851
|
Yang H, Shan W, Zhu F, Wu J, Wang Q. Ketone Bodies in Neurological Diseases: Focus on Neuroprotection and Underlying Mechanisms. Front Neurol 2019; 10:585. [PMID: 31244753 PMCID: PMC6581710 DOI: 10.3389/fneur.2019.00585] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
There is growing evidence that ketone bodies, which are derived from fatty acid oxidation and usually produced in fasting state or on high-fat diets have broad neuroprotective effects. Although the mechanisms underlying the neuroprotective effects of ketone bodies have not yet been fully elucidated, studies in recent years provided abundant shreds of evidence that ketone bodies exert neuroprotective effects through possible mechanisms of anti-oxidative stress, maintaining energy supply, modulating the activity of deacetylation and inflammatory responses. Based on the neuroprotective effects, the ketogenic diet has been used in the treatment of several neurological diseases such as refractory epilepsy, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. The ketogenic diet has great potential clinically, which should be further explored in future studies. It is necessary to specify the roles of components in ketone bodies and their therapeutic targets and related pathways to optimize the strategy and efficacy of ketogenic diet therapy in the future.
Collapse
Affiliation(s)
- Huajun Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China
| | - Wei Shan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fei Zhu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China
| | - Jianping Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
852
|
Cappel DA, Deja S, Duarte JAG, Kucejova B, Iñigo M, Fletcher JA, Fu X, Berglund ED, Liu T, Elmquist JK, Hammer S, Mishra P, Browning JD, Burgess SC. Pyruvate-Carboxylase-Mediated Anaplerosis Promotes Antioxidant Capacity by Sustaining TCA Cycle and Redox Metabolism in Liver. Cell Metab 2019; 29:1291-1305.e8. [PMID: 31006591 PMCID: PMC6585968 DOI: 10.1016/j.cmet.2019.03.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/12/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
The hepatic TCA cycle supports oxidative and biosynthetic metabolism. This dual responsibility requires anaplerotic pathways, such as pyruvate carboxylase (PC), to generate TCA cycle intermediates necessary for biosynthesis without disrupting oxidative metabolism. Liver-specific PC knockout (LPCKO) mice were created to test the role of anaplerotic flux in liver metabolism. LPCKO mice have impaired hepatic anaplerosis, diminution of TCA cycle intermediates, suppressed gluconeogenesis, reduced TCA cycle flux, and a compensatory increase in ketogenesis and renal gluconeogenesis. Loss of PC depleted aspartate and compromised urea cycle function, causing elevated urea cycle intermediates and hyperammonemia. Loss of PC prevented diet-induced hyperglycemia and insulin resistance but depleted NADPH and glutathione, which exacerbated oxidative stress and correlated with elevated liver inflammation. Thus, despite catalyzing the synthesis of intermediates also produced by other anaplerotic pathways, PC is specifically necessary for maintaining oxidation, biosynthesis, and pathways distal to the TCA cycle, such as antioxidant defenses.
Collapse
Affiliation(s)
- David A Cappel
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stanisław Deja
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - João A G Duarte
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Blanka Kucejova
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melissa Iñigo
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin A Fletcher
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaorong Fu
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric D Berglund
- Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tiemin Liu
- Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, Shanghai 200438, China
| | - Joel K Elmquist
- Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Suntrea Hammer
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shawn C Burgess
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
853
|
Poffé C, Ramaekers M, Van Thienen R, Hespel P. Ketone ester supplementation blunts overreaching symptoms during endurance training overload. J Physiol 2019; 597:3009-3027. [PMID: 31039280 PMCID: PMC6851819 DOI: 10.1113/jp277831] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Overload training is required for sustained performance gain in athletes (functional overreaching). However, excess overload may result in a catabolic state which causes performance decrements for weeks (non-functional overreaching) up to months (overtraining). Blood ketone bodies can attenuate training- or fasting-induced catabolic events. Therefore, we investigated whether increasing blood ketone levels by oral ketone ester (KE) intake can protect against endurance training-induced overreaching. We show for the first time that KE intake following exercise markedly blunts the development of physiological symptoms indicating overreaching, and at the same time significantly enhances endurance exercise performance. We provide preliminary data to indicate that growth differentiation factor 15 (GDF15) may be a relevant hormonal marker to diagnose the development of overtraining. Collectively, our data indicate that ketone ester intake is a potent nutritional strategy to prevent the development of non-functional overreaching and to stimulate endurance exercise performance. ABSTRACT It is well known that elevated blood ketones attenuate net muscle protein breakdown, as well as negate catabolic events, during energy deficit. Therefore, we hypothesized that oral ketones can blunt endurance training-induced overreaching. Fit male subjects participated in two daily training sessions (3 weeks, 6 days/week) while receiving either a ketone ester (KE, n = 9) or a control drink (CON, n = 9) following each session. Sustainable training load in week 3 as well as power output in the final 30 min of a 2-h standardized endurance session were 15% higher in KE than in CON (both P < 0.05). KE inhibited the training-induced increase in nocturnal adrenaline (P < 0.01) and noradrenaline (P < 0.01) excretion, as well as blunted the decrease in resting (CON: -6 ± 2 bpm; KE: +2 ± 3 bpm, P < 0.05), submaximal (CON: -15 ± 3 bpm; KE: -7 ± 2 bpm, P < 0.05) and maximal (CON: -17 ± 2 bpm; KE: -10 ± 2 bpm, P < 0.01) heart rate. Energy balance during the training period spontaneously turned negative in CON (-2135 kJ/day), but not in KE (+198 kJ/day). The training consistently increased growth differentiation factor 15 (GDF15), but ∼2-fold more in CON than in KE (P < 0.05). In addition, delta GDF15 correlated with the training-induced drop in maximal heart rate (r = 0.60, P < 0.001) and decrease in osteocalcin (r = 0.61, P < 0.01). Other measurements such as blood ACTH, cortisol, IL-6, leptin, ghrelin and lymphocyte count, and muscle glycogen content did not differentiate KE from CON. In conclusion, KE during strenuous endurance training attenuates the development of overreaching. We also identify GDF15 as a possible marker of overtraining.
Collapse
Affiliation(s)
- Chiel Poffé
- Exercise Physiology Research GroupDepartment of Movement SciencesKU LeuvenLeuvenBelgium
| | - Monique Ramaekers
- Exercise Physiology Research GroupDepartment of Movement SciencesKU LeuvenLeuvenBelgium
| | - Ruud Van Thienen
- Exercise Physiology Research GroupDepartment of Movement SciencesKU LeuvenLeuvenBelgium
| | - Peter Hespel
- Exercise Physiology Research GroupDepartment of Movement SciencesKU LeuvenLeuvenBelgium
- Bakala Academy‐Athletic Performance CenterKU LeuvenLeuvenBelgium
| |
Collapse
|
854
|
Wood TR, Stubbs BJ, Juul SE. Exogenous Ketone Bodies as Promising Neuroprotective Agents for Developmental Brain Injury. Dev Neurosci 2019; 40:451-462. [PMID: 31085911 DOI: 10.1159/000499563] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/12/2019] [Indexed: 11/19/2022] Open
Abstract
Ketone bodies are a promising area of neuroprotection research that may be ideally suited to the injured newborn. During normal development, the human infant is in significant ketosis for at least the first week of life. Ketone uptake and metabolism is upregulated in the both the fetus and neonate, with ketone bodies providing at least 10% of cerebral metabolic energy requirements, as well as being the preferred precursors for the synthesis of fatty acids and cholesterol. At the same time, ketone bodies have been shown to have multiple neuroprotective effects, including being anticonvulsant, decreasing oxidative stress and inflammation, and epigenetically upregulating the production of neurotrophic factors. While ketogenic diets and exogenous ketosis are largely being investigated in the setting of adult brain injury, the adaptation of the neonate to ketosis suggests that developmental brain injury may be the area most suited to the use of ketones for neuroprotection. Here, we describe the mechanisms by which ketone bodies exert their neuroprotective effects, and how these may translate to benefits within each of the phases of neonatal asphyxial brain injury.
Collapse
Affiliation(s)
- Thomas R Wood
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA, .,Institute for Human and Machine Cognition, Pensacola, Florida, USA,
| | - Brianna J Stubbs
- HVMN Inc., San Francisco, California, USA.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
855
|
Zhang Z, Hunter L, Wu G, Maidstone R, Mizoro Y, Vonslow R, Fife M, Hopwood T, Begley N, Saer B, Wang P, Cunningham P, Baxter M, Durrington H, Blaikley JF, Hussell T, Rattray M, Hogenesch JB, Gibbs J, Ray DW, Loudon ASI. Genome-wide effect of pulmonary airway epithelial cell-specific Bmal1 deletion. FASEB J 2019; 33:6226-6238. [PMID: 30794439 PMCID: PMC6463917 DOI: 10.1096/fj.201801682r] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
Pulmonary airway epithelial cells (AECs) form a critical interface between host and environment. We investigated the role of the circadian clock using mice bearing targeted deletion of the circadian gene brain and muscle ARNT-like 1 (Bmal1) in AECs. Pulmonary neutrophil infiltration, biomechanical function, and responses to influenza infection were all disrupted. A circadian time-series RNA sequencing study of laser-captured AECs revealed widespread disruption in genes of the core circadian clock and output pathways regulating cell metabolism (lipids and xenobiotics), extracellular matrix, and chemokine signaling, but strikingly also the gain of a novel rhythmic transcriptome in Bmal1-targeted cells. Many of the rhythmic components were replicated in primary AECs cultured in air-liquid interface, indicating significant cell autonomy for control of pulmonary circadian physiology. Finally, we found that metabolic cues dictate phasing of the pulmonary clock and circadian responses to immunologic challenges. Thus, the local circadian clock in AECs is vital in lung health by coordinating major cell processes such as metabolism and immunity.-Zhang, Z. Hunter, L., Wu, G., Maidstone, R., Mizoro, Y., Vonslow, R., Fife, M., Hopwood, T., Begley, N., Saer, B., Wang, P., Cunningham, P., Baxter, M., Durrington, H., Blaikley, J. F., Hussell, T., Rattray, M., Hogenesch, J. B., Gibbs, J., Ray, D. W., Loudon, A. S. I. Genome-wide effect of pulmonary airway epithelial cell-specific Bmal1 deletion.
Collapse
Affiliation(s)
- Zhenguang Zhang
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Louise Hunter
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Gang Wu
- Division of Human Genetics, Department of Pediatrics, Center for Chronobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Immunobiology, Department of Pediatrics, Center for Chronobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Robert Maidstone
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom; and
| | - Yasutaka Mizoro
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Ryan Vonslow
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Mark Fife
- Manchester Center for Collaborative Inflammation Research, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, United Kingdom
| | - Thomas Hopwood
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Nicola Begley
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Ben Saer
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Ping Wang
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom; and
| | - Peter Cunningham
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Matthew Baxter
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Hannah Durrington
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - John F. Blaikley
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Center for Collaborative Inflammation Research, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, United Kingdom
| | - Magnus Rattray
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom; and
| | - John B. Hogenesch
- Division of Human Genetics, Department of Pediatrics, Center for Chronobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Julie Gibbs
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - David W. Ray
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| | - Andrew S. I. Loudon
- Centre for Biological Timing, Faculty of Biology, Health, and Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
856
|
Antoniou SX, Gaude E, Ruparel M, van der Schee MP, Janes SM, Rintoul RC. The potential of breath analysis to improve outcome for patients with lung cancer. J Breath Res 2019; 13:034002. [PMID: 30822771 DOI: 10.1088/1752-7163/ab0bee] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung cancer remains the most common cause of cancer related death in both the UK and USA. Development of diagnostic approaches that have the ability to detect lung cancer early are a research priority with potential to improve survival. Analysis of exhaled breath metabolites, or volatile organic compounds (VOCs) is an area of considerable interest as it could fulfil such requirements. Numerous studies have shown that VOC profiles are different in the breath of patients with lung cancer compared to healthy individuals or those with non-malignant lung diseases. This review provides a scientific and clinical assessment of the potential value of a breath test in lung cancer. It discusses the current understanding of metabolic pathways that contribute to exhaled VOC production in lung cancer and reviews the research conducted to date. Finally, we highlight important areas for future research and discuss how a breath test could be incorporated into various clinical pathways.
Collapse
Affiliation(s)
- S X Antoniou
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom.,Equal contribution
| | - E Gaude
- Owlstone Medical, Cambridge, United Kingdom,Equal contribution
| | - M Ruparel
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | | | - S M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - R C Rintoul
- Papworth Trials Unit Collaboration, Royal Papworth Hospital, Cambridge, United Kingdom,Department of Oncology, University of Cambridge, United Kingdom
| | | |
Collapse
|
857
|
Paolicelli RC, Angiari S. Microglia immunometabolism: From metabolic disorders to single cell metabolism. Semin Cell Dev Biol 2019; 94:129-137. [PMID: 30954657 DOI: 10.1016/j.semcdb.2019.03.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/01/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022]
Abstract
Since the observation that obesity-associated low-grade chronic inflammation is a crucial driver for the onset of systemic metabolic disorders such as type 2 diabetes, a number of studies have highlighted the role of both the innate and the adaptive immune system in such pathologies. Moreover, researchers have recently demonstrated that immune cells can modulate their intracellular metabolic profile to control their activation and effector functions. These discoveries represent the foundations of a research area known as "immunometabolism", an emerging field of investigation that may lead to the development of new-generation therapies for the treatment of inflammatory and metabolic diseases. Most of the studies in the field have focused their attention on both circulating white blood cells and leukocytes residing within metabolic tissues such as adipose tissue, liver and pancreas. However, immunometabolism of immune cells in non-metabolic tissues, including central nervous system microglia, have long been neglected. In this review, we highlight the most recent findings suggesting that microglial cells play a central role in metabolic disorders and that interfering with the metabolic profile of microglia can modulate their functionality and pathogenicity in neurological diseases.
Collapse
Affiliation(s)
- Rosa C Paolicelli
- Department of Physiology, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland.
| | - Stefano Angiari
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, D02 R590, Dublin, Ireland.
| |
Collapse
|
858
|
Gross EC, Klement RJ, Schoenen J, D'Agostino DP, Fischer D. Potential Protective Mechanisms of Ketone Bodies in Migraine Prevention. Nutrients 2019; 11:E811. [PMID: 30974836 PMCID: PMC6520671 DOI: 10.3390/nu11040811] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
An increasing amount of evidence suggests that migraines are a response to a cerebral energy deficiency or oxidative stress levels that exceed antioxidant capacity. The ketogenic diet (KD), a diet mimicking fasting that leads to the elevation of ketone bodies (KBs), is a therapeutic intervention targeting cerebral metabolism that has recently shown great promise in the prevention of migraines. KBs are an alternative fuel source for the brain, and are thus likely able to circumvent some of the abnormalities in glucose metabolism and transport found in migraines. Recent research has shown that KBs-D-β-hydroxybutyrate in particular-are more than metabolites. As signalling molecules, they have the potential to positively influence other pathways commonly believed to be part of migraine pathophysiology, namely: mitochondrial functioning, oxidative stress, cerebral excitability, inflammation and the gut microbiome. This review will describe the mechanisms by which the presence of KBs, D-BHB in particular, could influence those migraine pathophysiological mechanisms. To this end, common abnormalities in migraines are summarised with a particular focus on clinical data, including phenotypic, biochemical, genetic and therapeutic studies. Experimental animal data will be discussed to elaborate on the potential therapeutic mechanisms of elevated KBs in migraine pathophysiology, with a particular focus on the actions of D-BHB. In complex diseases such as migraines, a therapy that can target multiple possible pathogenic pathways seems advantageous. Further research is needed to establish whether the absence/restriction of dietary carbohydrates, the presence of KBs, or both, are of primary importance for the migraine protective effects of the KD.
Collapse
Affiliation(s)
- Elena C Gross
- Division of Paediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, 4056 Basel, Switzerland.
| | - Rainer J Klement
- Department of Radiation Oncology, Leopoldina Hospital Schweinfurt, 97422 Schweinfurt, Germany.
| | - Jean Schoenen
- Headache Research Unit, University of Liège, Dept of Neurology-Citadelle Hospital, 4000 Liège, Belgium.
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Metabolic Medicine Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA.
| | - Dirk Fischer
- Division of Paediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
859
|
Durkalec-Michalski K, Nowaczyk PM, Siedzik K. Effect of a four-week ketogenic diet on exercise metabolism in CrossFit-trained athletes. J Int Soc Sports Nutr 2019; 16:16. [PMID: 30953522 PMCID: PMC6451242 DOI: 10.1186/s12970-019-0284-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The ketogenic diet is becoming a popular nutritional model among athletes. However, the relationship between its use and metabolism during exercise seems to have not been fully investigated. METHODS The aim of the study was to assess the effects of a four-week ketogenic diet (KD) on fat and carbohydrate (CHO) utilization during an incremental cycling test (ICT) in CrossFit-trained female (n = 11) and male (n = 11) athletes. During the ICT (while consuming the customary diet and after the KD), oxygen uptake and carbon dioxide exhalation were registered, and CHO and fat utilization as well as energy expenditure were calculated. RESULTS In males, the KD led to an increase in fat utilization (g·min- 1·kgFFM- 1 and % oxidation). It was particularly noticeable at exercise intensities up to 80% of VO2max. An increase in the area under the curve (AUC) was seen in males but not in females at up to ≤65% VO2max of fat utilization. CONCLUSIONS Male CrossFit-trained athletes seem to be more prone to shifts in macronutrient utilization (in favor of fat utilization) during submaximal intensity exercise under a ketogenic diet than are female athletes. TRIAL REGISTRATION Clinical Trials Gov, NCT03665948 . Registered 11 September 2018 (retrospectively registered).
Collapse
Affiliation(s)
- Krzysztof Durkalec-Michalski
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland
- Department of Food and Nutrition, Poznan University of Physical Education, 61-871 Poznań, Poland
| | - Paulina M. Nowaczyk
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Katarzyna Siedzik
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland
| |
Collapse
|
860
|
Lévesque S, Pol JG, Ferrere G, Galluzzi L, Zitvogel L, Kroemer G. Trial watch: dietary interventions for cancer therapy. Oncoimmunology 2019; 8:1591878. [PMID: 31143510 PMCID: PMC6527263 DOI: 10.1080/2162402x.2019.1591878] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/30/2022] Open
Abstract
Dietary interventions have a profound impact on whole body metabolism, including oncometabolism (the metabolic features allowing cancer cells to proliferate) and immunometabolism (the catabolic and anabolic reactions that regulate immune responses). Recent preclinical studies demonstrated that multiple dietary changes can improve anticancer immunosurveillance of chemo-, radio- and immunotherapy. These findings have fostered the design of clinical trials evaluating the capacity of dietary interventions to synergize with treatment and hence limit tumor progression. Here, we discuss the scientific rationale for harnessing dietary interventions to improve the efficacy of anticancer therapy and present up-to-date information on clinical trials currently investigating this possibility.
Collapse
Affiliation(s)
- Sarah Lévesque
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Fondation pour la Recherche Médicale, Paris, France
| | - Jonathan G Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Gladys Ferrere
- INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Laurence Zitvogel
- Université Paris-Saclay, Orsay, France.,INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
861
|
Huang J, Li YQ, Wu CH, Zhang YL, Zhao ST, Chen YJ, Deng YH, Xuan A, Sun XD. The effect of ketogenic diet on behaviors and synaptic functions of naive mice. Brain Behav 2019; 9:e01246. [PMID: 30848079 PMCID: PMC6456772 DOI: 10.1002/brb3.1246] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Beyond its application as an epilepsy therapy, the ketogenic diet (KD) has been considered a potential treatment for a variety of other neurological and metabolic disorders. However, whether KD promotes functional restoration by reducing the pathological processes underlying individual diseases or through some independent mechanisms is not clear. METHODS In this study, we evaluated the effect of KD on a series of behaviors and synaptic functions of young adult naive mice. Wild-type C57BL/6J mice at age of 2-3 months were fed with control diet or KD for three months. Body weight and caloric intake were monitored throughout the experiments. We assessed behavioral performance with seizure induction, motor coordination and activity, anxiety level, spatial learning and memory, sociability, and depression. Synaptic transmission and long-term potentiation were also recorded. RESULTS KD-fed mice performed equivalent to control-diet-fed mice in the behavioral tests and electrophysiological assays except exhibiting slower weight gain and increased seizure threshold. CONCLUSIONS Our results contribute to the better understanding of effects of the KD on physiological behaviors and synaptic functions.
Collapse
Affiliation(s)
- Jie Huang
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuan-Quan Li
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cui-Hong Wu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun-Long Zhang
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shen-Ting Zhao
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yong-Jun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu-Hong Deng
- Department of Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aiguo Xuan
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang-Dong Sun
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, School of Basic Medical Sciences, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, China
| |
Collapse
|
862
|
Kim JH, Lee M, Kim SH, Kim SR, Lee BW, Kang ES, Cha BS, Cho JW, Lee YH. Sodium-glucose cotransporter 2 inhibitors regulate ketone body metabolism via inter-organ crosstalk. Diabetes Obes Metab 2019; 21:801-811. [PMID: 30407726 DOI: 10.1111/dom.13577] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/18/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022]
Abstract
AIM To investigate sodium-glucose cotransporter 2 inhibitor (SGLT2i)-induced changes in ketogenic enzymes and transporters in normal and diabetic mice models. MATERIALS AND METHODS Normal mice were randomly assigned to receive either vehicle or SGLT2i (25 mg/kg/d by oral gavage) for 7 days. Diabetic mice were treated with vehicle, insulin (4.5 units/kg/d by subcutaneous injection) or SGLT2i (25 mg/kg/d by intra-peritoneal injection) for 5 weeks. Serum and tissues of ketogenic organs were analysed. RESULTS In both normal and diabetic mice, SGLT2i increased beta-hydroxybutyrate (BHB) content in liver, kidney and colon tissue, as well as in serum and urine. In these organs, SGLT2i upregulated mRNA expression of ketogenic enzymes, 3-hydroxy-3-methylglutaryl-coenzyme A synthase 2 and 3-hydroxy-3-methylglutaryl-coenzyme A lyase. Similar patterns were observed in the kidney, ileum and colon for mRNA and protein expression of sodium-dependent monocarboxylate transporters (SMCTs), which mediate the cellular uptake of BHB and butyrate, an important substrate for intestinal ketogenesis. In diabetic mice under euglycaemic conditions, SGLT2i increased major ketogenic enzymes and SMCTs, while insulin suppressed ketogenesis. CONCLUSIONS SGLT2i increased systemic and tissue BHB levels by upregulating ketogenic enzymes and transporters in the liver, kidney and intestine, suggesting the integrated physiological consequences for ketone body metabolism of SGLT2i administration.
Collapse
Affiliation(s)
- Jin Hee Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minyoung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo Hyun Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Ra Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Seok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Bong-Soo Cha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Won Cho
- Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
- Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
863
|
HDAC1 and HDAC2 independently regulate common and specific intrinsic responses in murine enteroids. Sci Rep 2019; 9:5363. [PMID: 30926862 PMCID: PMC6441098 DOI: 10.1038/s41598-019-41842-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023] Open
Abstract
Both HDAC1 and HDAC2 are class I deacetylases acting as erasers of lysine-acetyl marks on histones and non-histone proteins. Several histone deacetylase inhibitors, either endogenous to the cell, such as the ketogenic β-hydroxybutyrate metabolite, or exogenous, such as butyrate, a microbial-derived metabolite, regulate HDAC activity. Different combinations of intestinal epithelial cell (IEC)-specific Hdac1 and/or Hdac2 deletion differentially alter mucosal homeostasis in mice. Thus, HDAC1 and HDAC2 could act as sensors and transmitters of environmental signals to the mucosa. In this study, enteroid culture models deleted for Hdac1 or Hdac2 were established to determine IEC-specific function as assessed by global transcriptomic and proteomic approaches. Results show that Hdac1 or Hdac2 deficiency altered differentiation of Paneth and goblet secretory cells, which sustain physical and chemical protection barriers, and increased intermediate secretory cell precursor numbers. Furthermore, IEC Hdac1- and Hdac2-dependent common and specific biological processes were identified, including oxidation-reduction, inflammatory responses, and lipid-related metabolic processes, as well as canonical pathways and upstream regulators related to environment-dependent signaling through steroid receptor pathways, among others. These findings uncover unrecognized regulatory similarities and differences between Hdac1 and Hdac2 in IEC, and demonstrate how HDAC1 and HDAC2 may complement each other to regulate the intrinsic IEC phenotype.
Collapse
|
864
|
Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: A route to metastasization and a target for therapy. Eur J Med Chem 2019; 171:343-363. [PMID: 30928707 DOI: 10.1016/j.ejmech.2019.03.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed the peculiarities of metabolic reprogramming in tumour onset and progression, and their relevance in cancer therapy. Also, it has been indicated that the metastatic process may depend on the metabolic rewiring and adaptation of cancer cells to the pressure of tumour microenvironment and limiting nutrient availability. The present review gatherers the existent knowledge on the influence of tumour microenvironment and metabolic routes driving metastasis. A focus will be given to glycolysis, fatty acid metabolism, glutaminolysis, and amino acid handling. In addition, the role of metabolic waste driving metastasization will be explored. Finally, we discuss the status of cancer treatment approaches targeting metabolism. This knowledge revision will highlight the critical metabolic targets in metastasis and the chemicals already used in preclinical studies and clinical trials, providing clues that would be further exploited in medicinal chemistry research.
Collapse
Affiliation(s)
- Tiago Ma Carvalho
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Henrique J Cardoso
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
865
|
Lee WD, Mukha D, Aizenshtein E, Shlomi T. Spatial-fluxomics provides a subcellular-compartmentalized view of reductive glutamine metabolism in cancer cells. Nat Commun 2019; 10:1351. [PMID: 30903027 PMCID: PMC6430770 DOI: 10.1038/s41467-019-09352-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 03/01/2019] [Indexed: 12/23/2022] Open
Abstract
The inability to inspect metabolic activities within subcellular compartments has been a major barrier to our understanding of eukaryotic cell metabolism. Here, we describe a spatial-fluxomics approach for inferring metabolic fluxes in mitochondria and cytosol under physiological conditions, combining isotope tracing, rapid subcellular fractionation, LC-MS-based metabolomics, computational deconvolution, and metabolic network modeling. Applied to study reductive glutamine metabolism in cancer cells, shown to mediate fatty acid biosynthesis under hypoxia and defective mitochondria, we find a previously unappreciated role of reductive IDH1 as the sole net contributor of carbons to fatty acid biosynthesis under standard normoxic conditions in HeLa cells. In murine cells with defective SDH, we find that reductive biosynthesis of citrate in mitochondria is followed by a reversed CS activity, suggesting a new route for supporting pyrimidine biosynthesis. We expect this spatial-fluxomics approach to be a highly useful tool for elucidating the role of metabolic dysfunction in human disease. Measuring metabolic fluxes in cellular compartments is a challenge. Here, the authors introduce an approach to infer fluxes in mitochondria and cytosol, and find that IDH1 is the major producer of cytosolic citrate in HeLa cells and that in SDH- deficient cells citrate synthase functions in reverse.
Collapse
Affiliation(s)
- Won Dong Lee
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | | | - Elina Aizenshtein
- Lokey Center for Life Science and Engineering, Technion, 32000, Haifa, Israel
| | - Tomer Shlomi
- Faculty of Biology, Technion, 32000, Haifa, Israel. .,Lokey Center for Life Science and Engineering, Technion, 32000, Haifa, Israel. .,Faculty of Computer Science, Technion, 32000, Haifa, Israel.
| |
Collapse
|
866
|
Iso T, Haruyama H, Sunaga H, Matsui M, Matsui H, Tanaka R, Umbarawan Y, Syamsunarno MRAA, Yokoyama T, Kurabayashi M. Exercise endurance capacity is markedly reduced due to impaired energy homeostasis during prolonged fasting in FABP4/5 deficient mice. BMC PHYSIOLOGY 2019; 19:1. [PMID: 30866899 PMCID: PMC6415495 DOI: 10.1186/s12899-019-0038-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 03/06/2019] [Indexed: 11/25/2022]
Abstract
Background Skeletal muscle prefers carbohydrate use to fatty acid (FA) use as exercise intensity increases. In contrast, skeletal muscle minimizes glucose use and relies more on FA during fasting. In mice deficient for FABP4 and FABP5 (double knockout (DKO) mice), FA utilization by red skeletal muscle and the heart is markedly reduced by the impairment of trans-endothelial FA transport, with an increase in glucose use to compensate for reduced FA uptake even during fasting. We attempted to determine whether prolonged fasting affects exercise performance in DKO mice, where constant glucose utilization occurs. Results A single bout of treadmill exercise was performed in the fed and fasted states. The initial speed was 10 m/min, and gradually increased by 5 m/min every 5 min up to 30 m/min until the mice stopped running. Running distance was significantly reduced by DKO genotype and prior fasting, leading to the shortest distance in fasted DKO mice. Levels of glycogen in skeletal muscle and the liver were nearly depleted in both WT and DKO mice during prolonged fasting prior to exercise. Levels of TG in skeletal muscle were not reduced by exercise in fasted DKO mice, suggesting that intramuscular TG was not utilized during exercise. Hypoglycaemia was accelerated in fasted DKO mice, and this acceleration could be due to constant glucose utilization by red skeletal muscle and the heart where FA uptake is diminished due to defective trans-endothelial FA transport. Taken together, energy supply from serum and storage in skeletal muscle were very low in fasted DKO mice, which could lead to a significant reduction in exercise performance. Conclusions FABP4/5 have crucial roles in nutrient homeostasis during prolonged fasting for maintaining exercise endurance capacity.
Collapse
Affiliation(s)
- Tatsuya Iso
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Hikari Haruyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Miki Matsui
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Rina Tanaka
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yogi Umbarawan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Department of Internal Medicine, Faculty of Medicine Universitas Indonesia, Jl. Salemba Raya no. 6, Jakarta, 10430, Indonesia
| | - Mas Rizky A A Syamsunarno
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Department of Biochemistry and Molecular Biology, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM 21, Jatinangor, West Java, 45363, Indonesia
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
867
|
Liu K, Li F, Sun Q, Lin N, Han H, You K, Tian F, Mao Z, Li T, Tong T, Geng M, Zhao Y, Gu W, Zhao W. p53 β-hydroxybutyrylation attenuates p53 activity. Cell Death Dis 2019; 10:243. [PMID: 30858356 PMCID: PMC6411878 DOI: 10.1038/s41419-019-1463-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/08/2019] [Accepted: 02/01/2019] [Indexed: 12/16/2022]
Abstract
p53 is an essential tumor suppressor, whose activity is finely tuned by the posttranslational modifications. Previous research has reported that β-hydroxybutyrate (BHB) induces β-hydroxybutyrylation (Kbhb), which is a novel histone posttranslational modification. Here we report that p53 is modified by kbhb and that this modification occurs at lysines 120, 319, and 370 of p53. We demonstrate that the level of p53 kbhb is dramatically increased in cultured cells treated with BHB and in thymus tissues of fasted mice, and that CBP catalyze p53 kbhb. We show that p53 kbhb results in lower levels of p53 acetylation and reduced expression of the p53 downstream genes p21 and PUMA, as well as reduced cell growth arrest and apoptosis in cultured cells under p53-activating conditions. Similar results were observed in mouse thymus tissue under starvation conditions, which result in increased concentrations of serum BHB, and in response to genotoxic stress caused by γ-irradiation to activate p53. Our findings thus show that BHB-mediated p53 kbhb is a novel mechanism of p53 activity regulation, which may explain the link between ketone bodies and tumor, and which may provide promising therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Kun Liu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Fangzhou Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Qianqian Sun
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Ning Lin
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Haichao Han
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Kaiqiang You
- Department of Biomedical Informatics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Feng Tian
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Zebin Mao
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Tingting Li
- Department of Biomedical Informatics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Tanjun Tong
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China
| | - Meiyu Geng
- Department of Pharmacology I, Shanghai Institute of Materia Medica, 555 Zu Chong Zhi Road, Zhang Jiang Hi-Tech Park, 201203, Pudong, Shanghai, China
| | - Yingming Zhao
- Ben May Department of Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY, 10032, USA
| | - Wenhui Zhao
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 38 Xueyuan Road, 100191, Beijing, China.
| |
Collapse
|
868
|
Plutzky J, Bakris G. Sodium/Glucose Cotransporter 2 Inhibitors in Patients With Diabetes Mellitus and Chronic Kidney Disease: Turning the Page. Circulation 2019; 137:130-133. [PMID: 29311346 DOI: 10.1161/circulationaha.117.031422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jorge Plutzky
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (J.P.)
| | - George Bakris
- Comprehensive Hypertension Center, Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medicine, IL (G.B.)
| |
Collapse
|
869
|
Affiliation(s)
- Simon Sedej
- Department of Cardiology, Medical University of Graz, BioTechMed, Graz, Austria
| |
Collapse
|
870
|
Cortassa S, Aon MA, Sollott SJ. Control and Regulation of Substrate Selection in Cytoplasmic and Mitochondrial Catabolic Networks. A Systems Biology Analysis. Front Physiol 2019; 10:201. [PMID: 30906265 PMCID: PMC6418011 DOI: 10.3389/fphys.2019.00201] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
Appropriate substrate selection between fats and glucose is associated with the success of interventions that maintain health such as exercise or caloric restriction, or with the severity of diseases such as diabetes or other metabolic disorders. Although the interaction and mutual inhibition between glucose and fatty-acids (FAs) catabolism has been studied for decades, a quantitative and integrated understanding of the control and regulation of substrate selection through central catabolic pathways is lacking. We addressed this gap here using a computational model representing cardiomyocyte catabolism encompassing glucose (Glc) utilization, pyruvate transport into mitochondria and oxidation in the tricarboxylic acid (TCA) cycle, β-oxidation of palmitate (Palm), oxidative phosphorylation, ion transport, pH regulation, and ROS generation and scavenging in cytoplasmic and mitochondrial compartments. The model is described by 82 differential equations and 119 enzymatic, electron transport and substrate transport reactions accounting for regulatory mechanisms and key players, namely pyruvate dehydrogenase (PDH) and its modulation by multiple effectors. We applied metabolic control analysis to the network operating with various Glc to Palm ratios. The flux and metabolites’ concentration control were visualized through heat maps providing major insights into main control and regulatory nodes throughout the catabolic network. Metabolic pathways located in different compartments were found to reciprocally control each other. For example, glucose uptake and the ATP demand exert control on most processes in catabolism while TCA cycle activities and membrane-associated energy transduction reactions exerted control on mitochondrial processes namely β-oxidation. PFK and PDH, two highly regulated enzymes, exhibit opposite behavior from a control perspective. While PFK activity was a main rate-controlling step affecting the whole network, PDH played the role of a major regulator showing high sensitivity (elasticity) to substrate availability and key activators/inhibitors, a trait expected from a flexible substrate selector strategically located in the metabolic network. PDH regulated the rate of Glc and Palm consumption, consistent with its high sensitivity toward AcCoA, CoA, and NADH. Overall, these results indicate that the control of catabolism is highly distributed across the metabolic network suggesting that fuel selection between FAs and Glc goes well beyond the mechanisms traditionally postulated to explain the glucose-fatty-acid cycle.
Collapse
Affiliation(s)
- Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
871
|
Adam ME, Fehervari M, Boshier PR, Chin ST, Lin GP, Romano A, Kumar S, Hanna GB. Mass-Spectrometry Analysis of Mixed-Breath, Isolated-Bronchial-Breath, and Gastric-Endoluminal-Air Volatile Fatty Acids in Esophagogastric Cancer. Anal Chem 2019; 91:3740-3746. [PMID: 30699297 DOI: 10.1021/acs.analchem.9b00148] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A noninvasive breath test has the potential to improve survival from esophagogastric cancer by facilitating earlier detection. This study aimed to investigate the production of target volatile fatty acids (VFAs) in esophagogastric cancer through analysis of the ex vivo headspace above underivatized tissues and in vivo analysis within defined anatomical compartments, including analysis of mixed breath, isolated bronchial breath, and gastric-endoluminal air. VFAs were measured by PTR-ToF-MS and GC-MS. Levels of VFAs (acetic, butyric, pentanoic, and hexanoic acids) and acetone were elevated in ex vivo experiments in the headspace above esophagogastric cancer compared with the levels in samples from control subjects with morphologically normal and benign conditions of the upper gastrointestinal tract. In 25 patients with esophagogastric cancer and 20 control subjects, receiver-operating-characteristic analysis for the cancer-specific VFAs butyric acid ( P < 0.001) and pentatonic acid ( P = 0.005) within in vivo gastric-endoluminal air gave an area under the curve of 0.80 (95% confidence interval of 0.65 to 0.93, P = 0.01). Compared with mixed- and bronchial-breath samples, all examined VFAs were found in highest concentrations within esophagogastric-endoluminal air. In addition, VFAs were higher in all samples derived from cancer patients compared with in the controls. Equivalence of VFA levels within the mixed and bronchial breath of cancer patients suggests that their origin within breath is principally derived from the lungs and, by inference, from the systemic circulation as opposed to direct passage from the upper gastrointestinal tract. These findings highlight the potential to utilize VFAs for endoluminal-gas biopsies and noninvasive mixed-exhaled-breath testing for esophagogastric-cancer detection.
Collapse
Affiliation(s)
- Mina E Adam
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Matyas Fehervari
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Piers R Boshier
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Sung-Tong Chin
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Geng-Ping Lin
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Andrea Romano
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| | - Sacheen Kumar
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
- Department of Upper Gastrointestinal Surgery , The Royal Marsden Hospital , London SW3 6JJ , United Kingdom
| | - George B Hanna
- Department of Surgery & Cancer , Imperial College London , London W2 1NY , United Kingdom
| |
Collapse
|
872
|
Najac C, Radoul M, Le Page LM, Batsios G, Subramani E, Viswanath P, Gillespie AM, Ronen SM. In vivo investigation of hyperpolarized [1,3- 13C 2]acetoacetate as a metabolic probe in normal brain and in glioma. Sci Rep 2019; 9:3402. [PMID: 30833594 PMCID: PMC6399277 DOI: 10.1038/s41598-019-39677-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Dysregulation in NAD+/NADH levels is associated with increased cell division and elevated levels of reactive oxygen species in rapidly proliferating cancer cells. Conversion of the ketone body acetoacetate (AcAc) to β-hydroxybutyrate (β-HB) by the mitochondrial enzyme β-hydroxybutyrate dehydrogenase (BDH) depends upon NADH availability. The β-HB-to-AcAc ratio is therefore expected to reflect mitochondrial redox. Previous studies reported the potential of hyperpolarized 13C-AcAc to monitor mitochondrial redox in cells, perfused organs and in vivo. However, the ability of hyperpolarized 13C-AcAc to cross the blood brain barrier (BBB) and its potential to monitor brain metabolism remained unknown. Our goal was to assess the value of hyperpolarized [1,3-13C2]AcAc in healthy and tumor-bearing mice in vivo. Following hyperpolarized [1,3-13C2]AcAc injection, production of [1,3-13C2]β-HB was detected in normal and tumor-bearing mice. Significantly higher levels of [1-13C]AcAc and lower [1-13C]β-HB-to-[1-13C]AcAc ratios were observed in tumor-bearing mice. These results were consistent with decreased BDH activity in tumors and associated with increased total cellular NAD+/NADH. Our study confirmed that AcAc crosses the BBB and can be used for monitoring metabolism in the brain. It highlights the potential of AcAc for future clinical translation and its potential utility for monitoring metabolic changes associated with glioma, and other neurological disorders.
Collapse
Affiliation(s)
- Chloé Najac
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Marina Radoul
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Lydia M Le Page
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States.,Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA, United States
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Elavarasan Subramani
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
873
|
Alexandre-Gouabau MC, Moyon T, David-Sochard A, Fenaille F, Cholet S, Royer AL, Guitton Y, Billard H, Darmaun D, Rozé JC, Boquien CY. Comprehensive Preterm Breast Milk Metabotype Associated with Optimal Infant Early Growth Pattern. Nutrients 2019; 11:E528. [PMID: 30823457 PMCID: PMC6470768 DOI: 10.3390/nu11030528] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Early nutrition impacts preterm infant early growth rate and brain development but can have long lasting effects as well. Although human milk is the gold standard for feeding new born full-term and preterm infants, little is known about the effects of its bioactive compounds on breastfed preterm infants' growth outcomes. This study aims to determine whether breast milk metabolome, glycome, lipidome, and free-amino acids profiles analyzed by liquid chromatography-mass spectrometry had any impact on the early growth pattern of preterm infants. The study population consisted of the top tercile-Z score change in their weight between birth and hospital discharge ("faster grow", n = 11) and lowest tercile ("slower grow", n = 15) from a cohort of 138 premature infants (27⁻34 weeks gestation). This holistic approach combined with stringent clustering or classification statistical methods aims to discriminate groups of milks phenotype and identify specific metabolites associated with early growth of preterm infants. Their predictive reliability as biomarkers of infant growth was assessed using multiple linear regression and taking into account confounding clinical factors. Breast-milk associated with fast growth contained more branched-chain and insulino-trophic amino acid, lacto-N-fucopentaose, choline, and hydroxybutyrate, pointing to the critical role of energy utilization, protein synthesis, oxidative status, and gut epithelial cell maturity in prematurity.
Collapse
Affiliation(s)
- Marie-Cécile Alexandre-Gouabau
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
| | - Thomas Moyon
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
| | - Agnès David-Sochard
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
| | - François Fenaille
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB, F-91191 Gif-sur-Yvette, France.
| | - Sophie Cholet
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB, F-91191 Gif-sur-Yvette, France.
| | - Anne-Lise Royer
- LUNAM Université, ON;IRIS, Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), USC INRA 1329, Nantes F-44307, France.
| | - Yann Guitton
- LUNAM Université, ON;IRIS, Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), USC INRA 1329, Nantes F-44307, France.
| | - Hélène Billard
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
| | - Dominique Darmaun
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
- CHU, Centre Hospitalo-Universitaire Hôtel-Dieu, Nantes F-44093, France.
| | - Jean-Christophe Rozé
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
- CHU, Centre Hospitalo-Universitaire Hôtel-Dieu, Nantes F-44093, France.
| | - Clair-Yves Boquien
- INRA, UMR1280, Physiopathologie des Adaptations Nutritionnelles, Institut des maladies de l'appareil digestif (IMAD), Centre de Recherche en Nutrition Humaine Ouest (CRNH), Nantes F-44093, France.
- EMBA, European Milk Bank Association, Milano I-20126, Italy.
| |
Collapse
|
874
|
Kim S, Jeon JM, Kwon OK, Choe MS, Yeo HC, Peng X, Cheng Z, Lee MY, Lee S. Comparative Proteomic Analysis Reveals the Upregulation of Ketogenesis in Cardiomyocytes Differentiated from Induced Pluripotent Stem Cells. Proteomics 2019; 19:e1800284. [PMID: 30724459 DOI: 10.1002/pmic.201800284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/08/2018] [Indexed: 12/12/2022]
Abstract
Diverse metabolic pathways, such as the tricarboxylic acid cycle, pyruvate metabolism, and oxidative phosphorylation, regulate the differentiation of induced pluripotent stem cells (iPSCs) to cells of specific lineages and organs. Here, the protein dynamics during cardiac differentiation of human iPSCs into cardiomyocytes (CMs) are characterized. The differentiation is induced by N-(6-methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide, a Wnt signaling inhibitor, and confirmed by the mRNA and protein expression of cTnT and MLC2A in CMs. For comparative proteomics, cells from three stages, namely, hiPSCs, cardiac progenitor cells, and CMs, are prepared using the three-plex tandem mass tag labeling approach. In total, 3970 proteins in triplicate analysis are identified. As the result, the upregulation of proteins associated with branched chain amino acid degradation and ketogenesis by the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis are observed. The levels of 3-hydroxymethyl-3-methylglutaryl-CoA lyase, 3-hydroxymethyl-3-methylglutaryl-CoA synthase 2, and 3-hydroxybutyrate dehydrogenase 1, involved in ketone body metabolism, are determined using western blotting, and the level of acetoacetate, the final product of ketogenesis, is higher in CMs. Taken together, these observations indicate that proteins required for the production of diverse energy sources are naturally self-expressed during cardiomyogenic differentiation. Furthermore, acetoacetate concentration might act as a regulator of this differentiation.
Collapse
Affiliation(s)
- Sunjoo Kim
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ju Mi Jeon
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Oh Kwang Kwon
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Mu Seog Choe
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Han Cheol Yeo
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Xiaojun Peng
- PTM Biolab LLC, Hangzhou, Zhejiang, 310018, China
| | | | - Min Young Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| |
Collapse
|
875
|
Güntner AT, Abegg S, Königstein K, Gerber PA, Schmidt-Trucksäss A, Pratsinis SE. Breath Sensors for Health Monitoring. ACS Sens 2019; 4:268-280. [PMID: 30623644 DOI: 10.1021/acssensors.8b00937] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breath sensors can revolutionize medical diagnostics by on-demand detection and monitoring of health parameters in a noninvasive and personalized fashion. Despite extensive research for more than two decades, however, only a few breath sensors have been translated into clinical practice. Actually, most never even left the scientific laboratories. Here, we describe key challenges that currently impede realization of breath sensors and highlight strategies to overcome them. Specifically, we start with breath marker selection (with emphasis on metabolic and inflammatory markers) and breath sampling. Next, the sensitivity, stability, and selectivity requirements for breath sensors are described. Concepts are elaborated to systematically address these requirements by material design (focusing on chemoresistive metal oxides), orthogonal arrays, and filters. Finally, aspects of portable device integration, user communication, and clinical applicability are discussed.
Collapse
Affiliation(s)
- Andreas T. Güntner
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
- Department of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Sebastian Abegg
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
| | - Karsten Königstein
- Division Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, CH-4052 Basel, Switzerland
| | - Philipp A. Gerber
- Department of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Arno Schmidt-Trucksäss
- Division Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, CH-4052 Basel, Switzerland
| | - Sotiris E. Pratsinis
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
| |
Collapse
|
876
|
Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, Matsuura TR, Petucci C, Lewandowski ED, Crawford PA, Muoio DM, Recchia FA, Kelly DP. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 2019; 4:124079. [PMID: 30668551 DOI: 10.1172/jci.insight.124079] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
Evidence has emerged that the failing heart increases utilization of ketone bodies. We sought to determine whether this fuel shift is adaptive. Mice rendered incapable of oxidizing the ketone body 3-hydroxybutyrate (3OHB) in the heart exhibited worsened heart failure in response to fasting or a pressure overload/ischemic insult compared with WT controls. Increased delivery of 3OHB ameliorated pathologic cardiac remodeling and dysfunction in mice and in a canine pacing model of progressive heart failure. 3OHB was shown to enhance bioenergetic thermodynamics of isolated mitochondria in the context of limiting levels of fatty acids. These results indicate that the heart utilizes 3OHB as a metabolic stress defense and suggest that strategies aimed at increasing ketone delivery to the heart could prove useful in the treatment of heart failure.
Collapse
Affiliation(s)
- Julie L Horton
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA
| | - Michael T Davidson
- Departments of Medicine and Pharmacology, and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Clara Kurishima
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Rick B Vega
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA
| | - Jeffery C Powers
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Timothy R Matsuura
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher Petucci
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - E Douglas Lewandowski
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Davis Heart and Lung Research Institute and Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Peter A Crawford
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Departments of Medicine and Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology, and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Fabio A Recchia
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA.,Institute of Life Sciences, Scuola Superiore Sant'Anna Pisa, Fondazione G. Monasterio, Pisa, Italy
| | - Daniel P Kelly
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
877
|
HSIEH CL, LIN PY, AKITA T, MITA M, IDE T, LEE JA, HAMASE K. Development of a Three-Dimensional HPLC System for the Simultaneous Determination of Lactate and 3-Hydroxybutyrate Enantiomers in Mammalian Urine. CHROMATOGRAPHY 2019. [DOI: 10.15583/jpchrom.2018.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Chin-Ling HSIEH
- Graduate School of Pharmaceutical Sciences, Kyushu University
- School of Pharmacy, Taipei Medical University
| | - Po-Yeh LIN
- School of Pharmacy, Taipei Medical University
| | - Takeyuki AKITA
- Graduate School of Pharmaceutical Sciences, Kyushu University
| | | | - Tomomi IDE
- Graduate School of Medical Sciences, Kyushu University
| | - Jen-Ai LEE
- School of Pharmacy, Taipei Medical University
| | - Kenji HAMASE
- Graduate School of Pharmaceutical Sciences, Kyushu University
- School of Pharmacy, Taipei Medical University
| |
Collapse
|
878
|
Janssen JJE, Grefte S, Keijer J, de Boer VCJ. Mito-Nuclear Communication by Mitochondrial Metabolites and Its Regulation by B-Vitamins. Front Physiol 2019; 10:78. [PMID: 30809153 PMCID: PMC6379835 DOI: 10.3389/fphys.2019.00078] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are cellular organelles that control metabolic homeostasis and ATP generation, but also play an important role in other processes, like cell death decisions and immune signaling. Mitochondria produce a diverse array of metabolites that act in the mitochondria itself, but also function as signaling molecules to other parts of the cell. Communication of mitochondria with the nucleus by metabolites that are produced by the mitochondria provides the cells with a dynamic regulatory system that is able to respond to changing metabolic conditions. Dysregulation of the interplay between mitochondrial metabolites and the nucleus has been shown to play a role in disease etiology, such as cancer and type II diabetes. Multiple recent studies emphasize the crucial role of nutritional cofactors in regulating these metabolic networks. Since B-vitamins directly regulate mitochondrial metabolism, understanding the role of B-vitamins in mito-nuclear communication is relevant for therapeutic applications and optimal dietary lifestyle. In this review, we will highlight emerging concepts in mito-nuclear communication and will describe the role of B-vitamins in mitochondrial metabolite-mediated nuclear signaling.
Collapse
Affiliation(s)
| | | | | | - Vincent C. J. de Boer
- Human and Animal Physiology, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
879
|
Puchalska P, Martin SE, Huang X, Lengfeld JE, Daniel B, Graham MJ, Han X, Nagy L, Patti GJ, Crawford PA. Hepatocyte-Macrophage Acetoacetate Shuttle Protects against Tissue Fibrosis. Cell Metab 2019; 29:383-398.e7. [PMID: 30449686 PMCID: PMC6559243 DOI: 10.1016/j.cmet.2018.10.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/20/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Abstract
Metabolic plasticity has been linked to polarized macrophage function, but mechanisms connecting specific fuels to tissue macrophage function remain unresolved. Here we apply a stable isotope tracing, mass spectrometry-based untargeted metabolomics approach to reveal the metabolome penetrated by hepatocyte-derived glucose and ketone bodies. In both classically and alternatively polarized macrophages, [13C]acetoacetate (AcAc) labeled ∼200 chemical features, but its reduced form D-[13C]β-hydroxybutyrate (D-βOHB) labeled almost none. [13C]glucose labeled ∼500 features, and while unlabeled AcAc competed with only ∼15% of them, the vast majority required the mitochondrial enzyme succinyl-coenzyme A-oxoacid transferase (SCOT). AcAc carbon labeled metabolites within the cytoplasmic glycosaminoglycan pathway, which regulates tissue fibrogenesis. Accordingly, livers of mice lacking SCOT in macrophages were predisposed to accelerated fibrogenesis. Exogenous AcAc, but not D-βOHB, ameliorated diet-induced hepatic fibrosis. These data support a hepatocyte-macrophage ketone shuttle that segregates AcAc from D-βOHB, coordinating the fibrogenic response to hepatic injury via mitochondrial metabolism in tissue macrophages.
Collapse
Affiliation(s)
- Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Shannon E Martin
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Xiaojing Huang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Chemistry, Washington University, St. Louis, MO 63110, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Justin E Lengfeld
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA
| | - Bence Daniel
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Medicine, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA
| | | | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Barshop Institute for Longevity and Aging Studies, Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Laszlo Nagy
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Medicine, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA; Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63110, USA
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
880
|
Tiulganova DA, Nasaev SS, Chugreev IA, Rodionova MA, Zavyalov GA. [Mechanisms of ketogenic diet action]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:72-75. [PMID: 30698548 DOI: 10.17116/jnevro201811810272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The paper considers the necessity of using ketogenic diet and its efficacy in epilepsy. Direct and indirect effects of ketones on brain cells and molecular mechanisms of their action are discussed in detail.
Collapse
Affiliation(s)
- D A Tiulganova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Sh Sh Nasaev
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - I A Chugreev
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - M A Rodionova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - G A Zavyalov
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
881
|
Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci Rep 2019; 9:742. [PMID: 30679586 PMCID: PMC6346118 DOI: 10.1038/s41598-018-36941-9] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022] Open
Abstract
Butyrate and R-β-hydroxybutyrate are two related short chain fatty acids naturally found in mammals. Butyrate, produced by enteric butyric bacteria, is present at millimolar concentrations in the gastrointestinal tract and at lower levels in blood; R-β-hydroxybutyrate, the main ketone body, produced by the liver during fasting can reach millimolar concentrations in the circulation. Both molecules have been shown to be histone deacetylase (HDAC) inhibitors, and their administration has been associated to an improved metabolic profile and better cellular oxidative status, with butyrate inducing PGC1α and fatty acid oxidation and R-β-hydroxybutyrate upregulating oxidative stress resistance factors FOXO3A and MT2 in mouse kidney. Because of the chemical and functional similarity between the two molecules, we compared here their impact on multiple cell types, evaluating i) histone acetylation and hydroxybutyrylation levels by immunoblotting, ii) transcriptional regulation of metabolic and inflammatory genes by quantitative PCR and iii) cytokine secretion profiles using proteome profiling array analysis. We confirm that butyrate is a strong HDAC inhibitor, a characteristic we could not identify in R-β-hydroxybutyrate in vivo nor in vitro. Butyrate had an extensive impact on gene transcription in rat myotubes, upregulating PGC1α, CPT1b, mitochondrial sirtuins (SIRT3-5), and the mitochondrial anti-oxidative genes SOD2 and catalase. In endothelial cells, butyrate suppressed gene expression and LPS-induced secretion of several pro-inflammatory genes, while R-β-hydroxybutyrate acted as a slightly pro-inflammatory molecule. Our observations indicate that butyrate induces transcriptional changes to a higher extent than R-β-hydroxybutyrate in rat myotubes and endothelial cells, in keep with its HDAC inhibitory activity. Also, in contrast with previous reports, R-β-hydroxybutyrate, while inducing histone β-hydroxybutyrylation, did not display a readily detectable HDAC inhibitor activity and exerted a slight pro-inflammatory action on endothelial cells.
Collapse
|
882
|
Abstract
Significance: Diabetic cardiomyopathy (DCM) is a frequent complication occurring even in well-controlled asymptomatic diabetic patients, and it may advance to heart failure (HF). Recent Advances: The diabetic heart is characterized by a state of "metabolic rigidity" involving enhanced rates of fatty acid uptake and mitochondrial oxidation as the predominant energy source, and it exhibits mitochondrial electron transport chain defects. These alterations promote redox state changes evidenced by a decreased NAD+/NADH ratio associated with an increase in acetyl-CoA/CoA ratio. NAD+ is a co-substrate for deacetylases, sirtuins, and a critical molecule in metabolism and redox signaling; whereas acetyl-CoA promotes protein lysine acetylation, affecting mitochondrial integrity and causing epigenetic changes. Critical Issues: DCM lacks specific therapies with treatment only in later disease stages using standard, palliative HF interventions. Traditional therapy targeting neurohormonal signaling and hemodynamics failed to improve mortality rates. Though mitochondrial redox state changes occur in the heart with obesity and diabetes, how the mitochondrial NAD+/NADH redox couple connects the remodeled energy metabolism with mitochondrial and cytosolic antioxidant defense and nuclear epigenetic changes remains to be determined. Mitochondrial therapies targeting the mitochondrial NAD+/NADH redox ratio may alleviate cardiac dysfunction. Future Directions: Specific therapies must be supported by an optimal understanding of changes in mitochondrial redox state and how it influences other cellular compartments; this field has begun to surface as a therapeutic target for the diabetic heart. We propose an approach based on an alternate mitochondrial electron transport that normalizes the mitochondrial redox state and improves cardiac function in diabetes.
Collapse
Affiliation(s)
- Jessica M Berthiaume
- 1 Department of Physiology & Biophysics, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| | - Jacob G Kurdys
- 2 Department of Foundational Sciences, College of Medicine, Central Michigan University , Mount Pleasant, Michigan
| | - Danina M Muntean
- 3 Department of Functional Sciences-Pathophysiology, "Victor Babes" University of Medicine and Pharmacy , Timisoara, Romania
| | - Mariana G Rosca
- 2 Department of Foundational Sciences, College of Medicine, Central Michigan University , Mount Pleasant, Michigan
| |
Collapse
|
883
|
Tagawa R, Kawano Y, Minami A, Nishiumi S, Yano Y, Yoshida M, Kodama Y. β-hydroxybutyrate protects hepatocytes against endoplasmic reticulum stress in a sirtuin 1-independent manner. Arch Biochem Biophys 2019; 663:220-227. [PMID: 30664838 DOI: 10.1016/j.abb.2019.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 01/09/2023]
Abstract
β-hydroxybutyrate (BHB), a major ketone body in mammals, is produced from fatty acids through mitochondrial fatty acid oxidation in hepatocytes. To elucidate the role of BHB in the hepatic endoplasmic reticulum (ER), we examined the effects of BHB on hepatic ER stress induced by tunicamycin. In mouse hepatoma Hepa1c1c7 cells, BHB treatment suppressed the protein expression of ER stress responsive genes and increased cell viability, while reducing the protein expression of apoptosis inducible genes, without causing any alterations in the protein expression of sirtuin 1 (SIRT1) or the phosphorylation of AMP-activated protein kinase. The intraperitoneal administration of BHB also reduced the protein expression of ER stress responsive genes in mouse livers. In human hepatoma HepG2 cells, the protein expression levels of ER stress responsive genes were increased by the partial inhibition of BHB production with siRNA targeting endogenous 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) lyase, whereas they were decreased by promoting BHB production with fenofibrate. These findings revealed that BHB helps to suppress hepatic ER stress via a SIRT1-independent pathway, and it might be possible to manipulate ER stress by regulating BHB production genetically or pharmacologically.
Collapse
Affiliation(s)
- Ryoma Tagawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Kawano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Akihiro Minami
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shin Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiko Yano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaru Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Metabolomics Research, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan; AMED-CREST, AMED, Kobe, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
884
|
Interactions between Host PPARs and Gut Microbiota in Health and Disease. Int J Mol Sci 2019; 20:ijms20020387. [PMID: 30658440 PMCID: PMC6359605 DOI: 10.3390/ijms20020387] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/28/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022] Open
Abstract
The human gastrointestinal tract is inhabited by many types of microbiota, including bacteria, viruses, and fungi. Dysregulations of their microenvironment are associated with various health problems, not only limited to gastrointestinal disorders, such as inflammatory bowel disease, but to impacts beyond the intestine. For example, intestinal microbiota can affect the liver in non-alcoholic fatty liver disease, visceral adipose tissue during adipogenesis, and the heart in atherosclerosis. The factors contributing to these pathogeneses involve the gut microbiota and the effector organs of the host, and everything in between. The nuclear receptor peroxisome proliferator-activated receptors (PPARs) are pivotal for the modulation of many of the pathogeneses mentioned above. It is, therefore, conceivable that, in the process of host-microbiota interactions, PPARs play important roles. In this review, we focus on the interactions between host PPARs in different organs and gut microbiota and their impacts on maintaining health and various diseases.
Collapse
|
885
|
Amos D, Cook C, Santanam N. Omega 3 rich diet modulates energy metabolism via GPR120-Nrf2 crosstalk in a novel antioxidant mouse model. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:466-488. [PMID: 30658097 DOI: 10.1016/j.bbalip.2019.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/08/2023]
Abstract
With obesity rates reaching epidemic proportions, more studies concentrated on reducing the risk and treating this epidemic are vital. Redox stress is an important metabolic regulator involved in the pathophysiology of cardiovascular disease, Type 2 diabetes, and obesity. Oxygen and nitrogen-derived free radicals alter glucose and lipid homeostasis in key metabolic tissues, leading to increases in risk of developing metabolic syndrome. Oxidants derived from dietary fat differ in their metabolic regulation, with numerous studies showing benefits from a high omega 3 rich diet compared to the frequently consumed "western diet" rich in saturated fat. Omega 3 (OM3) fatty acids improve lipid profile, lower inflammation, and ameliorate insulin resistance, possibly through maintaining redox homeostasis. This study is based on the hypothesis that altering endogenous antioxidant production and/or increasing OM3 rich diet consumption will improve energy metabolism and maintain insulin sensitivity. We tested the comparative metabolic effects of a diet rich in saturated fat (HFD) and an omega 3-enriched diet (OM3) in the newly developed 'stress-less' mice model that overexpresses the endogenous antioxidant catalase. Eight weeks of dietary intervention showed that mice overexpressing endogenous catalase compared to their wild-type controls when fed an OM3 enriched diet, in contrast to HFD, activated GPR120-Nrf2 cross-talk to maintain balanced energy metabolism, normal circadian rhythm, and insulin sensitivity. These findings suggest that redox regulation of GPR120/FFAR4 might be an important target in reducing risk of metabolic syndrome and associated diseases.
Collapse
Affiliation(s)
- Deborah Amos
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States
| | - Carla Cook
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States.
| |
Collapse
|
886
|
Lundsgaard AM, Holm JB, Sjøberg KA, Bojsen-Møller KN, Myrmel LS, Fjære E, Jensen BAH, Nicolaisen TS, Hingst JR, Hansen SL, Doll S, Geyer PE, Deshmukh AS, Holst JJ, Madsen L, Kristiansen K, Wojtaszewski JFP, Richter EA, Kiens B. Mechanisms Preserving Insulin Action during High Dietary Fat Intake. Cell Metab 2019; 29:50-63.e4. [PMID: 30269983 DOI: 10.1016/j.cmet.2018.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/21/2018] [Accepted: 08/30/2018] [Indexed: 01/03/2023]
Abstract
Prolonged intervention studies investigating molecular metabolism are necessary for a deeper understanding of dietary effects on health. Here we provide mechanistic information about metabolic adaptation to fat-rich diets. Healthy, slightly overweight men ingested saturated or polyunsaturated fat-rich diets for 6 weeks during weight maintenance. Hyperinsulinemic clamps combined with leg balance technique revealed unchanged peripheral insulin sensitivity, independent of fatty acid type. Both diets increased fat oxidation potential in muscle. Hepatic insulin clearance increased, while glucose production, de novo lipogenesis, and plasma triacylglycerol decreased. High fat intake changed the plasma proteome in the immune-supporting direction and the gut microbiome displayed changes at taxonomical and functional level with polyunsaturated fatty acid (PUFA). In mice, eucaloric feeding of human PUFA and saturated fatty acid diets lowered hepatic triacylglycerol content compared with low-fat-fed control mice, and induced adaptations in the liver supportive of decreased gluconeogenesis and lipogenesis. Intake of fat-rich diets thus induces extensive metabolic adaptations enabling disposition of dietary fat without metabolic complications.
Collapse
Affiliation(s)
- Anne-Marie Lundsgaard
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Jacob B Holm
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Clinical Microbiomics, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | | | | | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | - Benjamin A H Jensen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Department of Medicine, Laval University, Quebec, QC, Canada
| | - Trine S Nicolaisen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Sine L Hansen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Sophia Doll
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Philip E Geyer
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Atul S Deshmukh
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Institute of Marine Research, Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Institute of Metagenomics, BGI-Shenzhen, Shenzhen, China
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark.
| |
Collapse
|
887
|
Wang D, Shang W, Zhang B, Jiang C, Qu F, Yang M. Manganese-doped zinc oxide hollow balls for chemiresistive sensing of acetone vapors. Mikrochim Acta 2019; 186:44. [PMID: 30607598 DOI: 10.1007/s00604-018-3108-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
Abstract
Both pure and Mn(II)-doped ZnO hollow structures were synthesized by a solvothermal reaction, and their phase structures, morphologies and elemental composition were characterized. SEM and TEM observations show the pure ZnO and the Mn(II)-doped ZnO balls to possess similar hollow structure with a particle size of about 1.5 μm. Their sensing properties were investigated, and the composite containing 1 atom% of Mn(II) (1% Mn-ZnO) is found be display the highest selectivity for acetone. The detection limit is 100 ppm acetone at 234 °C which is 4.6 times lower than that of the pure ZnO. In addition, the response time is shorter. Graphical abstract ZnO and Mn-doped ZnO hollow balls were prepared by a hydrothermal method, and their gas-sensing properties were investigated. Zinc(II) oxide doped with 1 atom% Mn(II) demonstrated an outstanding sensing behavior towards acetone vapors.
Collapse
Affiliation(s)
- Dongting Wang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China.,Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Wenan Shang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China.,Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Bingxue Zhang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China.,Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Chunjie Jiang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China.
| | - Fengdong Qu
- Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China.
| | - Minghui Yang
- Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China.
| |
Collapse
|
888
|
Depczynski B, Lee ATK, Varndell W, Chiew AL. The Significance of an Increased Beta-Hydroxybutyrate at Presentation to the Emergency Department in Patients with Diabetes in the Absence of a Hyperglycemic Emergency. J Diabetes Res 2019; 2019:7387128. [PMID: 31687409 PMCID: PMC6811785 DOI: 10.1155/2019/7387128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022] Open
Abstract
The significance of hyperketonemia in adults with diabetes presenting to the emergency department with acute illness, not due to a diabetic hyperglycemic emergency, has not been well characterized. Adult patients with diabetes presenting to the emergency department who had venous blood gas and beta-hydroxybutyrate levels measured whilst in the emergency department were retrospectively evaluated for the relationship between BHB and clinical outcomes. Over 6 months, 404 patients with diabetes had at least one beta-hydroxybutyrate level measured in the emergency department. There were 23 admissions for diabetic ketoacidosis (DKA) or hyperosmolar state. Of the remainder, 58 patients had a beta-hydroxybutyrate ≥ 1 mmol/L; this group had a higher glucose at presentation (19.0 (8.8) versus 10.4 (9.9) mmol/L), higher HbA1c (8.8 (5.4) versus 8.0 (3.3)%), lower bicarbonate (22.6 (6.2) versus 24.8 (4.7) mmol/L), and higher anion gap (14.8 (6.1) versus 12.6 (4.2)) than had those with BHB < 1 mmol/L. There was no association between the presence of ketosis and the length of stay (4.2 (7.3) versus (3.0) (7.2) days). Acute illness in those with diabetes associated with ketosis in the absence of DKA is associated with worse glycaemic control than in those without ketosis. Ketosis may represent an intermediate state of metabolic dysregulation rather than being associated with a more severe acute illness, as suggested by no relationship between BHB and length of stay.
Collapse
Affiliation(s)
- Barbara Depczynski
- Endocrinology Department, Prince of Wales Hospital, Randwick, NSW, Australia
| | | | - Wayne Varndell
- Emergency Department, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Angela L. Chiew
- Emergency Department, Prince of Wales Hospital, Randwick, NSW, Australia
| |
Collapse
|
889
|
Kovács Z, D'Agostino DP, Diamond D, Kindy MS, Rogers C, Ari C. Therapeutic Potential of Exogenous Ketone Supplement Induced Ketosis in the Treatment of Psychiatric Disorders: Review of Current Literature. Front Psychiatry 2019; 10:363. [PMID: 31178772 PMCID: PMC6543248 DOI: 10.3389/fpsyt.2019.00363] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Globally, psychiatric disorders, such as anxiety disorder, bipolar disorder, schizophrenia, depression, autism spectrum disorder, and attention-deficit/hyperactivity disorder (ADHD) are becoming more prevalent. Although the exact pathological alterations are not yet clear, recent studies have demonstrated that widespread changes of very complex metabolic pathways may partially underlie the pathophysiology of many psychiatric diseases. Thus, more attention should be directed to metabolic-based therapeutic interventions in the treatment of psychiatric disorders. Emerging evidence from numerous studies suggests that administration of exogenous ketone supplements, such as ketone salts or ketone esters, generates rapid and sustained nutritional ketosis and metabolic changes, which may evoke potential therapeutic effects in cases of central nervous system (CNS) disorders, including psychiatric diseases. Therefore, the aim of this review is to summarize the current information on ketone supplementation as a potential therapeutic tool for psychiatric disorders. Ketone supplementation elevates blood levels of the ketone bodies: D-β-hydroxybutyrate (βHB), acetoacetate (AcAc), and acetone. These compounds, either directly or indirectly, beneficially affect the mitochondria, glycolysis, neurotransmitter levels, activity of free fatty acid receptor 3 (FFAR3), hydroxycarboxylic acid receptor 2 (HCAR2), and histone deacetylase, as well as functioning of NOD-like receptor pyrin domain 3 (NLRP3) inflammasome and mitochondrial uncoupling protein (UCP) expression. The result of downstream cellular and molecular changes is a reduction in the pathophysiology associated with various psychiatric disorders. We conclude that supplement-induced nutritional ketosis leads to metabolic changes and improvements, for example, in mitochondrial function and inflammatory processes, and suggest that development of specific adjunctive ketogenic protocols for psychiatric diseases should be actively pursued.
Collapse
Affiliation(s)
- Zsolt Kovács
- Savaria Department of Biology, ELTE Eötvös Loránd University, Savaria University Centre, Szombathely, Hungary
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Ocala, FL, United States
| | - David Diamond
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Psychology, Hyperbaric Neuroscience Research Laboratory, University of South Florida, Tampa, FL, United States
| | - Mark S Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States.,James A. Haley VA Medical Center, Tampa, FL, United States.,Shriners Hospital for Children, Tampa, FL, United States
| | - Christopher Rogers
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Csilla Ari
- Department of Psychology, Hyperbaric Neuroscience Research Laboratory, University of South Florida, Tampa, FL, United States
| |
Collapse
|
890
|
Chen L, Liu Q, Tang Q, Kuang J, Li H, Pu S, Wu T, Yang X, Li R, Zhang J, Zhang Z, Huang Y, Li Y, Zou M, Jiang W, Li T, Gong M, Zhang L, Wang H, Qu A, Xie W, He J. Hepatocyte-specific Sirt6 deficiency impairs ketogenesis. J Biol Chem 2018; 294:1579-1589. [PMID: 30530497 DOI: 10.1074/jbc.ra118.005309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/15/2018] [Indexed: 02/05/2023] Open
Abstract
Sirt6 is an NADH (NAD+)-dependent deacetylase with a critical role in hepatic lipid metabolism. Ketogenesis is controlled by a signaling network of hepatic lipid metabolism. However, how Sirt6 functions in ketogenesis remains unclear. Here, we demonstrated that Sirt6 functions as a mediator of ketogenesis in response to a fasting and ketogenic diet (KD). The KD-fed hepatocyte-specific Sirt6 deficiency (HKO) mice exhibited impaired ketogenesis, which was due to enhanced Fsp27 (fat-specific induction of protein 27), a protein known to regulate lipid metabolism. In contrast, overexpression of Sirt6 in mouse primary hepatocytes promoted ketogenesis. Mechanistically, Sirt6 repressed Fsp27β expression by interacting with Crebh (cAMP response element-binding protein H) and preventing its recruitment to the Fsp27β gene promoter. The KD-fed HKO mice also showed exacerbated hepatic steatosis and inflammation. Finally, Fsp27 silencing rescued hypoketonemia and other metabolic phenotypes in KD-fed HKO mice. Our data suggest that the Sirt6-Crebh-Fsp27 axis is pivotal for hepatic lipid metabolism and inflammation. Sirt6 may be a pharmacological target to remedy metabolic diseases.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qin Tang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiangying Kuang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyun Pu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuping Yang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinhang Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zijing Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ya Huang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Jiang
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Li
- West China-Washington Mitochondria and Metabolism Center and Laboratory of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Gong
- West China-Washington Mitochondria and Metabolism Center and Laboratory of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Zhang
- West China-Washington Mitochondria and Metabolism Center and Laboratory of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hua Wang
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China 100069
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
891
|
Klos M, Morgenstern S, Hicks K, Suresh S, Devaney EJ. The effects of the ketone body β-hydroxybutyrate on isolated rat ventricular myocyte excitation-contraction coupling. Arch Biochem Biophys 2018; 662:143-150. [PMID: 30543786 DOI: 10.1016/j.abb.2018.11.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/06/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
β-hydroxybutyrate is the primary ketone body produced by the body during ketosis and is used to meet its metabolic demands. The healthy adult heart derives most of its energy from fatty acid oxidation. However, in certain diseases, the heart alters its substrate preference and increases its ketone body metabolism. Little is known about the effects of βOHB on ventricular myocyte excitation-contraction coupling. Therefore, we examined the effects of ketone body metabolism on single cell excitation-contraction coupling during normoxic and hypoxic conditions. Myocytes were isolated from adult rats, cultured for 18 h in RPMI 1640, RPMI 1640 no glucose, and M199, HEPES with/without various amount of βOHB added. To simulate hypoxia, myocytes were incubated at 1%O2, 5% CO2 for 1 h followed by incubation at atmospheric oxygen (21%O2,5% CO2) for 30 min before recordings. Recordings were obtained using an IonOptix system at 36±1ᵒ C. Myocytes were paced at 0.5, 1, 2, 3, and 4 Hz. We found that exposure to βOHB had no effect on excitation-contraction coupling. However, culturing cells with βOHB results in a significant increase in both contraction and calcium in RPMI 1640 media. Dose response experiments demonstrated 0.5 mM βOHB is enough to increase myocyte contraction in the absence of glucose. However, βOHB has no measurable effects on myocytes cultured in a nutrient rich media, M199, HEPES. Therefore, βOHB improves single cell excitation-contraction coupling, is protective against hypoxia, and may be a beneficial adaptation for the heart during periods of nutrient scarcity and or metabolic dysregulation.
Collapse
Affiliation(s)
- Matthew Klos
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA; Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Sherry Morgenstern
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA
| | - Kayla Hicks
- Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Shreyas Suresh
- Case Western Reserve University School of Graduate Studies, Cleveland, OH, 44106, USA
| | - Eric J Devaney
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA; Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
892
|
Shimizu K, Saito H, Sumi K, Sakamoto Y, Tachi Y, Iida K. Short-term and long-term ketogenic diet therapy and the addition of exercise have differential impacts on metabolic gene expression in the mouse energy-consuming organs heart and skeletal muscle. Nutr Res 2018; 60:77-86. [DOI: 10.1016/j.nutres.2018.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 09/06/2018] [Accepted: 09/14/2018] [Indexed: 11/24/2022]
|
893
|
Puchalska P, Huang X, Martin SE, Han X, Patti GJ, Crawford PA. Isotope Tracing Untargeted Metabolomics Reveals Macrophage Polarization-State-Specific Metabolic Coordination across Intracellular Compartments. iScience 2018; 9:298-313. [PMID: 30448730 PMCID: PMC6240706 DOI: 10.1016/j.isci.2018.10.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
We apply stable isotope tracing, mass-spectrometry-based untargeted metabolomics, to reveal the biochemical space labeled by 13C-substrates in bone-marrow-derived macrophages. At the pathway level, classically (lipopolysaccharide [LPS]-polarized, M1) and alternatively (interleukin [IL]-4-polarized, M2) polarized macrophages were 13C-labeled with surprising concordance. Total pools of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), an intermediate in the hexosamine biosynthetic pathway, were equally abundant in LPS- and IL-4-polarized macrophages. Informatic scrutiny of 13C-isotopologues revealed that LPS-polarized macrophages leverage the pentose phosphate pathway to generate UDP-GlcNAc, whereas IL-4-polarized macrophages rely on intact glucose and mitochondrial metabolism of glucose carbon. Labeling from [13C]glucose is competed by unlabeled fatty acids and acetoacetate, underscoring the broad roles for substrate metabolism beyond energy conversion. Finally, the LPS-polarized macrophage metabolite itaconate is imported into IL-4-polarized macrophages, in which it reprograms [13C]glucose metabolism. Thus, use of fully unsupervised isotope tracing metabolomics in macrophages reveals polarization-state-specific metabolic pathway connectivity, substrate competition, and metabolite allocation among cellular compartments.
Collapse
Affiliation(s)
- Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Xiaojing Huang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Chemistry, Washington University, St. Louis, MO 63110, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shannon E Martin
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Barshop Institute for Longevity and Aging Studies, Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63110, USA
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
894
|
Abstract
Nutrient composition and caloric intake have traditionally been used to devise optimized diets for various phases of life. Adjustment of meal size and frequency have emerged as powerful tools to ameliorate and postpone the onset of disease and delay aging, whereas periods of fasting, with or without reduced energy intake, can have profound health benefits. The underlying physiological processes involve periodic shifts of metabolic fuel sources, promotion of repair mechanisms, and the optimization of energy utilization for cellular and organismal health. Future research endeavors should be directed to the integration of a balanced nutritious diet with controlled meal size and patterns and periods of fasting to develop better strategies to prevent, postpone, and treat the socioeconomical burden of chronic diseases associated with aging.
Collapse
Affiliation(s)
- Andrea Di Francesco
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Clara Di Germanio
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
895
|
Arima Y, Izumiya Y, Ishida T, Takashio S, Ishii M, Sueta D, Fujisue K, Sakamoto K, Kaikita K, Tsujita K. Myocardial Ischemia Suppresses Ketone Body Utilization. J Am Coll Cardiol 2018; 73:246-247. [PMID: 30408507 DOI: 10.1016/j.jacc.2018.10.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/17/2018] [Accepted: 10/28/2018] [Indexed: 02/07/2023]
|
896
|
Erickson HL, Anakk S. Identification of IQ motif-containing GTPase-activating protein 1 as a regulator of long-term ketosis. JCI Insight 2018; 3:99866. [PMID: 30385723 DOI: 10.1172/jci.insight.99866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022] Open
Abstract
IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffolding protein that integrates multiple cellular processes, including motility, adhesion, and proliferation, but its role in metabolism is unknown. Here, we show that IQGAP1 is induced upon fasting and regulates β-oxidation of fatty acids and synthesis of ketone bodies in the liver. IQGAP1-null (Iqgap1-/-) mice exhibit reduced hepatic PPARα transcriptional activity, as evidenced during fasting, after ketogenic diet, and upon pharmacological activation. Conversely, we found that the activity of fed-state sensor mTORC1 is enhanced in Iqgap1-/- livers, but acute inhibition of mTOR in Iqgap1-/- mice was unable to rescue the defect in ketone body synthesis. However, reexpressing IQGAP1 in the livers of Iqgap1-/- mice was sufficient to promote ketone body synthesis, increase PPARα signaling, and suppress mTORC1 activity. Taken together, we uncover what we believe to be a previously unidentified role for IQGAP1 in regulating PPARα activity and ketogenesis.
Collapse
|
897
|
Güntner AT, Kompalla JF, Landis H, Theodore SJ, Geidl B, Sievi NA, Kohler M, Pratsinis SE, Gerber PA. Guiding Ketogenic Diet with Breath Acetone Sensors. SENSORS (BASEL, SWITZERLAND) 2018; 18:E3655. [PMID: 30373291 PMCID: PMC6264102 DOI: 10.3390/s18113655] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022]
Abstract
Ketogenic diet (KD; high fat, low carb) is a standard treatment for obesity, neurological diseases (e.g., refractory epilepsy) and a promising method for athletes to improve their endurance performance. Therein, the level of ketosis must be regulated tightly to ensure an effective therapy. Here, we introduce a compact and inexpensive breath sensor to monitor ketosis online and non-invasively. The sensor consists of Si-doped WO₃ nanoparticles that detect breath acetone selectively with non-linear response characteristics in the relevant range of 1 to 66 ppm, as identified by mass spectrometry. When tested on eleven subjects (five women and six men) undergoing a 36-h KD based on the Johns Hopkins protocol, this sensor clearly recognizes the onset and progression of ketosis. This is in good agreement to capillary blood β-hydroxybutyrate (BOHB) measurements. Despite similar dieting conditions, strong inter-subject differences in ketosis dynamics were observed and correctly identified by the sensor. These even included breath acetone patterns that could be linked to low tolerance to that diet. As a result, this portable breath sensor represents an easily applicable and reliable technology to monitor KD, possibly during medical treatment of epilepsy and weight loss.
Collapse
Affiliation(s)
- Andreas T Güntner
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| | - Julia F Kompalla
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| | - Henning Landis
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| | - S Jonathan Theodore
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| | - Bettina Geidl
- Department of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland.
| | - Noriane A Sievi
- Department of Pulmonology, University Hospital Zurich, CH-8091 Zurich, Switzerland.
| | - Malcolm Kohler
- Department of Pulmonology, University Hospital Zurich, CH-8091 Zurich, Switzerland.
| | - Sotiris E Pratsinis
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| | - Philipp A Gerber
- Department of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, CH-8091 Zurich, Switzerland.
| |
Collapse
|
898
|
Crawford PA. Refueling the Failing Heart: A Case for Sodium-Glucose Cotransporter 2 Inhibition in Cardiac Energy Homeostasis. JACC Basic Transl Sci 2018; 3:588-590. [PMID: 30456330 PMCID: PMC6234513 DOI: 10.1016/j.jacbts.2018.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
899
|
Thomsen HH, Rittig N, Johannsen M, Møller AB, Jørgensen JO, Jessen N, Møller N. Effects of 3-hydroxybutyrate and free fatty acids on muscle protein kinetics and signaling during LPS-induced inflammation in humans: anticatabolic impact of ketone bodies. Am J Clin Nutr 2018; 108:857-867. [PMID: 30239561 DOI: 10.1093/ajcn/nqy170] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 06/29/2018] [Indexed: 12/29/2022] Open
Abstract
Background Acute inflammation, and subsequent release of bacterial products (e.g. LPS), inflammatory cytokines, and stress hormones, is catabolic, and the loss of lean body mass predicts morbidity and mortality. Lipid intermediates may reduce protein loss, but the roles of free fatty acids (FFAs) and ketone bodies during acute inflammation are unclear. Objective We aimed to test whether infusions of 3-hydroxybutyrate (3OHB), FFAs, and saline reduce protein catabolism during exposure to LPS and Acipimox (to restrict and control endogenous lipolysis). Design A total of 10 healthy male subjects were randomly tested 3 times, with: 1) LPS, Acipimox (Olbetam) and saline, 2) LPS, Acipimox, and nonesterified fatty acids (Intralipid), and 3) LPS, Acipimox, and 3OHB, during a 5-h basal period and a 2-h hyperinsulinemic, euglycemic clamp. Labeled phenylalanine, tyrosine, and urea tracers were used to estimate protein kinetics, and muscle biopsies were taken for Western blot analysis of protein metabolic signaling. Results 3OHB infusion increased 3OHB concentrations (P < 0.0005) to 3.5 mM and decreased whole-body phenylalanine-to-tyrosine degradation. Basal and insulin-stimulated net forearm phenylalanine release decreased by >70% (P < 0.005), with both appearance and phenylalanine disappearance being profoundly decreased. Phosphorylation of eukaryotic initiation factor 2α at Ser51 was increased in skeletal muscle, and S6 kinase phosphorylation at Ser235/236 tended (P = 0.074) to be decreased with 3OHB infusion (suggesting inhibition of protein synthesis), whereas no detectable effects were seen on markers of protein breakdown. Lipid infusion did not affect phenylalanine kinetics, and insulin sensitivity was unaffected by interventions. Conclusion During acute inflammation, 3OHB has potent anticatabolic actions in muscle and at the whole-body level; in muscle, reduction of protein breakdown overrides inhibition of synthesis. This trial was registered at clinicaltrials.gov as NCT01752348.
Collapse
Affiliation(s)
- Henrik H Thomsen
- Department of Medicine, Viborg Regional Hospital, Viborg, Denmark.,Department of Internal Medicine and Endocrinology MEA, Aarhus University, Aarhus, Denmark
| | - Nikolaj Rittig
- Department of Internal Medicine and Endocrinology MEA, Aarhus University, Aarhus, Denmark
| | - Mogens Johannsen
- Department of Forensic Medicine, Bioanalytical Unit, Aarhus University, Aarhus, Denmark
| | - Andreas B Møller
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Otto Jørgensen
- Department of Internal Medicine and Endocrinology MEA, Aarhus University, Aarhus, Denmark
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Clinical Pharmacology, Aarhus University, Aarhus, Denmark
| | - Niels Møller
- Department of Internal Medicine and Endocrinology MEA, Aarhus University, Aarhus, Denmark
| |
Collapse
|
900
|
Sangalli JR, Sampaio RV, Del Collado M, da Silveira JC, De Bem THC, Perecin F, Smith LC, Meirelles FV. Metabolic gene expression and epigenetic effects of the ketone body β-hydroxybutyrate on H3K9ac in bovine cells, oocytes and embryos. Sci Rep 2018; 8:13766. [PMID: 30214009 PMCID: PMC6137158 DOI: 10.1038/s41598-018-31822-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/29/2018] [Indexed: 12/16/2022] Open
Abstract
The rapid decline in fertility that has been occurring to high-producing dairy cows in the past 50 years seems to be associated with metabolic disturbances such as ketosis, supporting the need for research to improve our understanding of the relations among the diet, metabolism and embryonic development. Recently, the ketone body β-hydroxybutyrate (BOHB) was demonstrated to be a potent inhibitor of histone deacetylases (HDACs). Herein, we performed a series of experiments aiming to investigate the epigenetic effects of BOHB on histone acetylation in somatic cells, cumulus-oocyte complexes (COCs) and somatic cell nuclear transfer (SCNT) embryos. Treatment with BOHB does not increase histone acetylation in cells but stimulates genes associated with ketolysis and master regulators of metabolism. We further demonstrated that maturing COCs with high levels of BOHB does not affect their maturation rate or histone acetylation but increases the expression of PPARA in cumulus cells. Treatment of somatic cell nuclear transfer zygotes with BOHB causes hyperacetylation, which is maintained until the blastocyst stage, causing enhanced FOXO3A expression and blastocyst production. Our data shed light on the epigenetic mechanisms caused by BOHB in bovine cells and embryos and provide a better understanding of the connection between nutrition and reproduction.
Collapse
Affiliation(s)
- Juliano Rodrigues Sangalli
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil.
| | - Rafael Vilar Sampaio
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| | - Maite Del Collado
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| | - Juliano Coelho da Silveira
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| | - Tiago Henrique Camara De Bem
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| | - Felipe Perecin
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| | - Lawrence Charles Smith
- Université de Montréal, Faculté de médecine vétérinaire, Centre de recherche en reproduction et fertilité, St. Hyacinthe, Québec, postcode: H3T 1J4, Canada
| | - Flávio Vieira Meirelles
- University of Sao Paulo, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Pirassununga, Sao Paulo, postcode: 13635-900, Brazil
| |
Collapse
|