondrial resilience, and suppression of oxidative stress and inflammation. Frequent switching between ketogenic and nonketogenic states may optimize health by increasing stress resistance, while also enhancing cell plasticity and functionality.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy.
| | - Grant M Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ravi Dhawan
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy
| |
Collapse
|
6
|
Paoli A, Cerullo G. Investigating the Link between Ketogenic Diet, NAFLD, Mitochondria, and Oxidative Stress: A Narrative Review. Antioxidants (Basel) 2023; 12:antiox12051065. [PMID: 37237931 DOI: 10.3390/antiox12051065] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Together with the global rise in obesity and metabolic syndrome, the prevalence of individuals who suffer from nonalcoholic fatty liver disease (NAFLD) has risen dramatically. NAFLD is currently the most common chronic liver disease and includes a continuum of liver disorders from initial fat accumulation to nonalcoholic steatohepatitis (NASH), considered the more severe forms, which can evolve in, cirrhosis, and hepatocellular carcinoma. Common features of NAFLD includes altered lipid metabolism mainly linked to mitochondrial dysfunction, which, as a vicious cycle, aggravates oxidative stress and promotes inflammation and, as a consequence, the progressive death of hepatocytes and the severe form of NAFLD. A ketogenic diet (KD), i.e., a diet very low in carbohydrates (<30 g/die) that induces "physiological ketosis", has been demonstrated to alleviate oxidative stress and restore mitochondrial function. Based on this, the aim of the present review is to analyze the body of evidence regarding the potential therapeutic role of KD in NAFLD, focusing on the interplay between mitochondria and the liver, the effects of ketosis on oxidative stress pathways, and the impact of KD on liver and mitochondrial function.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Research Center for High Performance Sport, UCAM Catholic University of Murcia, 30107 Murcia, Spain
| | - Giuseppe Cerullo
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
7
|
Kadir AA, Stubbs BJ, Chong C, Lee H, Cole M, Carr C, Hauton D, McCullagh J, Evans RD, Clarke K. On the interdependence of ketone body oxidation, glycogen content, glycolysis and energy metabolism in the heart. J Physiol 2023; 601:1207-1224. [PMID: 36799478 PMCID: PMC10684314 DOI: 10.1113/jp284270] [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: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
In heart, glucose and glycolysis are important for anaplerosis and potentially therefore for d-β-hydroxybutyrate (βHB) oxidation. As a glucose store, glycogen may also furnish anaplerosis. We determined the effects of glycogen content on βHB oxidation and glycolytic rates, and their downstream effects on energetics, in the isolated rat heart. High glycogen (HG) and low glycogen (LG) containing hearts were perfused with 11 mM [5-3 H]glucose and/or 4 mM [14 C]βHB to measure glycolytic rates or βHB oxidation, respectively, then freeze-clamped for glycogen and metabolomic analyses. Free cytosolic [NAD+ ]/[NADH] and mitochondrial [Q+ ]/[QH2 ] ratios were estimated using the lactate dehydrogenase and succinate dehydrogenase reaction, respectively. Phosphocreatine (PCr) and inorganic phosphate (Pi ) concentrations were measured using 31 P-nuclear magnetic resonance spectroscopy. Rates of βHB oxidation in LG hearts were half that in HG hearts, with βHB oxidation directly proportional to glycogen content. βHB oxidation decreased glycolysis in all hearts. Glycogenolysis in glycogen-replete hearts perfused with βHB alone was twice that of hearts perfused with βHB and glucose, which had significantly higher levels of the glycolytic intermediates fructose 1,6-bisphosphate and 3-phosphoglycerate, and higher free cytosolic [NAD+ ]/[NADH]. βHB oxidation increased the Krebs cycle intermediates citrate, 2-oxoglutarate and succinate, the total NADP/H pool, reduced mitochondrial [Q+ ]/[QH2 ], and increased the calculated free energy of ATP hydrolysis (∆GATP ). Although βHB oxidation inhibited glycolysis, glycolytic intermediates were not depleted, and cytosolic free NAD remained oxidised. βHB oxidation alone increased Krebs cycle intermediates, reduced mitochondrial Q and increased ∆GATP . We conclude that glycogen facilitates cardiac βHB oxidation by anaplerosis. KEY POINTS: Ketone bodies (d-β-hydroxybutyrate, acetoacetate) are increasingly recognised as important cardiac energetic substrates, in both healthy and diseased hearts. As 2-carbon equivalents they are cataplerotic, causing depletion of Krebs cycle intermediates; therefore their utilisation requires anaplerotic supplementation, and intra-myocardial glycogen has been suggested as a potential anaplerotic source during ketone oxidation. It is demonstrated here that cardiac glycogen does indeed provide anaplerotic substrate to facilitate β-hydroxybutyrate oxidation in isolated perfused rat heart, and this contribution was quantified using a novel pulse-chase metabolic approach. Further, using metabolomics and 31 P-MR, it was shown that glycolytic flux from myocardial glycogen increased the heart's ability to oxidise βHB, and βHB oxidation increased the mitochondrial redox potential, ultimately increasing the free energy of ATP hydrolysis.
Collapse
Affiliation(s)
- Azrul Abdul Kadir
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | | | - Cher‐Rin Chong
- Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
| | - Henry Lee
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Mark Cole
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Carolyn Carr
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - David Hauton
- Department of ChemistryUniversity of OxfordOxfordUK
| | | | - Rhys D. Evans
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Kieran Clarke
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
8
|
Fakhrolmobasheri M, Abhari AP, Manshaee B, Heidarpour M, Shafie D, Mohammadbeigi E, Mozafari AM, Mazaheri-Tehrani S. Effect of sodium-glucose cotransporter 2 inhibitors on insulin resistance; a systematic review and meta-analysis. Acta Diabetol 2023; 60:191-202. [PMID: 36264350 DOI: 10.1007/s00592-022-01981-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/25/2022] [Indexed: 01/24/2023]
Abstract
AIM Recent studies have indicated that Sodium-GLucose co-Transporter 2 Inhibitors (SGLT2Is) may increase insulin sensitivity (IS); however, these results are heterogeneous and need to be systematically assessed. METHOD We searched MEDLINE/PubMed, Embase, Web of Science, Scopus, Cochrane Library, Ovid, and ProQuest using a predefined search query. Randomized clinical trials on SGLT2Is with a passive control group or metformin controlled group were included. Risk of bias assessment was performed using the Cochrane risk of bias assessment tool. Meta-analysis was performed separately on studies with type 2 diabetes mellitus (T2DM) population and studies with non-T2DM population and also for passive- and active-controlled studies using standardized mean difference (SMD) as the measure of the effect size. Subgroup analysis was performed according to different types of SGLT2Is. Meta-regression analysis was performed according to the dose and duration of intervention. RESULTS Twenty-two studies (6 on non-T2DM population) with a total of 1421 (243 non-T2DM) patients were included. Six studies (3 on T2DM and 3 on non-T2DM) were controlled by metformin, and others were passively controlled. SGLT2Is could significantly increase IS in T2DM patients (SMD = 0.72 [0.32-1.12]). SGLT2Is could reduce insulin resistance in non-T2DM population, but this was not significant. SGLT2Is were not inferior to metformin in reducing insulin resistance. Subgroup analysis indicated that dapagliflozin could significantly increase IS, but empagliflozin was not associated with significant improvement in IS. Meta-regression analysis indicated no effect for dose but duration of SGLT2I administration on IS. CONCLUSION SGLT2Is, particularly dapagliflozin, could increase IS. These results need to be consolidated by further studies.
Collapse
Affiliation(s)
- Mohammad Fakhrolmobasheri
- Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Parsa Abhari
- Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrad Manshaee
- Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Heidarpour
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ehsan Mohammadbeigi
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Mohammad Mozafari
- Medical Library and Information Sciences Department, Health Information Technology Research Center, School of Management and Medical Information Sciences, Isfahan University of Medical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sadegh Mazaheri-Tehrani
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Saris CGJ, Timmers S. Ketogenic diets and Ketone suplementation: A strategy for therapeutic intervention. Front Nutr 2022; 9:947567. [PMID: 36458166 PMCID: PMC9705794 DOI: 10.3389/fnut.2022.947567] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/13/2022] [Indexed: 07/24/2023] Open
Abstract
Ketogenic diets and orally administered exogenous ketone supplements are strategies to increase serum ketone bodies serving as an alternative energy fuel for high energy demanding tissues, such as the brain, muscles, and the heart. The ketogenic diet is a low-carbohydrate and fat-rich diet, whereas ketone supplements are usually supplied as esters or salts. Nutritional ketosis, defined as serum ketone concentrations of ≥ 0.5 mmol/L, has a fasting-like effect and results in all sorts of metabolic shifts and thereby enhancing the health status. In this review, we thus discuss the different interventions to reach nutritional ketosis, and summarize the effects on heart diseases, epilepsy, mitochondrial diseases, and neurodegenerative disorders. Interest in the proposed therapeutic benefits of nutritional ketosis has been growing the past recent years. The implication of this nutritional intervention is becoming more evident and has shown interesting potential. Mechanistic insights explaining the overall health effects of the ketogenic state, will lead to precision nutrition for the latter diseases.
Collapse
Affiliation(s)
- Christiaan G. J. Saris
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
- Radboud Center for Mitochondrial Medicine, Nijmegen, Netherlands
| | - Silvie Timmers
- Department of Human and Animal Physiology, Wageningen University, Wageningen, Netherlands
| |
Collapse
|
10
|
Selvaraj S, Hu R, Vidula MK, Dugyala S, Tierney A, Ky B, Margulies KB, Shah SH, Kelly DP, Bravo PE. Acute Echocardiographic Effects of Exogenous Ketone Administration in Healthy Participants. J Am Soc Echocardiogr 2022; 35:305-311. [PMID: 34798244 PMCID: PMC8901445 DOI: 10.1016/j.echo.2021.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Interest in therapeutic applications of exogenous ketones has grown significantly, spanning patients with heart failure to endurance athletes. Exogenous ketones engender significant effects on cardiac function in heart failure and provide an ergogenic benefit in athletes. The aim of this study was to assess the effects of exogenous ketones on cardiac function in healthy participants. METHODS In a single-arm intervention study, 20 fasting, healthy participants underwent comprehensive echocardiography (two-dimensional, Doppler, and strain) before and 30 min after weight-based oral ketone ester administration. The relationship between changes in log-transformed biomarker levels and change in absolute global longitudinal strain (GLS) was assessed using linear regression. RESULTS The mean age was 30 ± 7 years, 50% were women, 45% were nonwhite, and the average body mass index was 24.3 ± 3.1 kg/m2. Ketone ingestion acutely elevated β-hydroxybutyrate levels from a median of 0.13 mmol/L (interquartile range, 0.10-0.37 mmol/L) to 3.23 mmol/L (interquartile range, 2.40-4.97 mmol/L) (P < .001). After ketone ester consumption, systolic blood pressure, heart rate, biventricular function, left ventricular GLS, and left atrial (LA) strain all augmented, while systemic vascular resistance decreased. Displayed as mean change, increases in ejection fraction (3.1%; 95% CI, 2.0%-4.2%; P < .001), GLS (2.0%; 95% CI, 1.4%-2.7%; P < .001), right ventricular S' (1.1 cm/sec; 95% CI, 0.4-1.8 cm/sec; P = .004), LA reservoir strain (7%; 95% CI, 3%-12%; P = .005), and LA contractile strain (4%; 2%-6%; P = .001) were observed. During robustly achieved ketosis, change in GLS was inversely associated with change in nonesterified fatty acids (P = .019). CONCLUSIONS In a single-arm study, systolic blood pressure, heart rate, biventricular function, and LV and LA strain acutely augmented after ketone ester ingestion in healthy, fasting participants, similar to several effects observed in the failing heart. These data may provide supporting data for the ergogenic benefits observed in athletes and may become increasingly relevant with exogenous ketone consumption across a variety of cardiovascular and noncardiovascular applications.
Collapse
Affiliation(s)
- Senthil Selvaraj
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Ray Hu
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mahesh K Vidula
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Supritha Dugyala
- Division of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ann Tierney
- Department of Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth B Margulies
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Daniel P Kelly
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paco E Bravo
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Recent Advances in Understanding of Alzheimer's Disease Progression through Mass Spectrometry-Based Metabolomics. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:1-17. [PMID: 35656096 PMCID: PMC9159642 DOI: 10.1007/s43657-021-00036-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the aging population, but despite extensive research, there is no consensus on the biological cause of AD. While AD research is dominated by protein/peptide-centric research based on the amyloid hypothesis, a theory that designates dysfunction in beta-amyloid production, accumulation, or disposal as the primary cause of AD, many studies focus on metabolomics as a means of understanding the biological processes behind AD progression. In this review, we discuss mass spectrometry (MS)-based AD metabolomics studies, including sample type and preparation, mass spectrometry specifications, and data analysis, as well as biological insights gleaned from these studies, with the hope of informing future AD metabolomic studies.
Collapse
|
12
|
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
|
13
|
Prins PJ, Buxton JD, McClure TS, D'Agostino DP, Ault DL, Welton GL, Jones DW, Atwell AD, Slack MA, Slack ML, Williams CE, Blanchflower ME, Kannel KK, Faulkner MN, Szmaciasz HL, Croll SM, Stanforth LM, Harris TD, Gwaltney HC, Koutnik AP. Ketone Bodies Impact on Hypoxic CO 2 Retention Protocol During Exercise. Front Physiol 2021; 12:780755. [PMID: 34966291 PMCID: PMC8711099 DOI: 10.3389/fphys.2021.780755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
Exogenous ketone esters have demonstrated the capacity to increase oxygen availability during acute hypoxic exposure leading to the potential application of their use to mitigate performance declines at high altitudes. Voluntary hypoventilation (VH) with exercise reliably reduces oxygen availability and increases carbon dioxide retention without alterations to ambient pressure or gas content. Utilizing a double-blind randomized crossover design, fifteen recreational male distance runners performed submaximal exercise (4 × 5 min; 70% VO2 Max) with VH. An exogenous ketone ester (KME; 573 mg⋅kg–1) or iso-caloric flavor matched placebo (PLA) was consumed prior to exercise. Metabolites, blood gases, expired air, heart rate, oxygen saturation, cognition, and perception metrics were collected throughout. KME rapidly elevated R-β-hydroxybutyrate and reduced blood glucose without altering lactate production. KME lowered pH, bicarbonate, and total carbon dioxide. VH with exercise significantly reduced blood (SpO2) and muscle (SmO2) oxygenation and increased cognitive mean reaction time and respiratory rate regardless of condition. KME administration significantly elevated respiratory exchange ratio (RER) at rest and throughout recovery from VH, compared to PLA. Blood carbon dioxide (PCO2) retention increased in the PLA condition while decreasing in the KME condition, leading to a significantly lower PCO2 value immediately post VH exercise (IPE; p = 0.031) and at recovery (p = 0.001), independent of respiratory rate. The KME’s ability to rapidly alter metabolism, acid/base balance, CO2 retention, and respiratory exchange rate independent of respiratory rate changes at rest, during, and/or following VH exercise protocol illustrates a rapid countermeasure to CO2 retention in concert with systemic metabolic changes.
Collapse
Affiliation(s)
- Philip J Prins
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Jeffrey D Buxton
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Tyler S McClure
- Human Healthspan, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, FL, United States
| | - Dominic P D'Agostino
- Human Healthspan, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, FL, United States.,Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | - Dana L Ault
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Gary L Welton
- Department of Psychology, Grove City College, Grove City, PA, United States
| | - Dalton W Jones
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Adam D Atwell
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Macey A Slack
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Marah L Slack
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Chloe E Williams
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | | | - Kristia K Kannel
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Madison N Faulkner
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Hannah L Szmaciasz
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Stephanie M Croll
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Lindsey M Stanforth
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Tim D Harris
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Holton C Gwaltney
- Department of Exercise Science, Grove City College, Grove City, PA, United States
| | - Andrew P Koutnik
- Human Healthspan, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, FL, United States
| |
Collapse
|
14
|
Kolwicz SC. Ketone Body Metabolism in the Ischemic Heart. Front Cardiovasc Med 2021; 8:789458. [PMID: 34950719 PMCID: PMC8688810 DOI: 10.3389/fcvm.2021.789458] [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: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 01/12/2023] Open
Abstract
Ketone bodies have been identified as an important, alternative fuel source in heart failure. In addition, the use of ketone bodies as a fuel source has been suggested to be a potential ergogenic aid for endurance exercise performance. These findings have certainly renewed interest in the use of ketogenic diets and exogenous supplementation in an effort to improve overall health and disease. However, given the prevalence of ischemic heart disease and myocardial infarctions, these strategies may not be ideal for individuals with coronary artery disease. Although research studies have clearly defined changes in fatty acid and glucose metabolism during ischemia and reperfusion, the role of ketone body metabolism in the ischemic and reperfused myocardium is less clear. This review will provide an overview of ketone body metabolism, including the induction of ketosis via physiological or nutritional strategies. In addition, the contribution of ketone body metabolism in healthy and diseased states, with a particular emphasis on ischemia-reperfusion (I-R) injury will be discussed.
Collapse
|
15
|
Poffé C, Robberechts R, Podlogar T, Kusters M, Debevec T, Hespel P. Exogenous ketosis increases blood and muscle oxygenation but not performance during exercise in hypoxia. Am J Physiol Regul Integr Comp Physiol 2021; 321:R844-R857. [PMID: 34668436 DOI: 10.1152/ajpregu.00198.2021] [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] [Indexed: 01/03/2023]
Abstract
Available evidence indicates that elevated blood ketones are associated with improved hypoxic tolerance in rodents. From this perspective, we hypothesized that exogenous ketosis by oral intake of the ketone ester (R)-3-hydroxybutyl (R)-3-hydroxybutyrate (KE) may induce beneficial physiological effects during prolonged exercise in acute hypoxia. As we recently demonstrated KE to deplete blood bicarbonate, which per se may alter the physiological response to hypoxia, we evaluated the effect of KE both in the presence and absence of bicarbonate intake (BIC). Fourteen highly trained male cyclists performed a simulated cycling race (RACE) consisting of 3-h intermittent cycling (IMT180') followed by a 15-min time-trial (TT15') and an all-out sprint at 175% of lactate threshold (SPRINT). During RACE, fraction of inspired oxygen ([Formula: see text]) was gradually decreased from 18.6% to 14.5%. Before and during RACE, participants received either 1) 75 g of ketone ester (KE), 2) 300 mg/kg body mass bicarbonate (BIC), 3) KE + BIC, or 4) a control drink in addition to 60 g of carbohydrates/h in a randomized, crossover design. KE counteracted the hypoxia-induced drop in blood ([Formula: see text]) and muscle oxygenation by ∼3%. In contrast, BIC decreased [Formula: see text] by ∼2% without impacting muscle oxygenation. Performance during TT15' and SPRINT were similar between all conditions. In conclusion, KE slightly elevated the degree of blood and muscle oxygenation during prolonged exercise in moderate hypoxia without impacting exercise performance. Our data warrant to further investigate the potential of exogenous ketosis to improve muscular and cerebral oxygenation status, and exercise tolerance in extreme hypoxia.
Collapse
Affiliation(s)
- Chiel Poffé
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Ruben Robberechts
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Tim Podlogar
- Department for Automation, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Martijn Kusters
- Bakala Academy-Athletic Performance Center, KU Leuven, Leuven, Belgium
| | - Tadej Debevec
- Department for Automation, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Sport, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Hespel
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium.,Bakala Academy-Athletic Performance Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Stubbs BJ, Koutnik AP, Goldberg EL, Upadhyay V, Turnbaugh PJ, Verdin E, Newman JC. Investigating Ketone Bodies as Immunometabolic Countermeasures against Respiratory Viral Infections. MED 2020; 1:43-65. [PMID: 32838361 PMCID: PMC7362813 DOI: 10.1016/j.medj.2020.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Respiratory viral infections remain a scourge, with seasonal influenza infecting millions and killing many thousands annually and viral pandemics, such as COVID-19, recurring every decade. Age, cardiovascular disease, and diabetes mellitus are risk factors for severe disease and death from viral infection. Immunometabolic therapies for these populations hold promise to reduce the risks of death and disability. Such interventions have pleiotropic effects that might not only target the virus itself but also enhance supportive care to reduce cardiopulmonary complications, improve cognitive resilience, and facilitate functional recovery. Ketone bodies are endogenous metabolites that maintain cellular energy but also feature drug-like signaling activities that affect immune activity, metabolism, and epigenetics. Here, we provide an overview of ketone body biology relevant to respiratory viral infection, focusing on influenza A and severe acute respiratory syndrome (SARS)-CoV-2, and discuss the opportunities, risks, and research gaps in the study of exogenous ketone bodies as novel immunometabolic interventions in these diseases.
Collapse
Affiliation(s)
| | - Andrew P Koutnik
- Institute for Human and Machine Cognition, Pensacola, FL, USA
- Department of Molecular Pharmacology and Physiology, USF, Tampa, FL, USA
| | | | - Vaibhav Upadhyay
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, UCSF, San Francisco, CA, USA
- Department of Microbiology and Immunology, UCSF, San Francisco, CA, USA
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, UCSF, San Francisco, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Division of Geriatrics, UCSF, San Francisco, CA, USA
| |
Collapse
|
17
|
Habieb ME, Mohamed MA, El Gamal DM, Hawas AM, Mohamed TM. Anti-aging effect of DL-β-hydroxybutyrate against hepatic cellular senescence induced by D-galactose or γ-irradiation via autophagic flux stimulation in male rats. Arch Gerontol Geriatr 2020; 92:104288. [PMID: 33147533 DOI: 10.1016/j.archger.2020.104288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/30/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022]
Abstract
The present study aims to shed new light on anti-aging effect of DL-β-hydroxybutyrate (βOHB) against hepatic cellular senescence induced by d-galactose or γ-irradiation. The rats divided into 6 groups. Group 1, control, group 2, exposed to γ-ray (5 GY), group 3, injected by d-galactose (150 mg/kg) daily for consecutive 6 weeks, which regarded to induce the aging, group 4, injected intraperitoneal by β-hydroxybutyrate (βOHB) (72.8 mg/kg) daily for consecutive 14 days, group 5, exposed to γ-ray then treated with βOHB daily for consecutive 14 days, group 6, injected daily with d-galactose for consecutive 6 weeks, then treated with βOHB daily at the last two weeks of d-galactose. Aspartate amino transferase (AST), alanine amino transferase (ALT), Insulin, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were estimated in serum. Moreover, protein expression of Microtubule-associated proteins 1A/1B light chain 3B (LC3-II/LC3-I) ratio, mechanistic target of rapamycin (mTOR), pAMPK, mRNA gene expression of 5' AMP-activated protein kinase (AMPK), Nucleoporin p62 (p62), cyclin-dependent kinase inhibitor 1(P21CIP1), cyclin-dependent kinase inhibitor 2A (p16INK4a) and DNA fragmentation percentage were measured in liver tissue as a biomarker of cellular senescence. The results confirmed that βOHB modulated serum level of AST, ALT, insulin, IL-6 and TNF-α, protein expression of mTOR and LC3-II/LC3-I ratio, pAMPK and p62 in liver aging model induced by d-galactose or γ-irradiation. Histopathological examination results of liver tissue indicated coincidence with those recorded by molecular biochemical inspection. Taken together, these findings suggest that βOHB may be useful in combating hepatic cellular senescence induced by d-galactose or γ-irradiation via autophagy dependent mechanisms.
Collapse
Affiliation(s)
- M E Habieb
- Drug Radiation Research Dept., National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, P.O. Box, 29 Nasr City, Cairo, Egypt.
| | - M A Mohamed
- Drug Radiation Research Dept., National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, P.O. Box, 29 Nasr City, Cairo, Egypt
| | - D M El Gamal
- Biochemistry Division, Chemistry Dept., Faculty of Science, Tanta University, Egypt
| | - A M Hawas
- Drug Radiation Research Dept., National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, P.O. Box, 29 Nasr City, Cairo, Egypt
| | - T M Mohamed
- Biochemistry Division, Chemistry Dept., Faculty of Science, Tanta University, Egypt
| |
Collapse
|
18
|
Stubbs BJ, Koutnik AP, Volek JS, Newman JC. From bedside to battlefield: intersection of ketone body mechanisms in geroscience with military resilience. GeroScience 2020; 43:1071-1081. [PMID: 33006708 PMCID: PMC8190215 DOI: 10.1007/s11357-020-00277-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Ketone bodies are endogenous metabolites that are linked to multiple mechanisms of aging and resilience. They are produced by the body when glucose availability is low, including during fasting and dietary carbohydrate restriction, but also can be consumed as exogenous ketone compounds. Along with supplying energy to peripheral tissues such as brain, heart, and skeletal muscle, they increasingly are understood to have drug-like protein binding activities that regulate inflammation, epigenetics, and other cellular processes. While these energy and signaling mechanisms of ketone bodies are currently being studied in a variety of aging-related diseases such as Alzheimer’s disease and type 2 diabetes mellitus, they may also be relevant to military service members undergoing stressors that mimic or accelerate aging pathways, particularly traumatic brain injury and muscle rehabilitation and recovery. Here we summarize the biology of ketone bodies relevant to resilience and rehabilitation, strategies for translational use of ketone bodies, and current clinical investigations in this area.
Collapse
Affiliation(s)
| | - Andrew P Koutnik
- Institute for Human and Machine Cognition, Pensacola, FL, USA.,Department of Molecular Pharmacology and Physiology, USF, Tampa, FL, USA
| | - Jeff S Volek
- Department of Human Sciences, Ohio State University, Columbus, OH, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato, CA, USA. .,Division of Geriatrics, UCSF, San Francisco, CA, USA.
| |
Collapse
|
19
|
Shippy DC, Wilhelm C, Viharkumar PA, Raife TJ, Ulland TK. β-Hydroxybutyrate inhibits inflammasome activation to attenuate Alzheimer's disease pathology. J Neuroinflammation 2020; 17:280. [PMID: 32958021 PMCID: PMC7507727 DOI: 10.1186/s12974-020-01948-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive, late-onset dementia with no effective treatment available. Recent studies suggest that AD pathology is driven by age-related changes in metabolism. Alterations in metabolism, such as placing patients on a ketogenic diet, can alter cognition by an unknown mechanism. One of the ketone bodies produced as a result of ketogenesis, β-hydroxybutyrate (BHB), is known to inhibit NLRP3 inflammasome activation. Therefore, we tested if BHB inhibition of the NLRP3 inflammasome reduces overall AD pathology in the 5XFAD mouse model of AD. Here, we find BHB levels are lower in red blood cells and brain parenchyma of AD patients when compared with non-AD controls. Furthermore, exogenous BHB administration reduced plaque formation, microgliosis, apoptosis-associated speck-like protein containing a caspase recruitment domain (Asc) speck formation, and caspase-1 activation in the 5XFAD mouse model of AD. Taken together, our findings demonstrate that BHB reduces AD pathology by inhibiting NLRP3 inflammasome activation. Additionally, our data suggest dietary or pharmacological approaches to increase BHB levels as promising therapeutic strategies for AD.
Collapse
Affiliation(s)
- Daniel C Shippy
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Connor Wilhelm
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Patel A Viharkumar
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Thomas J Raife
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
20
|
Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, Levasseur J, Leone T, Dyck JRB, Ussher JR, Muoio DM, Kelly DP, Lopaschuk GD. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res 2020; 115:1606-1616. [PMID: 30778524 DOI: 10.1093/cvr/cvz045] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/18/2018] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
AIMS The failing heart is energy-starved and inefficient due to perturbations in energy metabolism. Although ketone oxidation has been shown recently to increase in the failing heart, it remains unknown whether this improves cardiac energy production or efficiency. We therefore assessed cardiac metabolism in failing hearts and determined whether increasing ketone oxidation improves cardiac energy production and efficiency. METHODS AND RESULTS C57BL/6J mice underwent sham or transverse aortic constriction (TAC) surgery to induce pressure overload hypertrophy over 4-weeks. Isolated working hearts from these mice were perfused with radiolabelled β-hydroxybutyrate (βOHB), glucose, or palmitate to assess cardiac metabolism. Ejection fraction decreased by 45% in TAC mice. Failing hearts had decreased glucose oxidation while palmitate oxidation remained unchanged, resulting in a 35% decrease in energy production. Increasing βOHB levels from 0.2 to 0.6 mM increased ketone oxidation rates from 251 ± 24 to 834 ± 116 nmol·g dry wt-1 · min-1 in TAC hearts, rates which were significantly increased compared to sham hearts and occurred without decreasing glycolysis, glucose, or palmitate oxidation rates. Therefore, the contribution of ketones to energy production in TAC hearts increased to 18% and total energy production increased by 23%. Interestingly, glucose oxidation, in parallel with total ATP production, was also significantly upregulated in hearts upon increasing βOHB levels. However, while overall energy production increased, cardiac efficiency was not improved. CONCLUSIONS Increasing ketone oxidation rates in failing hearts increases overall energy production without compromising glucose or fatty acid metabolism, albeit without increasing cardiac efficiency.
Collapse
Affiliation(s)
- Kim L Ho
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Cory Wagg
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Rami Al Batran
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Jody Levasseur
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Teresa Leone
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, USA
| | - Jason R B Dyck
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, 300 N Duke St, Durham, NC, USA
| | - Daniel P Kelly
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, USA
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
21
|
Cardiac ketone body metabolism. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165739. [PMID: 32084511 DOI: 10.1016/j.bbadis.2020.165739] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022]
Abstract
The ketone bodies, d-β-hydroxybutyrate and acetoacetate, are soluble 4-carbon compounds derived principally from fatty acids, that can be metabolised by many oxidative tissues, including heart, in carbohydrate-depleted conditions as glucose-sparing energy substrates. They also have important signalling functions, acting through G-protein coupled receptors and histone deacetylases to regulate metabolism and gene expression including that associated with anti-oxidant activity. Their concentration, and hence availability, increases in diabetes mellitus and heart failure. Whilst known to be substrates for ATP production, especially in starvation, their role(s) in the heart, and in heart disease, is uncertain. Recent evidence, reviewed here, indicates that increased ketone body metabolism is a feature of heart failure, and is accompanied by other changes in substrate selection. Whether the change in myocardial ketone body metabolism is adaptive or maladaptive is unknown, but it offers the possibility of using exogenous ketones to treat the failing heart.
Collapse
|
22
|
Harvey KL, Holcomb LE, Kolwicz SC. Ketogenic Diets and Exercise Performance. Nutrients 2019; 11:nu11102296. [PMID: 31561520 PMCID: PMC6835497 DOI: 10.3390/nu11102296] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
The ketogenic diet (KD) has gained a resurgence in popularity due to its purported reputation for fighting obesity. The KD has also acquired attention as an alternative and/or supplemental method for producing energy in the form of ketone bodies. Recent scientific evidence highlights the KD as a promising strategy to treat obesity, diabetes, and cardiac dysfunction. In addition, studies support ketone body supplements as a potential method to induce ketosis and supply sustainable fuel sources to promote exercise performance. Despite the acceptance in the mainstream media, the KD remains controversial in the medical and scientific communities. Research suggests that the KD or ketone body supplementation may result in unexpected side effects, including altered blood lipid profiles, abnormal glucose homeostasis, increased adiposity, fatigue, and gastrointestinal distress. The purpose of this review article is to provide an overview of ketone body metabolism and a background on the KD and ketone body supplements in the context of obesity and exercise performance. The effectiveness of these dietary or supplementation strategies as a therapy for weight loss or as an ergogenic aid will be discussed. In addition, the recent evidence that indicates ketone body metabolism is a potential target for cardiac dysfunction will be reviewed.
Collapse
Affiliation(s)
- Kristin L Harvey
- Heart and Muscle Metabolism Laboratory, Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA.
| | - Lola E Holcomb
- Heart and Muscle Metabolism Laboratory, Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA.
| | - Stephen C Kolwicz
- Heart and Muscle Metabolism Laboratory, Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA.
| |
Collapse
|
23
|
Tentolouris A, Vlachakis P, Tzeravini E, Eleftheriadou I, Tentolouris N. SGLT2 Inhibitors: A Review of Their Antidiabetic and Cardioprotective Effects. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E2965. [PMID: 31426529 PMCID: PMC6720282 DOI: 10.3390/ijerph16162965] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is a chronic metabolic disease associated with high cardiovascular (CV) risk. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are the latest class of antidiabetic medication that inhibit the absorption of glucose from the proximal tubule of the kidney and hence cause glycosuria. Four SGLT2i are currently commercially available in many countries: canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin. SGLT2i reduce glycated hemoglobin by 0.5%-1.0% and have shown favorable effects on body weight, blood pressure, lipid profile, arterial stiffness and endothelial function. More importantly, SGLT2i have demonstrated impressive cardioprotective and renoprotective effects. The main mechanisms underlying their cardioprotective effects have been attributed to improvement in cardiac cell metabolism, improvement in ventricular loading conditions, inhibition of the Na+/H+ exchange in the myocardial cells, alteration in adipokines and cytokines production, as well as reduction of cardiac cells necrosis and cardiac fibrosis. The main adverse events of SGLT2i include urinary tract and genital infections, as well as euglycemic diabetic ketoacidosis. Concerns have also been raised about the association of SGLT2i with lower limb amputations, Fournier gangrene, risk of bone fractures, female breast cancer, male bladder cancer, orthostatic hypotension, and acute kidney injury.
Collapse
Affiliation(s)
- Anastasios Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | - Panayotis Vlachakis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | - Evangelia Tzeravini
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece.
| |
Collapse
|
24
|
Abstract
Diabetes is a global epidemic and a leading cause of death with more than 422 million patients worldwide out of whom around 392 million alone suffer from type 2 diabetes (T2D). Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are novel and effective drugs in managing glycemia of T2D patients. These inhibitors gained recent clinical and basic research attention due to their clinically observed cardiovascular protective effects. Although interest in the study of various SGLT isoforms and the effect of their inhibition on cardiovascular function extends over the past 20 years, an explanation of the effects observed clinically based on available experimental data is not forthcoming. The remarkable reduction in cardiovascular (CV) mortality (38%), major CV events (14%), hospitalization for heart failure (35%), and death from any cause (32%) observed over a period of 2.6 years in patients with T2D and high CV risk in the EMPA-REG OUTCOME trial involving the SGLT2 inhibitor empagliflozin (Empa) have raised the possibility that potential novel, more specific mechanisms of SGLT2 inhibition synergize with the known modest systemic improvements, such as glycemic, body weight, diuresis, and blood pressure control. Multiple studies investigated the direct impact of SGLT2i on the cardiovascular system with limited findings and the pathophysiological role of SGLTs in the heart. The direct impact of SGLT2i on cardiac homeostasis remains controversial, especially that SGLT1 isoform is the only form expressed in the capillaries and myocardium of human and rodent hearts. The direct impact of SGLT2i on the cardiovascular system along with potential lines of future research is summarized in this review.
Collapse
|
25
|
Veech RL, Todd King M, Pawlosky R, Kashiwaya Y, Bradshaw PC, Curtis W. The "great" controlling nucleotide coenzymes. IUBMB Life 2019; 71:565-579. [PMID: 30624851 PMCID: PMC6850382 DOI: 10.1002/iub.1997] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
Nucleotide coenzymes dot the map of metabolic pathways providing energy to drive the reactions of the pathway and play an important role in regulating and controlling energy metabolism through their shared potential energy, which is widely unobserved due to the paradox that the energy in the coenzyme pools cannot be determined from the concentration of the coenzyme couples. The potential energy of the nucleotide couples in the mitochondria or the cytoplasm is expressed in the enzyme reactions in which they take part. The energy in these couples, [NAD+]/[NADH], [NADP+]/[NADPH], [acetyl CoA]/[CoA], and [ATP]/[ADP]x[Pi], regulates energy metabolism. The energy contained in the couples can be altered by suppling energy equivalents in the form of ketones, such as, D-β-hydroxybutyrate to overcome insulin resistance, to restore antioxidants capacity, to form potential treatments for Alzheimer's and Parkinson's diseases, to enhance life span, and to increase physiological performance. © 2019 IUBMB Life, 71(5):565-579, 2019.
Collapse
Affiliation(s)
- Richard L Veech
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Michael Todd King
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Robert Pawlosky
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | | | - Patrick C Bradshaw
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - William Curtis
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| |
Collapse
|
26
|
Hansson NH, Harms HJ, Kim WY, Nielsen R, Tolbod LP, Frøkiær J, Bouchelouche K, Poulsen SH, Wiggers H, Parner ET, Sörensen J. Test-retest repeatability of myocardial oxidative metabolism and efficiency using standalone dynamic 11C-acetate PET and multimodality approaches in healthy controls. J Nucl Cardiol 2018; 25:1929-1936. [PMID: 29855984 DOI: 10.1007/s12350-018-1302-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/25/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Myocardial efficiency measured by 11C-acetate positron emission tomography (PET) has successfully been used in clinical research to quantify mechanoenergetic coupling. The objective of this study was to establish the repeatability of myocardial external efficiency (MEE) and work metabolic index (WMI) by non-invasive concepts. METHODS AND RESULTS Ten healthy volunteers (63 ± 4 years) were examined twice, one week apart, using 11C-acetate PET, cardiovascular magnetic resonance (CMR), and echocardiography. Myocardial oxygen consumption from PET was combined with stroke work data from CMR, echocardiography, or PET to obtain MEE and WMI for each modality. Repeatability was estimated as the coefficient of variation (CV) between test and retest. MEECMR, MEEEcho, and MEEPET values were 21.9 ± 2.7%, 16.4 ± 3.7%, and 23.8 ± 4.9%, respectively, P < .001. WMICMR, WMIEcho, and WMIPET values were 4.42 ± 0.90, 4.07 ± 0.63, and 4.58 ± 1.13 mmHg × mL/m2 × 106, respectively, P = .45. Repeatability for MEECMR was superior compared with MEEEcho but did not differ significantly compared with MEEPET (6.3% vs 12.9% and 9.4%, P = .04 and .25). CV values for WMICMR, WMIEcho, and WMIPET were 10.0%, 14.8%, and 12.0%, respectively, (P = .53). CONCLUSIONS Non-invasive measurements of MEE using 11C-acetate PET are highly repeatable. A PET-only approach did not differ significantly from CMR/PET and might facilitate further clinical research due to lower costs and broader applicability.
Collapse
Affiliation(s)
| | - Hendrik Johannes Harms
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Roni Nielsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars P Tolbod
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Frøkiær
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsten Bouchelouche
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Erik Thorlund Parner
- Section for Biostatistics, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Jens Sörensen
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
27
|
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: 165] [Impact Index Per Article: 23.6] [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
|
28
|
Karwi QG, Uddin GM, Ho KL, Lopaschuk GD. Loss of Metabolic Flexibility in the Failing Heart. Front Cardiovasc Med 2018; 5:68. [PMID: 29928647 PMCID: PMC5997788 DOI: 10.3389/fcvm.2018.00068] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
To maintain its high energy demand the heart is equipped with a highly complex and efficient enzymatic machinery that orchestrates ATP production using multiple energy substrates, namely fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The contribution of these individual substrates to ATP production can dramatically change, depending on such variables as substrate availability, hormonal status and energy demand. This "metabolic flexibility" is a remarkable virtue of the heart, which allows utilization of different energy substrates at different rates to maintain contractile function. In heart failure, cardiac function is reduced, which is accompanied by discernible energy metabolism perturbations and impaired metabolic flexibility. While it is generally agreed that overall mitochondrial ATP production is impaired in the failing heart, there is less consensus as to what actual switches in energy substrate preference occur. The failing heart shift toward a greater reliance on glycolysis and ketone body oxidation as a source of energy, with a decrease in the contribution of glucose oxidation to mitochondrial oxidative metabolism. The heart also becomes insulin resistant. However, there is less consensus as to what happens to fatty acid oxidation in heart failure. While it is generally believed that fatty acid oxidation decreases, a number of clinical and experimental studies suggest that fatty acid oxidation is either not changed or is increased in heart failure. Of importance, is that any metabolic shift that does occur has the potential to aggravate cardiac dysfunction and the progression of the heart failure. An increasing body of evidence shows that increasing cardiac ATP production and/or modulating cardiac energy substrate preference positively correlates with heart function and can lead to better outcomes. This includes increasing glucose and ketone oxidation and decreasing fatty acid oxidation. In this review we present the physiology of the energy metabolism pathways in the heart and the changes that occur in these pathways in heart failure. We also look at the interventions which are aimed at manipulating the myocardial metabolic pathways toward more efficient substrate utilization which will eventually improve cardiac performance.
Collapse
Affiliation(s)
| | | | | | - Gary D. Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
29
|
Holdsworth DA, Cox PJ, Kirk T, Stradling H, Impey SG, Clarke K. A Ketone Ester Drink Increases Postexercise Muscle Glycogen Synthesis in Humans. Med Sci Sports Exerc 2018; 49:1789-1795. [PMID: 28398950 PMCID: PMC5556006 DOI: 10.1249/mss.0000000000001292] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction Physical endurance can be limited by muscle glycogen stores, in that glycogen depletion markedly reduces external work. During carbohydrate restriction, the liver synthesizes the ketone bodies, d-β-hydroxybutyrate, and acetoacetate from fatty acids. In animals and in the presence of glucose, d-β-hydroxybutyrate promotes insulin secretion and increases glycogen synthesis. Here we determined whether a dietary ketone ester, combined with plentiful glucose, can increase postexercise glycogen synthesis in human skeletal muscle. Methods After an interval-based glycogen depletion exercise protocol, 12 well-trained male athletes completed a randomized, three-arm, blinded crossover recovery study that consisted of consumption of either a taste-matched, zero-calorie control or a ketone monoester drink, followed by a 10-mM glucose clamp or saline infusion for 2 h. The three postexercise conditions were control drink then saline infusion, control drink then hyperglycemic clamp, or ketone ester drink then hyperglycemic clamp. Skeletal muscle glycogen content was determined in muscle biopsies of vastus lateralis taken before and after the 2-h clamps. Results The ketone ester drink increased blood d-β-hydroxybutyrate concentrations to a maximum of 5.3 versus 0.7 mM for the control drink (P < 0.0001). During the 2-h glucose clamps, insulin levels were twofold higher (31 vs 16 mU·L−1, P < 0.01) and glucose uptake 32% faster (1.66 vs 1.26 g·kg−1, P < 0.001). The ketone drink increased by 61 g, the total glucose infused for 2 h, from 197 to 258 g, and muscle glycogen was 50% higher (246 vs 164 mmol glycosyl units per kilogram dry weight, P < 0.05) than after the control drink. Conclusion In the presence of constant high glucose concentrations, a ketone ester drink increased endogenous insulin levels, glucose uptake, and muscle glycogen synthesis.
Collapse
Affiliation(s)
- David A Holdsworth
- 1Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UNITED KINGDOM; and 2Research Institute for Sport and Exercise Sciences, Liverpool John Moore's University, Liverpool, UNITED KINGDOM
| | | | | | | | | | | |
Collapse
|
30
|
Evans M, Cogan KE, Egan B. Metabolism of ketone bodies during exercise and training: physiological basis for exogenous supplementation. J Physiol 2017; 595:2857-2871. [PMID: 27861911 PMCID: PMC5407977 DOI: 10.1113/jp273185] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/24/2016] [Indexed: 01/04/2023] Open
Abstract
Optimising training and performance through nutrition strategies is central to supporting elite sportspeople, much of which has focused on manipulating the relative intake of carbohydrate and fat and their contributions as fuels for energy provision. The ketone bodies, namely acetoacetate, acetone and β-hydroxybutyrate (βHB), are produced in the liver during conditions of reduced carbohydrate availability and serve as an alternative fuel source for peripheral tissues including brain, heart and skeletal muscle. Ketone bodies are oxidised as a fuel source during exercise, are markedly elevated during the post-exercise recovery period, and the ability to utilise ketone bodies is higher in exercise-trained skeletal muscle. The metabolic actions of ketone bodies can alter fuel selection through attenuating glucose utilisation in peripheral tissues, anti-lipolytic effects on adipose tissue, and attenuation of proteolysis in skeletal muscle. Moreover, ketone bodies can act as signalling metabolites, with βHB acting as an inhibitor of histone deacetylases, an important regulator of the adaptive response to exercise in skeletal muscle. Recent development of ketone esters facilitates acute ingestion of βHB that results in nutritional ketosis without necessitating restrictive dietary practices. Initial reports suggest this strategy alters the metabolic response to exercise and improves exercise performance, while other lines of evidence suggest roles in recovery from exercise. The present review focuses on the physiology of ketone bodies during and after exercise and in response to training, with specific interest in exploring the physiological basis for exogenous ketone supplementation and potential benefits for performance and recovery in athletes.
Collapse
Affiliation(s)
- Mark Evans
- Institute for Sport and Health, School of Public Health, Physiotherapy and Sports ScienceUniversity College DublinBelfieldDublin4Ireland
| | - Karl E. Cogan
- Institute for Sport and Health, School of Public Health, Physiotherapy and Sports ScienceUniversity College DublinBelfieldDublin4Ireland
| | - Brendan Egan
- Institute for Sport and Health, School of Public Health, Physiotherapy and Sports ScienceUniversity College DublinBelfieldDublin4Ireland
- School of Health and Human PerformanceDublin City UniversityGlasnevinDublin9Ireland
| |
Collapse
|
31
|
Veech RL, Bradshaw PC, Clarke K, Curtis W, Pawlosky R, King MT. Ketone bodies mimic the life span extending properties of caloric restriction. IUBMB Life 2017; 69:305-314. [PMID: 28371201 DOI: 10.1002/iub.1627] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/10/2017] [Indexed: 12/27/2022]
Abstract
The extension of life span by caloric restriction has been studied across species from yeast and Caenorhabditis elegans to primates. No generally accepted theory has been proposed to explain these observations. Here, we propose that the life span extension produced by caloric restriction can be duplicated by the metabolic changes induced by ketosis. From nematodes to mice, extension of life span results from decreased signaling through the insulin/insulin-like growth factor receptor signaling (IIS) pathway. Decreased IIS diminishes phosphatidylinositol (3,4,5) triphosphate (PIP3 ) production, leading to reduced PI3K and AKT kinase activity and decreased forkhead box O transcription factor (FOXO) phosphorylation, allowing FOXO proteins to remain in the nucleus. In the nucleus, FOXO proteins increase the transcription of genes encoding antioxidant enzymes, including superoxide dismutase 2, catalase, glutathione peroxidase, and hundreds of other genes. An effective method for combating free radical damage occurs through the metabolism of ketone bodies, ketosis being the characteristic physiological change brought about by caloric restriction from fruit flies to primates. A dietary ketone ester also decreases circulating glucose and insulin leading to decreased IIS. The ketone body, d-β-hydroxybutyrate (d-βHB), is a natural inhibitor of class I and IIa histone deacetylases that repress transcription of the FOXO3a gene. Therefore, ketosis results in transcription of the enzymes of the antioxidant pathways. In addition, the metabolism of ketone bodies results in a more negative redox potential of the NADP antioxidant system, which is a terminal destructor of oxygen free radicals. Addition of d-βHB to cultures of C. elegans extends life span. We hypothesize that increasing the levels of ketone bodies will also extend the life span of humans and that calorie restriction extends life span at least in part through increasing the levels of ketone bodies. An exogenous ketone ester provides a new tool for mimicking the effects of caloric restriction that can be used in future research. The ability to power mitochondria in aged individuals that have limited ability to oxidize glucose metabolites due to pyruvate dehydrogenase inhibition suggests new lines of research for preventative measures and treatments for aging and aging-related disorders. © 2017 The Authors IUBMB Life published by Wiley Periodicals, Inc. on behalf of International Union of Biochemistry and Molecular Biology, 69(5):305-314, 2017.
Collapse
Affiliation(s)
| | - Patrick C Bradshaw
- East Tennessee State University College of Medicine, Johnson City, TN, USA
| | | | | | | | - M Todd King
- Lab of Metabolic Control, NIH/NIAAA, Rockville, MD, USA
| |
Collapse
|
32
|
|
33
|
Murray AJ, Knight NS, Cole MA, Cochlin LE, Carter E, Tchabanenko K, Pichulik T, Gulston MK, Atherton HJ, Schroeder MA, Deacon RMJ, Kashiwaya Y, King MT, Pawlosky R, Rawlins JNP, Tyler DJ, Griffin JL, Robertson J, Veech RL, Clarke K. Novel ketone diet enhances physical and cognitive performance. FASEB J 2016; 30:4021-4032. [PMID: 27528626 PMCID: PMC5102124 DOI: 10.1096/fj.201600773r] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Ketone bodies are the most energy-efficient fuel and yield more ATP per mole of substrate than pyruvate and increase the free energy released from ATP hydrolysis. Elevation of circulating ketones via high-fat, low-carbohydrate diets has been used for the treatment of drug-refractory epilepsy and for neurodegenerative diseases, such as Parkinson's disease. Ketones may also be beneficial for muscle and brain in times of stress, such as endurance exercise. The challenge has been to raise circulating ketone levels by using a palatable diet without altering lipid levels. We found that blood ketone levels can be increased and cholesterol and triglycerides decreased by feeding rats a novel ketone ester diet: chow that is supplemented with (R)-3-hydroxybutyl (R)-3-hydroxybutyrate as 30% of calories. For 5 d, rats on the ketone diet ran 32% further on a treadmill than did control rats that ate an isocaloric diet that was supplemented with either corn starch or palm oil (P < 0.05). Ketone-fed rats completed an 8-arm radial maze test 38% faster than did those on the other diets, making more correct decisions before making a mistake (P < 0.05). Isolated, perfused hearts from rats that were fed the ketone diet had greater free energy available from ATP hydrolysis during increased work than did hearts from rats on the other diets as shown by using [31P]-NMR spectroscopy. The novel ketone diet, therefore, improved physical performance and cognitive function in rats, and its energy-sparing properties suggest that it may help to treat a range of human conditions with metabolic abnormalities.-Murray, A. J., Knight, N. S., Cole, M. A., Cochlin, L. E., Carter, E., Tchabanenko, K., Pichulik, T., Gulston, M. K., Atherton, H. J., Schroeder, M. A., Deacon, R. M. J., Kashiwaya, Y., King, M. T., Pawlosky, R., Rawlins, J. N. P., Tyler, D. J., Griffin, J. L., Robertson, J., Veech, R. L., Clarke, K. Novel ketone diet enhances physical and cognitive performance.
Collapse
Affiliation(s)
- Andrew J Murray
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom;
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas S Knight
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Mark A Cole
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Lowri E Cochlin
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Emma Carter
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Tica Pichulik
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Melanie K Gulston
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Helen J Atherton
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Marie A Schroeder
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Robert M J Deacon
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Yoshihiro Kashiwaya
- Laboratory of Metabolic Control, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - M Todd King
- Laboratory of Metabolic Control, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - Robert Pawlosky
- Laboratory of Metabolic Control, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - J Nicholas P Rawlins
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Damian J Tyler
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Jeremy Robertson
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Richard L Veech
- Laboratory of Metabolic Control, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - Kieran Clarke
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Cook GA, Lavrentyev EN, Pham K, Park EA. Streptozotocin diabetes increases mRNA expression of ketogenic enzymes in the rat heart. Biochim Biophys Acta Gen Subj 2016; 1861:307-312. [PMID: 27845231 DOI: 10.1016/j.bbagen.2016.11.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Diabetic cardiomyopathy develops in insulin-dependent diabetic patients who have no hypertension, cardiac hypertrophy or vascular disease. Diabetes increases cardiac fatty acid oxidation, but cardiac hypertrophy limits fatty acid oxidation. Here we examined effects of diabetes on gene expression in rat hearts. METHODS We used oligonucleotide microarrays to examine effects of insulindependent diabetes in the rat heart. RTQ PCR confirmed results of microarrays. Specific antibodies were used to examine changes in the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2). RESULTS A surprising result of diabetes was increased mRNA encoding all enzymes of the ketone body synthesis pathway. Increased mRNA expression for these enzymes was confirmed by RTQ PCR. The mRNA encoding HMGCS2, the rate-controlling enzyme, was 27 times greater in diabetic hearts. Total HMGCS2 protein increased 8-fold in diabetic hearts, but no difference was found in HMGCS2 protein in control vs. diabetic liver. CONCLUSIONS Insulin-dependent diabetes induced the enzymes of ketone body synthesis in the heart, including HMGCS2, as well as increasing enzymes of fatty acid oxidation. GENERAL SIGNIFICANCE The mammalian heart does not export ketone bodies to other tissues, but rather is a major consumer of ketone bodies. Induction of HMGCS2, which is normally expressed only in the fetal and newborn heart, may indicate an adaptation by the heart to combat "metabolic inflexibility" by shifting the flux of excess intramitochondrial acetyl-CoA derived from elevated fatty acid oxidation into ketone bodies, liberating free CoA to balance the acetyl-CoA/CoA ratio in favor of increased glucose oxidation through the pyruvate dehydrogenase complex.
Collapse
Affiliation(s)
- George A Cook
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - Eduard N Lavrentyev
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Kevin Pham
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN 38104, United States
| | - Edwards A Park
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN 38104, United States
| |
Collapse
|
35
|
Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, King MT, Dodd MS, Holloway C, Neubauer S, Drawer S, Veech RL, Griffin JL, Clarke K. Nutritional Ketosis Alters Fuel Preference and Thereby Endurance Performance in Athletes. Cell Metab 2016; 24:256-68. [PMID: 27475046 DOI: 10.1016/j.cmet.2016.07.010] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/18/2016] [Accepted: 07/17/2016] [Indexed: 11/28/2022]
Abstract
Ketosis, the metabolic response to energy crisis, is a mechanism to sustain life by altering oxidative fuel selection. Often overlooked for its metabolic potential, ketosis is poorly understood outside of starvation or diabetic crisis. Thus, we studied the biochemical advantages of ketosis in humans using a ketone ester-based form of nutrition without the unwanted milieu of endogenous ketone body production by caloric or carbohydrate restriction. In five separate studies of 39 high-performance athletes, we show how this unique metabolic state improves physical endurance by altering fuel competition for oxidative respiration. Ketosis decreased muscle glycolysis and plasma lactate concentrations, while providing an alternative substrate for oxidative phosphorylation. Ketosis increased intramuscular triacylglycerol oxidation during exercise, even in the presence of normal muscle glycogen, co-ingested carbohydrate and elevated insulin. These findings may hold clues to greater human potential and a better understanding of fuel metabolism in health and disease.
Collapse
Affiliation(s)
- Pete J Cox
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; Department of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK.
| | - Tom Kirk
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Tom Ashmore
- Department of Biochemistry & Cambridge Systems Biology Centre, University of Cambridge & MRC Human Nutrition Research, Cambridge CB1 9NL, UK
| | - Kristof Willerton
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Rhys Evans
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Alan Smith
- UK Sport, 40 Bernard Street, London WC1N 1ST, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Brianna Stubbs
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - James West
- Department of Biochemistry & Cambridge Systems Biology Centre, University of Cambridge & MRC Human Nutrition Research, Cambridge CB1 9NL, UK
| | - Stewart W McLure
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - M Todd King
- Laboratory of Metabolic Control, NIAAA/NIH, Rockville, MD 20852, USA
| | - Michael S Dodd
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Cameron Holloway
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; Department of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Stefan Neubauer
- Department of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Scott Drawer
- UK Sport, 40 Bernard Street, London WC1N 1ST, UK
| | - Richard L Veech
- Laboratory of Metabolic Control, NIAAA/NIH, Rockville, MD 20852, USA
| | - Julian L Griffin
- Department of Biochemistry & Cambridge Systems Biology Centre, University of Cambridge & MRC Human Nutrition Research, Cambridge CB1 9NL, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
36
|
Ferrannini E, Mark M, Mayoux E. CV Protection in the EMPA-REG OUTCOME Trial: A "Thrifty Substrate" Hypothesis. Diabetes Care 2016; 39:1108-14. [PMID: 27289126 DOI: 10.2337/dc16-0330] [Citation(s) in RCA: 721] [Impact Index Per Article: 80.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/28/2016] [Indexed: 02/03/2023]
Abstract
The striking and unexpected relative risk reductions in cardiovascular (CV) mortality (38%), hospitalization for heart failure (35%), and death from any cause (32%) observed in the EMPA-REG OUTCOME trial using an inhibitor of sodium-glucose cotransporter 2 (SGLT2) in patients with type 2 diabetes and high CV risk have raised the possibility that mechanisms other than those observed in the trial-modest improvement in glycemic control, small decrease in body weight, and persistent reductions in blood pressure and uric acid level-may be at play. We hypothesize that under conditions of mild, persistent hyperketonemia, such as those that prevail during treatment with SGLT2 inhibitors, β-hydroxybutyrate is freely taken up by the heart (among other organs) and oxidized in preference to fatty acids. This fuel selection improves the transduction of oxygen consumption into work efficiency at the mitochondrial level. In addition, the hemoconcentration that typically follows SGLT2 inhibition enhances oxygen release to the tissues, thereby establishing a powerful synergy with the metabolic substrate shift. These mechanisms would cooperate with other SGLT2 inhibition-induced changes (chiefly, enhanced diuresis and reduced blood pressure) to achieve the degree of cardioprotection revealed in the EMPA-REG OUTCOME trial. This hypothesis opens up new lines of investigation into the pathogenesis and treatment of diabetic and nondiabetic heart disease.
Collapse
Affiliation(s)
| | - Michael Mark
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Eric Mayoux
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
37
|
Pedersen BA, Yazdi PG, Taylor JF, Khattab OS, Chen YH, Chen Y, Wang PH. Novel remodeling of the mouse heart mitochondrial proteome in response to acute insulin stimulation. Nutr Metab Cardiovasc Dis 2015; 25:1152-1158. [PMID: 26610654 PMCID: PMC4684478 DOI: 10.1016/j.numecd.2015.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 09/25/2015] [Accepted: 09/30/2015] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND AIM Mitochondrial dysfunction contributes to the pathophysiology of diabetic cardiomyopathy. The aim of this study was to investigate the acute changes in the mitochondrial proteome in response to insulin stimulation. METHODS AND RESULTS Cardiac mitochondria from C57BL/6 mice after insulin stimulation were analyzed using two-dimensional fluorescence difference gel electrophoresis. MALDI-TOF MS/MS was utilized to identify differences. Two enzymes involved in metabolism and four structural proteins were identified. Succinyl-CoA ligase [ADP forming] subunit beta was identified as one of the differentially regulated proteins. Upon insulin stimulation, a relatively more acidic isoform of this protein was increased by 53% and its functional activity was decreased by ∼32%. CONCLUSIONS This proteomic remodeling in response to insulin stimulation may play an important role in the normal and diabetic heart.
Collapse
Affiliation(s)
- B A Pedersen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - P G Yazdi
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA; Systomic Health LLC, Los Angeles, CA 90039, USA
| | - J F Taylor
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - O S Khattab
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
| | - Y-H Chen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Y Chen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - P H Wang
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA; Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California at Irvine, Irvine, CA 92697, USA; Department of Physiology & Biophysics, University of California at Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
38
|
Bouteldja N, Andersen LT, Møller N, Gormsen LC. Using positron emission tomography to study human ketone body metabolism: a review. Metabolism 2014; 63:1375-84. [PMID: 25195069 DOI: 10.1016/j.metabol.2014.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/14/2014] [Accepted: 08/02/2014] [Indexed: 11/25/2022]
Abstract
Ketone bodies - 3-hydroxybutyrate and acetoacetate - are important fuel substrates, which can be oxidized by most tissues in the body. They are synthesized in the liver and are derived from fatty acids released from adipose tissue. Intriguingly, under conditions of stress such as fasting, arterio-venous catheterization studies have shown that the brain switches from the use of almost 100% glucose to the use of >50-60% ketone bodies. A similar adaptive mechanism is observed in the heart, where fasting induces a shift toward ketone body uptake that provides the myocardium with an alternate fuel source and also favorably affects myocardial contractility. Within the past years there has been a renewed interest in ketone bodies and the possible beneficial effects of fasting/semi-fasting/exercising and other "ketogenic" regimens have received much attention. In this perspective, it is promising that positron emission tomography (PET) techniques with isotopically labeled ketone bodies, fatty acids and glucose offer an opportunity to study interactions between ketone body, fatty acid and glucose metabolism in tissues such as the brain and heart. PET scans are non-invasive and thus eliminates the need to place catheters in vascular territories not easily accessible. The short half-life of e.g. 11C-labeled PET tracers even allows multiple scans on the same study day and reduces the total radiation burden associated with the procedure. This short review aims to give an overview of current knowledge on ketone body metabolism obtained by PET studies and discusses the methodological challenges and perspectives involved in PET ketone body research.
Collapse
Affiliation(s)
- Nadia Bouteldja
- Department of Radiology, Hospital of Southwest Denmark, 6700 Esbjerg, Denmark
| | - Lone Thing Andersen
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - Niels Møller
- Department of Endocrinology, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - Lars Christian Gormsen
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
39
|
Cox PJ, Clarke K. Acute nutritional ketosis: implications for exercise performance and metabolism. EXTREME PHYSIOLOGY & MEDICINE 2014; 3:17. [PMID: 25379174 PMCID: PMC4212585 DOI: 10.1186/2046-7648-3-17] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/29/2014] [Indexed: 01/13/2023]
Abstract
Ketone bodies acetoacetate (AcAc) and D-β-hydroxybutyrate (βHB) may provide an alternative carbon source to fuel exercise when delivered acutely in nutritional form. The metabolic actions of ketone bodies are based on sound evolutionary principles to prolong survival during caloric deprivation. By harnessing the potential of these metabolic actions during exercise, athletic performance could be influenced, providing a useful model for the application of ketosis in therapeutic conditions. This article examines the energetic implications of ketone body utilisation with particular reference to exercise metabolism and substrate energetics.
Collapse
Affiliation(s)
- Pete J Cox
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- Department of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
40
|
Abstract
Ketosis induced by starvation or feeding a ketogenic diet has widespread and often contradictory effects due to the simultaneous elevation of both ketone bodies and free fatty acids. The elevation of ketone bodies increases the energy of ATP hydrolysis by reducing the mitochondrial NAD couple and oxidizing the coenzyme Q couple, thus increasing the redox span between site I and site II. In contrast, metabolism of fatty acids leads to a reduction of both mitochondrial NAD and mitochondrial coenzyme Q causing a decrease in the ΔG of ATP hydrolysis. In contrast, feeding ketone body esters leads to pure ketosis, unaccompanied by elevation of free fatty acids, producing a physiological state not previously seen in nature. The effects of pure ketosis on transcription and upon certain neurodegenerative diseases make approach not only interesting, but of potential therapeutic value.
Collapse
Affiliation(s)
- Richard L Veech
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
41
|
Martinez-Outschoorn U, Sotgia F, Lisanti MP. Tumor microenvironment and metabolic synergy in breast cancers: critical importance of mitochondrial fuels and function. Semin Oncol 2014; 41:195-216. [PMID: 24787293 DOI: 10.1053/j.seminoncol.2014.03.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metabolic synergy or metabolic coupling between glycolytic stromal cells (Warburg effect) and oxidative cancer cells occurs in human breast cancers and promotes tumor growth. The Warburg effect or aerobic glycolysis is the catabolism of glucose to lactate to obtain adenosine triphosphate (ATP). This review summarizes the main findings on this stromal metabolic phenotype, and the associated signaling pathways, as well as the critical role of oxidative stress and autophagy, all of which promote carcinoma cell mitochondrial metabolism and tumor growth. Loss of Caveolin 1 (Cav-1) and the upregulation of monocarboxylate transporter 4 (MCT4) in stromal cells are novel markers of the Warburg effect and metabolic synergy between stromal and carcinoma cells. MCT4 and Cav-1 are also breast cancer prognostic biomarkers. Reactive oxygen species (ROS) are key mediators of the stromal Warburg effect. High ROS also favors cancer cell mitochondrial metabolism and tumorigenesis, and anti-oxidants can reverse this altered stromal and carcinoma metabolism. A pseudo-hypoxic state with glycolysis and low mitochondrial metabolism in the absence of hypoxia is a common feature in breast cancer. High ROS induces loss of Cav-1 in stromal cells and is sufficient to generate a pseudo-hypoxic state. Loss of Cav-1 in the stroma drives glycolysis and lactate extrusion via HIF-1α stabilization and the upregulation of MCT4. Stromal cells with loss of Cav-1 and/or high expression of MCT4 also show a catabolic phenotype, with enhanced macroautophagy. This catabolic state in stromal cells is driven by hypoxia-inducible factor (HIF)-1α, nuclear factor κB (NFκB), and JNK activation and high ROS generation. A feed-forward loop in stromal cells regulates pseudo-hypoxia and metabolic synergy, with Cav-1, MCT4, HIF-1α, NFκB, and ROS as its key elements. Metabolic synergy also may occur between cancer cells and cells in distant organs from the tumor. Cancer cachexia, which is due to severe organismal metabolic dysregulation in myocytes and adipocytes, shares similarities with stromal-carcinoma metabolic synergy, as well. In summary, metabolic synergy occurs when breast carcinoma cells induce a nutrient-rich microenvironment to promote tumor growth. The process of tumor metabolic synergy is a multistep process, due to the generation of ROS, and the induction of catabolism with autophagy, mitophagy and glycolysis. Studying epithelial-stromal interactions and metabolic synergy is important to better understand the ecology of cancer and the metabolic role of different cell types in tumor progression.
Collapse
Affiliation(s)
| | - Federica Sotgia
- University of Manchester, Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Manchester, United Kingdom
| | - Michael P Lisanti
- University of Manchester, Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Manchester, United Kingdom
| |
Collapse
|
42
|
Martinez-Outschoorn UE, Lisanti MP, Sotgia F. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol 2014; 25:47-60. [PMID: 24486645 DOI: 10.1016/j.semcancer.2014.01.005] [Citation(s) in RCA: 319] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/17/2014] [Accepted: 01/17/2014] [Indexed: 12/22/2022]
Abstract
Fibroblasts are the most abundant "non-cancerous" cells in tumors. However, it remains largely unknown how these cancer-associated fibroblasts (CAFs) promote tumor growth and metastasis, driving chemotherapy resistance and poor clinical outcome. This review summarizes new findings on CAF signaling pathways and their emerging metabolic phenotypes that promote tumor growth. Although it is well established that altered cancer metabolism enhances tumor growth, little is known about the role of fibroblast metabolism in tumor growth. New studies reveal that metabolic coupling occurs between catabolic fibroblasts and anabolic cancer cells, in many types of human tumors, including breast, prostate, and head & neck cancers, as well as lymphomas. These catabolic phenotypes observed in CAFs are secondary to a ROS-induced metabolic stress response. Mechanistically, this occurs via HIF1-alpha and NFκB signaling, driving oxidative stress, autophagy, glycolysis and senescence in stromal fibroblasts. These catabolic CAFs then create a nutrient-rich microenvironment, to metabolically support tumor growth, via the local stromal generation of mitochondrial fuels (lactate, ketone bodies, fatty acids, glutamine, and other amino acids). New biomarkers of this catabolic CAF phenotype (such as caveolin-1 (Cav-1) and MCT4), which are reversible upon treatment with anti-oxidants, are strong predictors of poor clinical outcome in various types of human cancers. How cancer cells metabolically reprogram fibroblasts can also help us to understand the effects of cancer cells at an organismal level, explaining para-neoplastic phenomena, such as cancer cachexia. In conclusion, cancer should be viewed more as a systemic disease, that engages the host-organism in various forms of energy-transfer and metabolic co-operation, across a whole-body "ecosystem".
Collapse
Affiliation(s)
| | - Michael P Lisanti
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, UK.
| | - Federica Sotgia
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, UK.
| |
Collapse
|
43
|
Veech RL. Ketone esters increase brown fat in mice and overcome insulin resistance in other tissues in the rat. Ann N Y Acad Sci 2013; 1302:42-48. [PMID: 23909803 DOI: 10.1111/nyas.12222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Brown adipose tissue (BAT) is classically activated by sympathetic nervous stimulation resulting from exposure to cold. Feeding a high-fat diet also induces development of brown fat, but is decreased by caloric restriction. Blood ketone bodies, which function as alternative energy substrates to glucose, are increased during caloric restriction. Here we discuss the unexpected observation that feeding an ester of ketone bodies to the mouse, which increases blood ketone body concentrations, results in an activation of brown fat. The mechanism of this activation of brown fat is similar to that occurring from cold exposure in that cyclic adenosine monophosphate (AMP) levels are increased as are levels of the transcription factor cyclic AMP-responsive element-binding protein, which is also increased by ketone ester feeding. Other effects of feeding ketone esters, in addition to their ability to induce brown fat, are discussed such as their ability to overcome certain aspects of insulin resistance and to ameliorate the accumulation of amyloid and phosphorylated tau protein in brain, and improve cognitive function, in a triple transgenic mouse model of Alzheimer's disease.
Collapse
Affiliation(s)
- Richard L Veech
- Laboratory of Metabolic Control, Department of Health and Human Services, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
44
|
Rider OJ, Cox P, Tyler D, Clarke K, Neubauer S. Myocardial substrate metabolism in obesity. Int J Obes (Lond) 2013; 37:972-9. [PMID: 23069666 DOI: 10.1038/ijo.2012.170] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/29/2012] [Accepted: 09/02/2012] [Indexed: 12/28/2022]
Abstract
Obesity is linked to a wide variety of cardiac changes, from subclinical diastolic dysfunction to end-stage systolic heart failure. Obesity causes changes in cardiac metabolism, which make ATP production and utilization less efficient, producing functional consequences that are linked to the increased rate of heart failure in this population. As a result of the increases in circulating fatty acids and insulin resistance that accompanies excess fat storage, several of the proteins and genes that are responsible for fatty acid uptake and metabolism are upregulated, and the metabolic machinery responsible for glucose utilization and oxidation are inhibited. The resultant increase in fatty acid metabolism, and the inherent alterations in the proteins of the electron transport chain used to create the gradient needed to drive mitochondrial ATP production, results in a decrease in efficiency of cardiac work and a relative increase in oxygen usage. These changes in cardiac mitochondrial metabolism are potential therapeutic targets for the treatment and prevention of obesity-related heart failure.
Collapse
Affiliation(s)
- O J Rider
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | | | | | | |
Collapse
|
45
|
Kashiwaya Y, Bergman C, Lee JH, Wan R, King MT, Mughal MR, Okun E, Clarke K, Mattson MP, Veech RL. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer's disease. Neurobiol Aging 2012; 34:1530-9. [PMID: 23276384 DOI: 10.1016/j.neurobiolaging.2012.11.023] [Citation(s) in RCA: 252] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) involves progressive accumulation of amyloid β-peptide (Aβ) and neurofibrillary pathologies, and glucose hypometabolism in brain regions critical for memory. The 3xTgAD mouse model was used to test the hypothesis that a ketone ester-based diet can ameliorate AD pathogenesis. Beginning at a presymptomatic age, 2 groups of male 3xTgAD mice were fed a diet containing a physiological enantiomeric precursor of ketone bodies (KET) or an isocaloric carbohydrate diet. The results of behavioral tests performed at 4 and 7 months after diet initiation revealed that KET-fed mice exhibited significantly less anxiety in 2 different tests. 3xTgAD mice on the KET diet also exhibited significant, albeit relatively subtle, improvements in performance on learning and memory tests. Immunohistochemical analyses revealed that KET-fed mice exhibited decreased Aβ deposition in the subiculum, CA1 and CA3 regions of the hippocampus, and the amygdala. KET-fed mice exhibited reduced levels of hyperphosphorylated tau deposition in the same regions of the hippocampus, amygdala, and cortex. Thus, a novel ketone ester can ameliorate proteopathic and behavioral deficits in a mouse AD model.
Collapse
Affiliation(s)
- Yoshihiro Kashiwaya
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kabiraj P, Pal R, Varela-Ramirez A, Miranda M, Narayan M. Nitrosative stress mediated misfolded protein aggregation mitigated by Na-d-β-hydroxybutyrate intervention. Biochem Biophys Res Commun 2012; 426:438-44. [DOI: 10.1016/j.bbrc.2012.08.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 08/25/2012] [Indexed: 10/27/2022]
|
47
|
The unrecognized effects of the volume and composition of the resuscitation fluid used during the administration of blood products. Transfus Apher Sci 2012; 46:121-3. [PMID: 22364841 DOI: 10.1016/j.transci.2012.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Recent publications have reported the severe adverse events associated with blood products but have not considered the effect of the volume and composition of the resuscitative fluids infused with the blood products. METHODS Injury leads to cellular reaction characterized by insulin resistance during which glucose cannot enter muscle and fat cells. In all cells, mitochondrial pyruvate dehydrogenase activity is decreased during insulin deficiency leaving cells deficient in substrates needed to power the Krebs cycle and make ATP. RESULTS d-β-Hydroxybutyrate, a normal ketone body metabolite, enters cells on the monocarboxylate transport mimicking the action of insulin and bypassing the enzymatic block at PDH. Metabolism of ketone bodies increases efficiency of mitochondrial energy production and cellular ATP level. CONCLUSION Infusion of 250 ml of 600 mM Na d-β-hydroxybutyrate solution, with the same osmotic strength as the hypertonic NaCl solution currently being used, would correct insulin resistance, provide energy substrates for cells to produce ATP, correct the tendency of injured tissue to swell due to decreased energy of ionic gradients and correct acidosis observed in hemorrhage.
Collapse
|
48
|
Metabolomic analysis of serum by (1) H NMR spectroscopy in amyotrophic lateral sclerosis. Clin Chim Acta 2010; 411:563-7. [PMID: 20096678 DOI: 10.1016/j.cca.2010.01.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/11/2010] [Accepted: 01/11/2010] [Indexed: 12/13/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS), an invariably fatal neurological disorder shows complicated pathogenesis that poses challenges with respect to diagnosis as well as monitoring of disease progression. METHODS We investigated metabolite profiles in the serum of 30 patients with ALS, 10 patients of Hirayama disease, which served as a neurological disease control and 25 healthy controls by using (1) H NMR spectroscopy. RESULTS Compared to healthy controls, the ALS patients had higher quantities of glutamate (P<0.001), beta-hydroxybutyrate (P<0.001), acetate (P<0.01), acetone (P<0.05), and formate (P<0.001), and lower concentrations of glutamine (P<0.02), histidine (P<0.001) and N-acetyl derivatives. On the other hand, Hirayama disease patients had significantly higher median concentrations of pyruvate (P<0.05), glutamate (P<0.001), formate (P<0.05) and lower median concentrations of N-acetyl derivatives. Furthermore, we also found that serum glutamate showed a positive correlation (P<0.001, r=0.6487) whereas, histidine showed a negative correlation (P<0.001, r=-0.5641) with the duration of the disease in ALS. CONCLUSIONS Such (1) H NMR study of serum may reveal abnormal metabolite patterns, which could have the potential to serve as surrogate markers for monitoring ALS disease progression.
Collapse
|
49
|
Pumain R, Ahmed MS, Kurcewicz I, Trottier S, Louvel J, Turak B, Devaux B, Laschet J. Lability of GABAAreceptor function in human partial epilepsy: Possible relationship to hypometabolism. Epilepsia 2008; 49 Suppl 8:87-90. [DOI: 10.1111/j.1528-1167.2008.01845.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Briede J, Stivrina M, Vigante B, Stoldere D, Duburs G. Acute effect of antidiabetic 1,4-dihydropyridine compound cerebrocrast on cardiac function and glucose metabolism in the isolated, perfused normal rat heart. Cell Biochem Funct 2008; 26:238-45. [PMID: 17990288 DOI: 10.1002/cbf.1442] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Diabetes mellitus (DM) is an important cardiovascular risk factor and is associated with abnormalities in endothelial and vascular smooth muscle cell function, evoked by chronic hyperglycemia and hyperlipidemia. Chronic insulin deficiency or resistance is marked by decreases in the intensity of glucose transport, glucose phosphorylation, and glucose oxidation, plus decreases in ATP levels in cardiac myocytes. It is important to search for new agents that promote glucose consumption in the heart and partially inhibit extensive fatty acid beta-oxidation observed in diabetic, ischemia. When the oxygen supply for myocardium is decreased, the heart accumulates potentially toxic intermediates of fatty acid beta-oxidation, that is, long-chain acylcarnitine and long-chain acyl-CoA metabolites. Exogenous glucose and heart glycogen become an important compensatory source of energy. Therefore we studied the effect of the antidiabetic 1,4-dihydropyridine compound cerebrocrast at concentrations from 10(-10) M to 10(-7) M on isolated rat hearts using the method of Langendorff, on physiological parameters and energy metabolism. Cerebrocrast at concentrations from 10(-10) M to 10(-7) M has a negative inotropic effect on the rat heart. It inhibits L-type Ca(2+)channels thereby diminishing the cellular Ca(2+) supply, reducing contractile activity, and oxygen consumption, that normally favors enhanced glucose uptake, metabolism, and production of high-energy phosphates (ATP content) in myocardium. Cerebrocrast decreases heart rate and left ventricular (LV) systolic pressure; at concentrations of 10(-10) M and 10(-9) M it evokes short-term vasodilatation of coronary arteries. Increase of ATP content in the myocytes induced by cerebrocrast has a ubiquitous role. It can preserve the integrity of the cell plasma membranes, maintain normal cellular function, and inhibit release of lactate dehydrogenase (LDH) from cells that is associated with diabetes and heart ischemia. Administration of cerebrocrast together with insulin shows that both compounds only slightly enhance glucose uptake in myocardium, but significantly normalize the rate of contraction and relaxation ( +/- dp/dt). The effect of insulin on coronary flow is more pronounced by administration of insulin together with cerebrocrast at a concentration of 10(-7) M. Cerebrocrast may promote a shift of glucose consumption from aerobic to anerobic conditions (through the negative inotropic properties), and may be very significant in prevention of cardiac ischemic episodes.
Collapse
Affiliation(s)
- Janina Briede
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | | | | | | |
Collapse
|