1
|
Wu Y, Chan AY, Hauke J, Htin Aung O, Foollee A, Cleofe MAS, Stölting H, Han ML, Jeppe KJ, Barlow CK, Okun JG, Rusu PM, Rose AJ. Variable glucagon metabolic actions in diverse mouse models of obesity and type 2 diabetes. Mol Metab 2024; 90:102064. [PMID: 39536823 PMCID: PMC11617456 DOI: 10.1016/j.molmet.2024.102064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The study aimed to investigate the effects of glucagon on metabolic pathways in mouse models of obesity, fatty liver disease, and type 2 diabetes (T2D) to determine the extent and variability of hepatic glucagon resistance in these conditions. METHODS We investigated glucagon's effects in mouse models of fatty liver disease, obesity, and type 2 diabetes (T2D), including male BKS-db/db, high-fat diet-fed, and western diet-fed C57Bl/6 mice. Glucagon tolerance tests were performed using the selective glucagon receptor agonist acyl-glucagon (IUB288). Blood glucose, serum and liver metabolites include lipids and amino acids were measured. Additionally, liver protein expression related to glucagon signalling and a comprehensive liver metabolomics were performed. RESULTS Western diet-fed mice displayed impaired glucagon response, with reduced blood glucose and PKA activation. In contrast, high-fat diet-fed and db/db mice maintained normal glucagon sensitivity, showing significant elevations in blood glucose and phospho-PKA motif protein expression. Acyl-glucagon treatment also lowered liver alanine and histidine levels in high-fat diet-fed mice, but not in western diet-fed mice. Additionally, some amino acids, such as methionine, were increased by acyl-glucagon only in chow diet control mice. Despite normal glucagon sensitivity in PKA signalling, db/db mice had a distinct metabolomic response, with acyl-glucagon significantly altering 90 metabolites in db/+ mice but only 42 in db/db mice, and classic glucagon-regulated metabolites, such as cyclic adenosine monophosphate (cAMP), being less responsive in db/db mice. CONCLUSIONS The study reveals that hepatic glucagon resistance in obesity and T2D is complex and not uniform across metabolic pathways, underscoring the complexity of glucagon action in these conditions.
Collapse
Affiliation(s)
- Yuqin Wu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Andrea Y Chan
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Jana Hauke
- Division of Inherited Metabolic Diseases, University Children's Hospital, 69120 Heidelberg, Germany
| | - Okka Htin Aung
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Maria Almira S Cleofe
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Helen Stölting
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Mei-Ling Han
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Katherine J Jeppe
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, 69120 Heidelberg, Germany
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
2
|
Ait Tayeb AEK, Colle R, Chappell K, El-Asmar K, Acquaviva-Bourdain C, David DJ, Trabado S, Chanson P, Feve B, Becquemont L, Verstuyft C, Corruble E. Metabolomic profiles of 38 acylcarnitines in major depressive episodes before and after treatment. Psychol Med 2024; 54:289-298. [PMID: 37226550 DOI: 10.1017/s003329172300140x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
BACKGROUND Major depression is associated with changes in plasma L-carnitine and acetyl-L-carnitine. But its association with acylcarnitines remains unclear. The aim of this study was to assess metabolomic profiles of 38 acylcarnitines in patients with major depression before and after treatment compared to healthy controls (HCs). METHODS Metabolomic profiles of 38 plasma short-, medium-, and long-chain acylcarnitines were performed by liquid chromatography-mass spectrometry in 893 HCs from the VARIETE cohort and 460 depressed patients from the METADAP cohort before and after 6 months of antidepressant treatment. RESULTS As compared to HCs, depressed patients had lower levels of medium- and long-chain acylcarnitines. After 6 months of treatment, increased levels of medium- and long-chain acyl-carnitines were observed that no longer differed from those of controls. Accordingly, several medium- and long-chain acylcarnitines were negatively correlated with depression severity. CONCLUSIONS These medium- and long-chain acylcarnitine dysregulations argue for mitochondrial dysfunction through fatty acid β-oxidation impairment during major depression.
Collapse
Affiliation(s)
- Abd El Kader Ait Tayeb
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, Paris, F-94275, France
| | - Romain Colle
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, Paris, F-94275, France
| | - Kenneth Chappell
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
| | - Khalil El-Asmar
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Department of Epidemiology and Population Health, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Cécile Acquaviva-Bourdain
- Service de Biochimie et Biologie Moléculaire; Unité Médicale Pathologies Héréditaires du Métabolisme et du Globule Rouge; Centre de Biologie et Pathologie Est; CHU de Lyon; F-69500 Bron, France
| | - Denis J David
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
| | - Séverine Trabado
- INSERM UMR-S U1185, Physiologie et Physiopathologie Endocriniennes, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Philippe Chanson
- INSERM UMR-S U1185, Physiologie et Physiopathologie Endocriniennes, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l'Hypophyse, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Bruno Feve
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire ICAN, Service d'Endocrinologie, CRMR PRISIS, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris, F-75012, France
| | - Laurent Becquemont
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Centre de recherche clinique, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, Paris, F-94275, France
| | - Céline Verstuyft
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, Paris, F-94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, Paris, F-94275, France
| |
Collapse
|
3
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Biol Chem 2021; 297:101278. [PMID: 34717954 PMCID: PMC8605089 DOI: 10.1016/j.jbc.2021.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/21/2022] Open
|
4
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Lipid Res 2021; 62:100132. [PMID: 34717951 PMCID: PMC8721491 DOI: 10.1016/j.jlr.2021.100132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/05/2022] Open
|
5
|
Odo S, Tanabe K, Yohda M, Yamauchi M. Liver-Oriented Acute Metabolic Effects of A Low Dose of L-Carnitine under Fat-Mobilizing Conditions: Pilot Human Clinical Trial. J Nutr Sci Vitaminol (Tokyo) 2021; 66:136-149. [PMID: 32350175 DOI: 10.3177/jnsv.66.136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The acute metabolic effect of low dosages of L-carnitine under fat-mobilizing conditions was investigated. Healthy subjects (Study 1: n=5; Study 2: n=6) were asked to fast overnight. Then, 30 min of aerobic exercise on a cycle ergometer was performed after supplementation, followed by a 3.5-h sedentary recovery phase. The following ingestion patterns were used: Study 1 (i) noningestion, (ii) 750 mg of L-carnitine (LC), and (iii) 750 mg of LC+50 g of carbohydrate (CHO); Study 2 (iv) noningestion, (v) 500 mg of LC, (vi) 30 mg of CoQ10, and (vii) 500 mg of LC+30 mg of CoQ10. The energy expenditure (EE) and nonprotein respiratory quotient (npRQ) were measured during the pre-exercise, postexercise, and recovery periods. Serum free carnitine, acetylcarnitine, total carnitine (Study 1 and 2), and ketone bodies (Study 2) were measured. The 750 mg LC treatment significantly facilitated fat oxidation during the recovery phases (p<0.05) without elevating EE. The higher fat oxidation associated with LC was completely suppressed by CHO. CoQ10 affected neither npRQ nor EE. npRQ was significantly correlated with the serum total ketone bodies (R=-0.68, p<0.001) and acetylcarnitine (R=-0.61--0.70, p<0.001). The highest correlation was found between acetylcarnitine and total ketone bodies immediately after exercise (R=0.85, p<0.001). In conclusion, LC enhanced liver fat utilization and ketogenesis in an acute manner without stimulating EE under fat-mobilizing conditions.
Collapse
Affiliation(s)
- Satoshi Odo
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology.,Lonza Japan Ltd
| | | | - Masafumi Yohda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology
| | | |
Collapse
|
6
|
Savic D, Hodson L, Neubauer S, Pavlides M. The Importance of the Fatty Acid Transporter L-Carnitine in Non-Alcoholic Fatty Liver Disease (NAFLD). Nutrients 2020; 12:E2178. [PMID: 32708036 PMCID: PMC7469009 DOI: 10.3390/nu12082178] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
L-carnitine transports fatty acids into the mitochondria for oxidation and also buffers excess acetyl-CoA away from the mitochondria. Thus, L-carnitine may play a key role in maintaining liver function, by its effect on lipid metabolism. The importance of L-carnitine in liver health is supported by the observation that patients with primary carnitine deficiency (PCD) can present with fatty liver disease, which could be due to low levels of intrahepatic and serum levels of L-carnitine. Furthermore, studies suggest that supplementation with L-carnitine may reduce liver fat and the liver enzymes alanine aminotransferase (ALT) and aspartate transaminase (AST) in patients with Non-Alcoholic Fatty Liver Disease (NAFLD). L-carnitine has also been shown to improve insulin sensitivity and elevate pyruvate dehydrogenase (PDH) flux. Studies that show reduced intrahepatic fat and reduced liver enzymes after L-carnitine supplementation suggest that L-carnitine might be a promising supplement to improve or delay the progression of NAFLD.
Collapse
Affiliation(s)
- Dragana Savic
- Radcliffe Department of Medicine, Oxford Centre for Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK; (S.N.); (M.P.)
| | - Leanne Hodson
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford OX3 7LE, UK;
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Stefan Neubauer
- Radcliffe Department of Medicine, Oxford Centre for Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK; (S.N.); (M.P.)
| | - Michael Pavlides
- Radcliffe Department of Medicine, Oxford Centre for Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK; (S.N.); (M.P.)
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
7
|
Schlaepfer IR, Joshi M. CPT1A-mediated Fat Oxidation, Mechanisms, and Therapeutic Potential. Endocrinology 2020; 161:5695911. [PMID: 31900483 DOI: 10.1210/endocr/bqz046] [Citation(s) in RCA: 368] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022]
Abstract
Energy homeostasis during fasting or prolonged exercise depends on mitochondrial fatty acid oxidation (FAO). This pathway is crucial in many tissues with high energy demand and its disruption results in inborn FAO deficiencies. More than 15 FAO genetic defects have been currently described, and pathological variants described in circumpolar populations provide insights into its critical role in metabolism. The use of fatty acids as energy requires more than 2 dozen enzymes and transport proteins, which are involved in the activation and transport of fatty acids into the mitochondria. As the key rate-limiting enzyme of FAO, carnitine palmitoyltransferase I (CPT1) regulates FAO and facilitates adaptation to the environment, both in health and in disease, including cancer. The CPT1 family of proteins contains 3 isoforms: CPT1A, CPT1B, and CPT1C. This review focuses on CPT1A, the liver isoform that catalyzes the rate-limiting step of converting acyl-coenzyme As into acyl-carnitines, which can then cross membranes to get into the mitochondria. The regulation of CPT1A is complex and has several layers that involve genetic, epigenetic, physiological, and nutritional modulators. It is ubiquitously expressed in the body and associated with dire consequences linked with genetic mutations, metabolic disorders, and cancers. This makes CPT1A an attractive target for therapeutic interventions. This review discusses our current understanding of CPT1A expression, its role in heath and disease, and the potential for therapeutic opportunities targeting this enzyme.
Collapse
Affiliation(s)
- Isabel R Schlaepfer
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora
| | - Molishree Joshi
- University of Colorado School of Medicine, Department of Pharmacology, Aurora, Colorado
| |
Collapse
|
8
|
Ringseis R, Keller J, Eder K. Basic mechanisms of the regulation of L-carnitine status in monogastrics and efficacy of L-carnitine as a feed additive in pigs and poultry. J Anim Physiol Anim Nutr (Berl) 2018; 102:1686-1719. [PMID: 29992642 DOI: 10.1111/jpn.12959] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/19/2022]
Abstract
A great number of studies have investigated the potential of L-carnitine as feed additive to improve performance of different monogastric and ruminant livestock species, with, however, discrepant outcomes. In order to understand the reasons for these discrepant outcomes, it is important to consider the determinants of L-carnitine status and how L-carnitine status is regulated in the animal's body. While it is a long-known fact that L-carnitine is endogenously biosynthesized in certain tissues, it was only recently recognized that critical determinants of L-carnitine status, such as intestinal L-carnitine absorption, tissue L-carnitine uptake, endogenous L-carnitine synthesis and renal L-carnitine reabsorption, are regulated by specific nutrient sensing nuclear receptors. This review aims to give a more in-depth understanding of the basic mechanisms of the regulation of L-carnitine status in monogastrics taking into account the most recent evidence on nutrient sensing nuclear receptors and evaluates the efficacy of L-carnitine as feed additive in monogastric livestock by providing an up-to-date overview about studies with L-carnitine supplementation in pigs and poultry.
Collapse
Affiliation(s)
- Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Janine Keller
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
9
|
Servillo L, D'Onofrio N, Neglia G, Casale R, Cautela D, Marrelli M, Limone A, Campanile G, Balestrieri ML. Carnitine Precursors and Short-Chain Acylcarnitines in Water Buffalo Milk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:8142-8149. [PMID: 30011990 DOI: 10.1021/acs.jafc.8b02963] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ruminants' milk contains δ-valerobetaine originating from rumen through the transformation of dietary Nε-trimethyllysine. Among ruminant's milk, the occurrence of δ-valerobetaine, along with carnitine precursors and metabolites, has not been investigated in buffalo milk, the second most worldwide consumed milk, well-known for its nutritional value. HPLC-ESI-MS/MS analyses of bulk milk revealed that the Italian Mediterranean buffalo milk contains δ-valerobetaine at levels higher than those in bovine milk. Importantly, we detected also γ-butyrobetaine, the l-carnitine precursor, never described so far in any milk. Of interest, buffalo milk shows higher levels of acetylcarnitine, propionylcarnitine, butyrylcarnitine, isobutyrylcarnitine, and 3-methylbutyrylcarnitine (isovalerylcarnitine) than cow milk. Moreover, buffalo milk shows isobutyrylcarnitine and butyrylcarnitine at a 1-to-1 molar ratio, while in cow's milk this ratio is 5 to 1. Results indicate a peculiar short-chain acylcarnitine profile characterizing buffalo milk, widening the current knowledge about its composition and nutritional value.
Collapse
Affiliation(s)
- Luigi Servillo
- Department of Precision Medicine , University of Campania "L. Vanvitelli" , 80138 Naples , Italy
| | - Nunzia D'Onofrio
- Department of Precision Medicine , University of Campania "L. Vanvitelli" , 80138 Naples , Italy
| | - Gianluca Neglia
- Department of Veterinary Medicine and Animal Production , Federico II University , 80137 Naples , Italy
| | - Rosario Casale
- Department of Precision Medicine , University of Campania "L. Vanvitelli" , 80138 Naples , Italy
| | - Domenico Cautela
- Stazione Sperimentale per le Industrie delle Essenze e dei derivati dagli Agrumi , Azienda Speciale della Camera di Commercio di Reggio Calabria , 89125 Reggio Calabria , Italy
| | - Massimo Marrelli
- Maxillofacial Surgery Section , Marrelli Health , 88900 Crotone , Italy
| | - Antonio Limone
- Istituto Zooprofilattico Sperimentale del Mezzogiorno , 80055 Naples , Italy
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production , Federico II University , 80137 Naples , Italy
| | - Maria Luisa Balestrieri
- Department of Precision Medicine , University of Campania "L. Vanvitelli" , 80138 Naples , Italy
| |
Collapse
|
10
|
Ringseis R, Keller J, Eder K. Regulation of carnitine status in ruminants and efficacy of carnitine supplementation on performance and health aspects of ruminant livestock: a review. Arch Anim Nutr 2018; 72:1-30. [PMID: 29313385 DOI: 10.1080/1745039x.2017.1421340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Carnitine has long been known to play a critical role for energy metabolism. Due to this, a large number of studies have been carried out to investigate the potential of supplemental carnitine in improving performance of livestock animals including ruminants, with however largely inconsistent results. An important issue that has to be considered when using carnitine as a feed additive is that the efficacy of supplemental carnitine is probably dependent on the animal's carnitine status, which is affected by endogenous carnitine synthesis, carnitine uptake from the gastrointestinal tract and carnitine excretion. The present review aims to summarise the current knowledge of the regulation of carnitine status and carnitine homeostasis in ruminants, and comprehensively evaluate the efficacy of carnitine supplementation on performance and/or health in ruminant livestock by comparing the outcomes of studies with carnitine supplementation in dairy cattle, growing and finishing cattle and sheep. While most of the studies show that supplemental carnitine, even in ruminally unprotected form, is bioavailable in ruminants, its effect on either milk or growth performance is largely disappointing. However, supplemental carnitine appears to be a useful strategy to offer protection against ammonia toxicity caused by consumption of high levels of non-protein N or forages with high levels of soluble N both, in cattle and sheep.
Collapse
Affiliation(s)
- Robert Ringseis
- a Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Gießen , Gießen , Germany
| | - Janine Keller
- a Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Gießen , Gießen , Germany
| | - Klaus Eder
- a Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Gießen , Gießen , Germany
| |
Collapse
|
11
|
Humer E, Khol-Parisini A, Metzler-Zebeli BU, Gruber L, Zebeli Q. Alterations of the Lipid Metabolome in Dairy Cows Experiencing Excessive Lipolysis Early Postpartum. PLoS One 2016; 11:e0158633. [PMID: 27383746 PMCID: PMC4934687 DOI: 10.1371/journal.pone.0158633] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/20/2016] [Indexed: 11/18/2022] Open
Abstract
A decrease in insulin sensitivity enhances adipose tissue lipolysis helping early lactation cows counteracting their energy deficit. However, excessive lipolysis poses serious health risks for cows, and its underlying mechanisms are not clearly understood. The present study used targeted ESI-LC-MS/MS-based metabolomics and indirect insulin sensitivity measurements to evaluate metabolic alterations in the serum of dairy cows of various parities experiencing variable lipolysis early postpartum. Thirty (12 primiparous and 18 multiparous) cows of Holstein Friesian and Simmental breeds, fed the same diet and kept under the same management conditions, were sampled at d 21 postpartum and classified as low (n = 10), medium (n = 8), and high (n = 12) lipolysis groups, based on serum concentration of nonesterified fatty acids. Overall, excessive lipolysis in the high group came along with impaired estimated insulin sensitivity and characteristic shifts in acylcarnitine, sphingomyelin, phosphatidylcholine and lysophospholipid metabolome profiles compared to the low group. From the detected phosphatidylcholines mainly those with diacyl-residues showed differences among lipolysis groups. Furthermore, more than half of the detected sphingomyelins were increased in cows experiencing high lipomobilization. Additionally, strong differences in serum acylcarnitines were noticed among lipolysis groups. The study suggests an altered serum phospholipidome in dairy cows associated with an increase in certain long-chain sphingomyelins and the progression of disturbed insulin function. In conclusion, the present study revealed 37 key metabolites as part of alterations in the synthesis or breakdown of sphingolipids and phospholipids associated with lowered estimated insulin sensitivity and excessive lipolysis in early-lactating cows.
Collapse
Affiliation(s)
- Elke Humer
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Annabella Khol-Parisini
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara U. Metzler-Zebeli
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Leonhard Gruber
- Institute of Livestock Research, Agricultural Research and Education Centre Raumberg-Gumpenstein, Irdning, Austria
| | - Qendrim Zebeli
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
12
|
Ciavardelli D, Piras F, Consalvo A, Rossi C, Zucchelli M, Di Ilio C, Frazzini V, Caltagirone C, Spalletta G, Sensi SL. Medium-chain plasma acylcarnitines, ketone levels, cognition, and gray matter volumes in healthy elderly, mildly cognitively impaired, or Alzheimer's disease subjects. Neurobiol Aging 2016; 43:1-12. [PMID: 27255810 DOI: 10.1016/j.neurobiolaging.2016.03.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 02/02/2016] [Accepted: 03/06/2016] [Indexed: 12/21/2022]
Abstract
Aging, amyloid deposition, and tau-related pathology are key contributors to the onset and progression of Alzheimer's disease (AD). However, AD is also associated with brain hypometabolism and deficits of mitochondrial bioenergetics. Plasma acylcarnitines (ACCs) are indirect indices of altered fatty acid beta-oxidation, and ketogenesis has been found to be decreased on aging. Furthermore, in elderly subjects, alterations in plasma levels of specific ACCs have been suggested to predict conversion to mild cognitive impairment (MCI) or AD. In this study, we assayed plasma profiles of ACCs in a cohort of healthy elderly control, MCI subjects, and AD patients. Compared with healthy controls or MCI subjects, AD patients showed significant lower plasma levels of several medium-chain ACCs. Furthermore, in AD patients, these lower concentrations were associated with lower prefrontal gray matter volumes and the presence of cognitive impairment. Interestingly, lower levels of medium-chain ACCs were also found to be associated with lower plasma levels of 2-hydroxybutyric acid. Overall, these findings suggest that altered metabolism of medium-chain ACCs and impaired ketogenesis can be metabolic features of AD.
Collapse
Affiliation(s)
- Domenico Ciavardelli
- School of Human and Social Science, "Kore" University of Enna, Enna, Italy; Molecular Neurology Unit, Center of Excellence on Aging and Translational Medicine (Ce.S.I.-MeT), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Fabrizio Piras
- Department of Clinical and Behavioral Neurology, Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; "Enrico Fermi" Centre for Study and Research, Rome, Italy
| | - Ada Consalvo
- Department of Medical, Oral, and Biotechnological Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Claudia Rossi
- Department of Medical, Oral, and Biotechnological Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Mirco Zucchelli
- Department of Medical, Oral, and Biotechnological Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Carmine Di Ilio
- Department of Medical, Oral, and Biotechnological Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Valerio Frazzini
- Molecular Neurology Unit, Center of Excellence on Aging and Translational Medicine (Ce.S.I.-MeT), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Neuroscience, University "Tor Vergata", Rome, Italy
| | - Gianfranco Spalletta
- Department of Clinical and Behavioral Neurology, Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Stefano L Sensi
- Molecular Neurology Unit, Center of Excellence on Aging and Translational Medicine (Ce.S.I.-MeT), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Neurology, and Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, USA; Department of Pharmacology, and Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
13
|
Isolation of soluble scFv antibody fragments specific for small biomarker molecule, L-Carnitine, using phage display. J Immunol Methods 2015; 428:9-19. [PMID: 26608419 DOI: 10.1016/j.jim.2015.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 10/23/2015] [Accepted: 11/18/2015] [Indexed: 11/22/2022]
Abstract
Isolation of single chain antibody fragment (scFv) clones from naïve Tomlinson I+J phage display libraries that specifically bind a small biomarker molecule, L-Carnitine, was performed using iterative affinity selection procedures. L-Carnitine has been described as a conditionally essential nutrient for humans. Abnormally high concentrations of L-Carnitine in urine are related to many health disorders including diabetes mellitus type 2 and lung cancer. ELISA-based affinity characterization results indicate that selectants preferentially bind to L-Carnitine in the presence of key bioselecting component materials and closely related L-Carnitine derivatives. In addition, the affinity results were confirmed using biophysical fluorescence quenching for tyrosine residues in the V segment. Small-scale production of the soluble fragment yielded 1.3mg/L using immunopure-immobilized protein A affinity column. Circular Dichroism data revealed that the antibody fragment (Ab) represents a folded protein that mainly consists of β-sheets. These novel antibody fragments may find utility as molecular affinity interface receptors in various electrochemical biosensor platforms to provide specific L-Carnitine binding capability with potential applications in metabolomic devices for companion diagnostics and personalized medicine applications. It may also be used in any other biomedical application where detection of the L-Carnitine level is important.
Collapse
|
14
|
McCarty MF, DiNicolantonio JJ, O’Keefe JH. Ketosis may promote brain macroautophagy by activating Sirt1 and hypoxia-inducible factor-1. Med Hypotheses 2015; 85:631-9. [PMID: 26306884 DOI: 10.1016/j.mehy.2015.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/06/2015] [Accepted: 08/01/2015] [Indexed: 12/25/2022]
|
15
|
Luo H, Zhang Y, Guo H, Zhang L, Li X, Ringseis R, Wen G, Hui D, Liang A, Eder K, He D. Transcriptional regulation of the human, porcine and bovine OCTN2 gene by PPARα via a conserved PPRE located in intron 1. BMC Genet 2014; 15:90. [PMID: 25299939 PMCID: PMC4363911 DOI: 10.1186/s12863-014-0090-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/06/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The novel organic cation transporter 2 (OCTN2) is the physiologically most important carnitine transporter in tissues and is responsible for carnitine absorption in the intestine, carnitine reabsorption in the kidney and distribution of carnitine between tissues. Genetic studies clearly demonstrated that the mouse OCTN2 gene is directly regulated by peroxisome proliferator-activated receptor α (PPARα). Despite its well conserved role as an important regulator of lipid catabolism in general, the specific genes under control of PPARα within each lipid metabolic pathway were shown to differ between species and it is currently unknown whether the OCTN2 gene is also a PPARα target gene in pig, cattle, and human. In the present study we examined the hypothesis that the porcine, bovine, and human OCTN2 gene are also PPARα target genes. RESULTS Using positional cloning and reporter gene assays we identified a functional PPRE, each in the intron 1 of the porcine, bovine, and human OCTN2 gene. Gel shift assay confirmed binding of PPARα to this PPRE in the porcine, bovine, and the human OCTN2 gene. CONCLUSIONS The results of the present study show that the porcine, bovine, and human OCTN2 gene, like the mouse OCTN2 gene, is directly regulated by PPARα. This suggests that regulation of genes involved in carnitine uptake by PPARα is highly conserved across species.
Collapse
Affiliation(s)
- Huidi Luo
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| | - Yuanqing Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| | - Huihui Guo
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| | - Li Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| | - Xi Li
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, 35392, Giessen, Germany.
| | - Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, 35392, Giessen, Germany.
| | - Dequan Hui
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, P. R. China.
| | - Aihua Liang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, P. R. China.
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, 35392, Giessen, Germany.
| | - Dongchang He
- Institute of Animal Husbandry and Veterinary Medicine, Shanxi Provincial Academy of Agricultural Sciences, Taiyuan, 030031, P. R. China.
| |
Collapse
|
16
|
Nassiri A, Dashti-Khavidaki S, Khalili H, Nassiri-Toosi M, Abdollahi A. Serum carnitine level and its associated factors in patients with chronic viral hepatitis. Future Virol 2014. [DOI: 10.2217/fvl.14.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT: Aim: Serum carnitine level and its associated factors have been evaluated in patients with chronic viral hepatitis. Methods: Patients with confirmed chronic viral hepatitis based on the serological markers and liver biopsy were included. In total, 86 volunteers and 86 patients with chronic viral hepatitis completed the study. Demographic data, type of treatment regimen and nutritional status of the patients were recorded and one blood sample was collected from each patient after an overnight fasting. A double antibody sandwich ELISA kit was used to measure carnitine serum level. Results: Mean ± standard deviation of serum carnitine level in the case and control groups were 34.3 ± 15.3 and 55.7 ± 28.4 μmol/l, respectively (p = 0.001). Regarding carnitine deficiency definition, 64 out of 86 patients (74.4%) and 21 out of 86 (24.5%) healthy individuals suffered from carnitine deficiency (p < 0.001). Carnitine dietary intake was significantly lower (p < 0.001). Compared with patients with chronic hepatitis C infection, a more severe form of carnitine deficiency was detected in patients with chronic hepatitis B infection (18.39 ± 15.68 μmol/l vs 42.30 ± 32.92 μmol/l; p = 0.03). In addition, serum carnitine level (41.1 ± 14.8 μmol/l) was significantly higher in the cirrhotic than noncirrhotic patients (31.60 ± 13.2 μmol/l; p = 0.04). Conclusion: Although the cirrhotic patients had higher serum carnitine level compared with noncirrhotic patients, serum carnitine level in the patients with chronic hepatitis was significantly lower than the healthy individuals. Also compared with the defined cut-off point for normal carnitine serum level, carnitine deficiency was common in Iranian patients with chronic hepatitis.
Collapse
Affiliation(s)
- Azin Nassiri
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khalili
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nassiri-Toosi
- Imam-Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Abdollahi
- Vali-E-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Ringseis R, Wen G, Eder K. Regulation of Genes Involved in Carnitine Homeostasis by PPARα across Different Species (Rat, Mouse, Pig, Cattle, Chicken, and Human). PPAR Res 2012; 2012:868317. [PMID: 23150726 PMCID: PMC3486131 DOI: 10.1155/2012/868317] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 09/27/2012] [Indexed: 11/17/2022] Open
Abstract
Recent studies in rodents convincingly demonstrated that PPARα is a key regulator of genes involved in carnitine homeostasis, which serves as a reasonable explanation for the phenomenon that energy deprivation and fibrate treatment, both of which cause activation of hepatic PPARα, causes a strong increase of hepatic carnitine concentration in rats. The present paper aimed to comprehensively analyse available data from genetic and animal studies with mice, rats, pigs, cows, and laying hens and from human studies in order to compare the regulation of genes involved in carnitine homeostasis by PPARα across different species. Overall, our comparative analysis indicates that the role of PPARα as a regulator of carnitine homeostasis is well conserved across different species. However, despite demonstrating a well-conserved role of PPARα as a key regulator of carnitine homeostasis in general, our comprehensive analysis shows that this assumption particularly applies to the regulation by PPARα of carnitine uptake which is obviously highly conserved across species, whereas regulation by PPARα of carnitine biosynthesis appears less well conserved across species.
Collapse
Affiliation(s)
- Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Heinrich-Buff-Ring 26-32, 35390 Giessen, Germany
| | - Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Heinrich-Buff-Ring 26-32, 35390 Giessen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Heinrich-Buff-Ring 26-32, 35390 Giessen, Germany
| |
Collapse
|
18
|
Effects of carnitine and its congeners on eicosanoid discharge from rat cells: implications for release of TNFalpha. Mediators Inflamm 2012; 2:S57-62. [PMID: 18475573 PMCID: PMC2365447 DOI: 10.1155/s0962935193000778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
THE acyl carrier coenzyme A (CoA) is involved in fatty acid metabolism. The carnitine/CoA ratio is of particular importance in regulating the transport of long-chain fatty acids into mitochondria for oxidation. Also CoA has a role in the formation and breakdown of products from both the cyclooxygenase and lipoxygenase pathways of the precursor arachidonic acid. In the present study the effect of 4 days feeding of 300 mg/kg/day of L-carnitine, acetyl Lcarnitine and propionyl L-carnitine on the basal and calcium ionophore (A23187) stimulated release of arachidonic acid metabolites from rat carrageenin elicited peritoneal cells was investigated. There were two series of experiments carried out. In the first, the harvested peritoneal cell population consisted of less than 90% macrophages and additional polymorphonuclear (PMN) leucocytes. The basal release of prostaglandin E2 (PGE2), 6-ketoprostaglandin F1α (6-keto-PGF1α) and leukotriene B4 (LTB4) was stimulated by all treatments. The A23187 stimulated release of 6-keto-PGF1α and LTB4 was increased by all three compounds. The 6-keto-PGF1α:TxB2 and 6-keto-PGF1α:LTB4 ratios were increased by carnitine treatment. These results suggested that carnitine could modify the macrophage component of an inflammatory site in vivo. In the second series of experiments the harvested cell population was highly purified (>95% macrophages) and none of the compounds fed to the rats caused a change of either eicosanoid or TNFα formation. Moreover the 6-keto-PGF1α:TxB2 and 6-keto-PGF1α:LTB4 ratios were not enhanced by any of the compounds tested. It is conceivable that in the first series the increased ratios 6-keto-PGF1α:TxB2 and 6-keto-PGF1α:LTB4 reflected the effect of carnitine or its congeners on PMN leucocytes rather than on macrophages.
Collapse
|
19
|
Abstract
The purpose of this study was to determine if free or esterified carnitine could alter fatty acid metabolism and ameliorate sepsis in lipopolysaccharide (LPS)-treated rats. Throughout a 96 h observation post-LPS, i.p. administration of both markedly reduced illness and accelerated recovery. Carnitine prevented the acute LPS-induced rise in serum triglycerides (45 ± 6, 59 ± 5 vs. 83 ± 8 mg/ml, p < 0.001), respectively. This difference was accompanied by a significant increase in liver lipogenesis in LPS controls compared to both carnitines and normal rats (6.1 ± 0.3 vs. 3.9 ± 0.5, 4.3 ± 0.5, and 1.8 ± 0.4 μmol/h, respectively, p < 0.04). Compared to normal rats, total liver carnitine was significantly elevated in LPS controls and even higher in the carnitine groups (357 ± 40 vs. 736 ± 38, 796 ± 79, and 1081 ± 21 nmol/g). The data suggest that carnitines may be of therapeutic value in sepsis treatment and one action may be to partition fatty acids from esterification to oxidation.
Collapse
|
20
|
Xia Y, Li Q, Zhong W, Dong J, Wang Z, Wang C. L-carnitine ameliorated fatty liver in high-calorie diet/STZ-induced type 2 diabetic mice by improving mitochondrial function. Diabetol Metab Syndr 2011; 3:31. [PMID: 22082204 PMCID: PMC3226540 DOI: 10.1186/1758-5996-3-31] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 11/15/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are an increasing number of patients suffering from fatty liver caused by type 2 diabetes. We intended to study the preventive and therapeutic effect of L-carnitine (LC) on nonalcoholic fatty liver disease (NAFLD) in streptozotocin (STZ)-induced type 2 diabetic mice and to explore its possible mechanism. METHODS Thirty male Kungming mice were randomly divided into five groups: control group, diabetic group, pre-treatment group (125 mg/kg BW), low-dose (125 mg/kg BW) therapeutic group and high-dose (250 mg/kg BW) therapeutic group. The morphology of hepatocytes was observed by light and electron microscopy. LC and ALC (acetyl L-carnitine) concentrations in the liver were determined by high-performance liquid chromatography (HPLC). Moreover, liver weight, insulin levels and free fatty acid (FFA) and triglyceride (TG) levels in the liver and plasma were measured. RESULTS Average liver LC and ALC levels were 33.7% and 20% lower, respectively, in diabetic mice compared to control mice (P < 0.05). After preventive and therapeutic treatment with LC, less hepatocyte steatosis, clearer crista and fewer glycogen granules in the mitochondria were observed. Decreased liver weight, TG levels, and FFA concentrations (P < 0.05) in the liver were also observed after treatment with LC in diabetic mice. Moreover, liver LC and ALC levels increased upon treatment with LC, whereas the ratio of LC and ALC decreased significantly (P < 0.01). CONCLUSION LC supplements ameliorated fatty liver in type 2 diabetic mice by increasing fatty acid oxidation and decreasing the LC/ALC ratio in the liver. Therefore, oral administration of LC protected mitochondrial function in liver.
Collapse
Affiliation(s)
- Yunqiu Xia
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Qing Li
- Class 5, Grade 2007, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Weizhen Zhong
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Jing Dong
- Physiology Department of the Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Zhulin Wang
- Class 5, Grade 2007, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Chunbo Wang
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| |
Collapse
|
21
|
Alexandre-Gouabau MC, Courant F, Le Gall G, Moyon T, Darmaun D, Parnet P, Coupé B, Antignac JP. Offspring Metabolomic Response to Maternal Protein Restriction in a Rat Model of Intrauterine Growth Restriction (IUGR). J Proteome Res 2011; 10:3292-302. [DOI: 10.1021/pr2003193] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Marie-Cécile Alexandre-Gouabau
- INRA and University of Nantes, UMR-1280 Physiologie des Adaptations Nutritionnelles CHU Hôtel Dieu, 44093 Nantes cedex 1, France
| | - Frédérique Courant
- ONIRIS, USC 2013, Laboratoire d’Etude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| | - Gwénaëlle Le Gall
- Institute of Food Research, Norwich Research Park, Colney, Norwich, NR4 7UA, United Kingdom
| | - Thomas Moyon
- INRA and University of Nantes, UMR-1280 Physiologie des Adaptations Nutritionnelles CHU Hôtel Dieu, 44093 Nantes cedex 1, France
| | - Dominique Darmaun
- INRA and University of Nantes, UMR-1280 Physiologie des Adaptations Nutritionnelles CHU Hôtel Dieu, 44093 Nantes cedex 1, France
| | - Patricia Parnet
- INRA and University of Nantes, UMR-1280 Physiologie des Adaptations Nutritionnelles CHU Hôtel Dieu, 44093 Nantes cedex 1, France
| | - Bérengère Coupé
- INRA and University of Nantes, UMR-1280 Physiologie des Adaptations Nutritionnelles CHU Hôtel Dieu, 44093 Nantes cedex 1, France
| | - Jean-Philippe Antignac
- ONIRIS, USC 2013, Laboratoire d’Etude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| |
Collapse
|
22
|
The role of peroxisome proliferator-activated receptor α in transcriptional regulation of novel organic cation transporters. Eur J Pharmacol 2010; 628:1-5. [DOI: 10.1016/j.ejphar.2009.11.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Revised: 11/08/2009] [Accepted: 11/17/2009] [Indexed: 12/26/2022]
|
23
|
Wen G, Ringseis R, Eder K. Mouse OCTN2 is directly regulated by peroxisome proliferator-activated receptor alpha (PPARalpha) via a PPRE located in the first intron. Biochem Pharmacol 2009; 79:768-76. [PMID: 19819229 DOI: 10.1016/j.bcp.2009.10.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 09/30/2009] [Accepted: 10/01/2009] [Indexed: 10/20/2022]
Abstract
Recent studies provided strong evidence to suggest that organic cation transporter 2 (OCTN2) is a direct target gene of peroxisome proliferator-activated receptor alpha (PPARalpha). However, subsequent studies failed to demonstrate a functional peroxisome proliferator response element (PPRE) in the promoter region of the OCTN2 gene. In the present study we hypothesized that the OCTN2 gene is transcriptionally induced by PPARalpha via a functional PPRE located in the first intron. In silico-analysis of the first intron of mouse OCTN2 revealed 11 putative PPRE with high similarity to the consensus PPRE. In addition, reporter gene assays using a mouse OCTN2 intron reporter construct containing a cluster of three partially overlapping PPRE (PPREint-1-8-10) revealed a marked response to exogenous mouse PPARalpha/RXRalpha and subsequent stimulation with PPARalpha agonist WY-14,643. Introduction of a selective mutation in either PPRE8 or PPRE10 in the PPREint-1-8-10 reporter constructs caused a substantial loss of the responsiveness to PPARalpha activation, but a selective mutation in PPRE1 resulted in a complete loss of responsiveness to PPARalpha activation. Moreover, gel shift assays revealed binding of PPARalpha/RXRalpha heterodimer to the PPRE1 of mouse OCTN2 first intron. In conclusion, the present study shows that mouse OCTN2 is a direct target gene of PPARalpha and that transcriptional upregulation of OCTN2 by PPARalpha is likely mediated via PPRE1 in its first intron.
Collapse
Affiliation(s)
- Gaiping Wen
- Institute of Agricultural and Nutritional Sciences, Martin-Luther-Universität Halle-Wittenberg, Von-Danckelmann-Platz 2, 06120 Halle (Saale), Germany
| | | | | |
Collapse
|
24
|
García-Delgado M, Peral MJ, Durán JM, García-Miranda P, Calonge ML, Ilundáin AA. Ontogeny of Na+/l-carnitine transporter and of γ-trimethylaminobutyraldehyde dehydrogenase and γ-butyrobetaine hydroxylase genes expression in rat kidney. Mech Ageing Dev 2009; 130:227-33. [DOI: 10.1016/j.mad.2008.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 11/20/2008] [Accepted: 12/13/2008] [Indexed: 11/26/2022]
|
25
|
Citil M, Karapehlivan M, Erdogan HM, Yucayurt R, Atakisi E, Atakisi O. Effect of orally administered l-carnitine on selected biochemical indicators of lactating Tuj-ewes. Small Rumin Res 2009. [DOI: 10.1016/j.smallrumres.2008.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Abstract
Ketone bodies accumulate in the plasma in conditions of fasting and uncontrolled diabetes. The initiating event is a change in the molar ratio of glucagon:insulin. Insulin deficiency triggers the lipolytic process in adipose tissue with the result that free fatty acids pass into the plasma for uptake by liver and other tissues. Glucagon appears to be the primary hormone involved in the induction of fatty acid oxidation and ketogenesis in the liver. It acts by acutely dropping hepatic malonyl-CoA concentrations as a consequence of inhibitory effects exerted in the glycolytic pathway and on acetyl-CoA carboxylase (EC 6.4.1.2). The fall in malonyl-CoA concentration activates carnitine acyltransferase I (EC 2.3.1.21) such that long-chain fatty acids can be transported through the inner mitochondrial membrane to the enzymes of fatty acid oxidation and ketogenesis. The latter are high-capacity systems assuring that fatty acids entering the mitochondria are rapidly oxidized to ketone bodies. Thus, the rate-controlling step for ketogenesis is carnitine acyltransferase I. Administration of food after a fast, or of insulin to the diabetic subject, reduces plasma free fatty acid concentrations, increases the liver concentration of malonyl-CoA, inhibits carnitine acyltransferase I and reverses the ketogenic process.
Collapse
|
27
|
Ringseis R, Luci S, Spielmann J, Kluge H, Fischer M, Geissler S, Wen G, Hirche F, Eder K. Clofibrate treatment up-regulates novel organic cation transporter (OCTN)-2 in tissues of pigs as a model of non-proliferating species. Eur J Pharmacol 2008; 583:11-7. [DOI: 10.1016/j.ejphar.2008.01.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 12/17/2007] [Accepted: 01/14/2008] [Indexed: 01/09/2023]
|
28
|
Luci S, Hirche F, Eder K. Fasting and Caloric Restriction Increases mRNA Concentrations of Novel Organic Cation Transporter-2 and Carnitine Concentrations in Rat Tissues. ANNALS OF NUTRITION AND METABOLISM 2008; 52:58-67. [DOI: 10.1159/000118872] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 11/06/2007] [Indexed: 11/19/2022]
|
29
|
van Vlies N, Ferdinandusse S, Turkenburg M, Wanders RJA, Vaz FM. PPAR alpha-activation results in enhanced carnitine biosynthesis and OCTN2-mediated hepatic carnitine accumulation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2007; 1767:1134-42. [PMID: 17692817 DOI: 10.1016/j.bbabio.2007.07.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 06/18/2007] [Accepted: 07/05/2007] [Indexed: 11/16/2022]
Abstract
In fasted rodents hepatic carnitine concentration increases considerably which is not observed in PPAR alpha-/- mice, indicating that PPAR alpha is involved in carnitine homeostasis. To investigate the mechanisms underlying the PPAR alpha-dependent hepatic carnitine accumulation we measured carnitine biosynthesis enzyme activities, levels of carnitine biosynthesis intermediates, acyl-carnitines and OCTN2 mRNA levels in tissues of untreated, fasted or Wy-14643-treated wild type and PPAR alpha-/- mice. Here we show that both enhancement of carnitine biosynthesis (due to increased gamma-butyrobetaine dioxygenase activity), extra-hepatic gamma-butyrobetaine synthesis and increased hepatic carnitine import (OCTN2 expression) contributes to the increased hepatic carnitine levels after fasting and that these processes are PPAR alpha-dependent.
Collapse
Affiliation(s)
- Naomi van Vlies
- Department of Clinical Chemistry, Academic Medical Center, University of Amsterdam, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Koch A, König B, Luci S, Stangl GI, Eder K. Dietary oxidised fat up regulates the expression of organic cation transporters in liver and small intestine and alters carnitine concentrations in liver, muscle and plasma of rats. Br J Nutr 2007; 98:882-9. [PMID: 17524183 DOI: 10.1017/s000711450775691x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It has been shown that treatment of rats with clofibrate, a synthetic agonist of PPARalpha, increases mRNA concentration of organic cation transporters (OCTN)-1 and -2 and concentration of carnitine in the liver. Since oxidised fats have been demonstrated in rats to activate hepatic PPARalpha, we tested the hypothesis that they also up regulate OCTN. Eighteen rats were orally administered either sunflower-seed oil (control group) or an oxidised fat prepared by heating sunflower-seed oil, for 6 d. Rats administered the oxidised fat had higher mRNA concentrations of typical PPARalpha target genes such as acyl-CoA oxidase, cytochrome P450 4A1 and carnitine palmitoyltransferases-1A and -2 in liver and small intestine than control rats (P < 0.05). Furthermore, rats treated with oxidised fat had higher hepatic mRNA concentrations of OCTN1 (1.5-fold) and OCTN2 (3.1-fold), a higher carnitine concentration in the liver and lower carnitine concentrations in plasma, gastrocnemius and heart muscle than control rats (P < 0.05). Moreover, rats administered oxidised fat had a higher mRNA concentration of OCTN2 in small intestine (2.4-fold; P < 0.05) than control rats. In conclusion, the present study shows that an oxidised fat causes an up regulation of OCTN in the liver and small intestine. An increased hepatic carnitine concentration in rats treated with the oxidised fat is probably at least in part due to an increased uptake of carnitine into the liver which in turn leads to reduced plasma and muscle carnitine concentrations. The present study supports the hypothesis that nutrients acting as PPARalpha agonists influence whole-body carnitine homeostasis.
Collapse
Affiliation(s)
- Alexander Koch
- Institute of Agricultural and Nutritional Sciences, Martin-Luther-University of Halle-Wittenberg, Emil-Abderhalden-Strasse 26, D-06108 Halle (Saale), Germany
| | | | | | | | | |
Collapse
|
31
|
López-Viñas E, Bentebibel A, Gurunathan C, Morillas M, de Arriaga D, Serra D, Asins G, Hegardt FG, Gómez-Puertas P. Definition by functional and structural analysis of two malonyl-CoA sites in carnitine palmitoyltransferase 1A. J Biol Chem 2007; 282:18212-18224. [PMID: 17452323 DOI: 10.1074/jbc.m700885200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Carnitine palmitoyltransferase 1 (CPT1) catalyzes the conversion of palmitoyl-CoA to palmitoylcarnitine in the presence of l-carnitine, thus facilitating the entry of fatty acids to mitochondria, in a process that is physiologically inhibited by malonyl-CoA. To examine the mechanism of CPT1 liver isoform (CPT1A) inhibition by malonyl-CoA, we constructed an in silico model of both its NH2- and COOH-terminal domains. Two malonyl-CoA binding sites were found. One of these, the "CoA site" or "A site," is involved in the interactions between NH2- and COOH-terminal domains and shares the acyl-CoA hemitunnel. The other, the "opposite-to-CoA site" or "O site," is on the opposite side of the enzyme, in the catalytic channel. The two sites share the carnitine-binding locus. To prevent the interaction between NH2- and COOH-terminal regions, we produced CPT1A E26K and K561E mutants. A double mutant E26K/K561E (swap), which was expected to conserve the interaction, was also produced. Inhibition assays showed a 12-fold decrease in the sensitivity (IC50) toward malonyl-CoA for CPT1A E26K and K561E single mutants, whereas swap mutant reverts to wild-type IC50 value. We conclude that structural interaction between both domains is critical for enzyme sensitivity to malonyl-CoA inhibition at the "A site." The location of the "O site" for malonyl-CoA binding was supported by inhibition assays of expressed R243T mutant. The model is also sustained by kinetic experiments that indicated linear mixed type malonyl-CoA inhibition for carnitine. Malonyl-CoA alters the affinity of carnitine, and there appears to be an exponential inverse relation between carnitine Km and malonyl-CoA IC50.
Collapse
Affiliation(s)
- Eduardo López-Viñas
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, E-28049 Madrid, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Assia Bentebibel
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Chandrashekaran Gurunathan
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Montserrat Morillas
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Dolores de Arriaga
- Departamento de Biología Molecular, Universidad de León, E-24071 León, Spain
| | - Dolors Serra
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Guillermina Asins
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Fausto G Hegardt
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Barcelona, E-08028 Barcelona, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain.
| | - Paulino Gómez-Puertas
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, E-28049 Madrid, Spain; CIBER Institute of Fisiopatología de la Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28049 Madrid, Spain
| |
Collapse
|
32
|
Luci S, Geissler S, König B, Koch A, Stangl GI, Hirche F, Eder K. PPARalpha agonists up-regulate organic cation transporters in rat liver cells. Biochem Biophys Res Commun 2006; 350:704-8. [PMID: 17011512 DOI: 10.1016/j.bbrc.2006.09.099] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 09/20/2006] [Indexed: 11/19/2022]
Abstract
It has been shown that clofibrate treatment increases the carnitine concentration in the liver of rats. However, the molecular mechanism is still unknown. In this study, we observed for the first time that treatment of rats with the peroxisome proliferator activated receptor (PPAR)-alpha agonist clofibrate increases hepatic mRNA concentrations of organic cation transporters (OCTNs)-1 and -2 which act as transporters of carnitine into the cell. In rat hepatoma (Fao) cells, treatment with WY-14,643 also increased the mRNA concentration of OCTN-2. mRNA concentrations of enzymes involved in carnitine biosynthesis were not altered by treatment with the PPARalpha agonists in livers of rats and in Fao cells. We conclude that PPARalpha agonists increase carnitine concentrations in livers of rats and cells by an increased uptake of carnitine into the cell but not by an increased carnitine biosynthesis.
Collapse
Affiliation(s)
- Sebastian Luci
- Institute of Agricultural and Nutritional Sciences, Martin-Luther-University Halle-Wittenberg, Emil-Abderhalden-Strasse 26, D-06108 Halle (Saale), Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Shin ES, Cho SY, Lee EH, Lee SJ, Chang IS, Lee TR. Positive regulation of hepatic carnitine palmitoyl transferase 1A (CPT1A) activities by soy isoflavones and L-carnitine. Eur J Nutr 2006; 45:159-64. [PMID: 16362726 DOI: 10.1007/s00394-005-0576-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2004] [Accepted: 07/05/2005] [Indexed: 02/02/2023]
Abstract
BACKGROUND Genistein increases CPT1A, a rate-limiting enzyme in the beta-oxidation pathway, enzyme activity by increasing CPT1A transcription in HepG2 cells and, consequently, suppresses high fat induced obesity in C57BL/6J mice. Genistein and daidzein are the most abundant isoflavones in soy. AIM OF STUDY To investigate the effect of co-treatment of genistein and L-carnitine on CPT1A enzyme activity and to determine whether daidzein also increases CPT1A activity and to establish a cell line that can be used to screen chemicals to regulate CPT1A transcription. METHODS The enzyme activities of CPT1A were determined after HepG2 cells were incubated with 10 microM genistein or 10 microM daidzein or 1 mM L-carnitine or in combination with 10 microM genistein and 1 mM L-carnitine or in combination with 10 microM daidzein and 1 mM L-carnitine. The mRNA expression levels of CPT1A were determined by real time PCR method after HepG2 cells were incubated with 10 microM genistein or 10 microM daidzein. A suggested CPT1A promoter region was cloned from human genomic DNA and the CPT1A promoter-luciferase reporter gene construct was made, and the promoter-reporter gene construct was transfected into human hepatoma cell line Huh7. RESULTS The enzyme activity of CPT1A was at least 2.3- fold higher in L-carnitine and genistein co-treated HepG2 cells than either single-agent treated cells. Daidzein also significantly increased the mRNA expression of CPT1A as well as the enzyme activity of CPT1A. A stable Huh7 cell line, which was selected after Huh7 cells were transfected with CPT1A promoter luciferase reporter gene construct, was characterized by confirming that luciferase activity of the cell line can be regulated by genistein and daidzein as well as clofibrate, a well-known CPT1A mRNA up-regulating drug. CONCLUSIONS Genistein and daidzein can up-regulate CPT1A enzyme activity through up-regulation of CPT1A transcription. Co-treatment of L-carnitine and genistein additively increases CPT1A enzyme activity in HepG2 cells. A stable Huh7 cell line transfected with the CPT1A promoter luciferase reporter gene was established and characterized.
Collapse
Affiliation(s)
- E S Shin
- BioResearch, R&D Center AmorePacific Corporation, 314-1, Bora-Ri, Giheung-Eup, Yongin-Si, Gyeonggi-Do, Korea
| | | | | | | | | | | |
Collapse
|
34
|
García-Miranda P, Durán JM, Peral MJ, Ilundáin AA. Developmental maturation and segmental distribution of rat small intestinal L-carnitine uptake. J Membr Biol 2006; 206:9-16. [PMID: 16440177 DOI: 10.1007/s00232-005-0769-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Indexed: 01/20/2023]
Abstract
Oral L-carnitine supplementation is commonly used in sports nutrition and in medicine; however, there is controversy regarding the mechanisms that mediate intestinal L-carnitine transport. We have previously reported that the Na(+)/L-carnitine transporter OCTN2 is present in the small intestinal apical membrane. Herein we aimed to find out if this step of intestinal L-carnitine absorption is ontogenically regulated, and if so, to determine the molecular mechanism(s) involved. L-[(3)H]-Carnitine uptake was measured in the jejunum and ileum of fetuses (E17 and E21), newborn (1 day-old), suckling (15 day-old), weaning (1 month-old) and adult (2 and 6 month-old) Wistar rats. Both, Na(+) -dependent and Na(+) -independent L-carnitine uptake rates, normalized to intestinal weight, significantly increased during the late gestation period, and then declined during the suckling period. After weaning, the rate of Na(+) -dependent L-carnitine uptake is no longer measurable. In E21- fetuses and newborn rats, L-carnitine uptake was higher in the ileum than in the jejunum. The decline in Na(+) -dependent L-carnitine uptake with maturation was mediated via a decrease in the V(max) of the uptake process with no change in its apparent K(m). Semi-quantitative RT-PCR assays showed that OCTN2 mRNA levels were significantly higher in E21-fetuses and newborn rats compared to suckling rats, which were in turn significantly higher than that in adult rats. Neither retardation of weaning nor L-carnitine supplementation prevented the down-regulation of Na(+)/L-carnitine transport activity. The results demonstrate for the first time that intestinal Na(+) -dependent L-carnitine uptake activity is under genetic regulation at the transcriptional level.
Collapse
Affiliation(s)
- P García-Miranda
- Depto. Fisiología y Zoología, Facultad de Farmacia, Universidad de Sevilla, Spain
| | | | | | | |
Collapse
|
35
|
Davis AT, Monroe TJ. Carnitine deficiency and supplementation do not affect the gene expression of carnitine biosynthetic enzymes in rats. J Nutr 2005; 135:761-4. [PMID: 15795431 DOI: 10.1093/jn/135.4.761] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Starved male weanling rats supplemented with 20 mmol/L pivalate in their drinking water exhibit significantly depressed concentrations of carnitine in tissues and plasma. In addition, pivalate supplementation has been linked with increased renal and hepatic trimethyllysine hydroxylase (TMLH) activity, whereas carnitine supplementation has been associated with significantly decreased hepatic gamma-butyrobetaine hydroxylase (BBH) activity. The purpose of this study was to determine whether pivalate or carnitine supplementation affects the activity and genetic expression of 2 enzymes of carnitine (Cn) biosynthesis, TMLH and BBH, expressed as mRNA abundance, relative to the abundance of beta-actin mRNA. Male weanling rats were administered the control treatment (C; n = 6), the pivalate treatment (P; n = 7), or the pivalate treatment plus supplemental dietary carnitine (P+Cn; n = 7). Rats in group P had elevated renal TMLH activity, relative to the other groups (P < 0.05). The groups did not differ in the abundance of renal or hepatic TMLH or BBH mRNA. A previously unreported finding was the quantifiable level of renal BBH mRNA, which was verified by direct sequencing of the BBH cDNA product amplified from kidney RNA. The groups did not differ in renal BBH mRNA abundance and renal BBH enzyme activity was not detected. Thus, the alterations in enzyme activities in the pivalate-treated rats are not regulated at the transcriptional level, and are apparently related to post-transcriptional effects on the enzymes themselves.
Collapse
Affiliation(s)
- Alan T Davis
- Department of Surgery, Michigan State University, Grand Rapids, MI, USA.
| | | |
Collapse
|
36
|
Abstract
Metabolism cycles daily between the fed and fasted states. The pathways of energy production are reversible and distinct. In the anabolic (fed) state, the liver stores glucose as glycogen, and fatty acid/triglyceride synthesis is active. In the catabolic (fasted) state, the liver becomes a glucose producer, lipogenesis is slowed, and fatty acid oxidation/ketogenesis is activated. The rate-limiting step for the latter is vested in the carnitine/carnitine palmitoyltransferase (CPT) system, and the off/on regulator of this is malonyl CoA. The AMP-induced protein kinase primarily determines the concentration of malonyl CoA. Four other systems have significant influence: two on fatty acid oxidation and two on lipogenesis. Peroxisome proliferator-activated receptor gamma-1 alpha, a master regulator of metabolism, induces hepatic gluconeogenesis and fatty acid oxidation in the catabolic phase. Deficiency of stearoyl CoA desaturase, although having no role in gluconeogenesis, powerfully induces fatty acid oxidation and weight loss despite increased food intake in rodents. Major stimulators of lipogenesis are carbohydrate-responsive element binding protein and the Insig system. The malonyl CoA-regulated CPT system has been firmly established in humans. The other systems have not yet been confirmed in humans, but likely are active there as well. Activation of fatty acid oxidation has considerable clinical promise for the treatment of obesity, type 2 diabetes, steatohepatitis, and lipotoxic damage to the heart.
Collapse
Affiliation(s)
- Daniel W Foster
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9030, USA.
| |
Collapse
|
37
|
Abstract
It has been shown previously that supplementation of sows with l-carnitine increases their reproduction performance. The current study was carried out to investigate if feeding of l-carnitine also affects the reproduction performance of rats. Thirty female rats at 4 weeks of age were divided into two groups. The rats were fed diets with or without l-carnitine (1 g/kg diet) over a period of 34 weeks. After 8 weeks of feeding, the female rats were mated the first time. Two more reproductive cycles followed, with 3-week intervals in between. Body weight development of the females was similar in both groups during the whole experimental period. Number of pregnancies and number of total rat pups, pups born alive and stillborn pups were not influenced by l-carnitine. There was also no influence of dietary l-carnitine on the body weight of individual pups and the litter weights at birth. Weight development of litters differed between both groups on several days, but no uniform effect of l-carnitine was observed. Body weight development of weaned rats fed a commercial diet was different between both groups, but only in one reproduction cycle. In conclusion, this study shows that l-carnitine supplementation does not improve the reproductive performance of rats.
Collapse
Affiliation(s)
- C Brandsch
- Institute of Nutritional Sciences, Martin-Luther University Halle-Wittenberg, Halle/Saale, Germany
| | | |
Collapse
|
38
|
Murata M, Sano Y, Ishihara K, Uchida M. Dietary fish oil and Undaria pinnatifida (wakame) synergistically decrease rat serum and liver triacylglycerol. J Nutr 2002; 132:742-7. [PMID: 11925471 DOI: 10.1093/jn/132.4.742] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Japanese eating habits are characterized by the consumption of various food materials such as cereals, vegetables, fish, shellfish, marine algae and meat. Therefore, properties of functional substances in food materials may be enhanced or lessened by the combination of various food materials. In the present study, we examined how the combination of wakame and fish containing polyunsaturated fatty acids, which are typical Japanese food materials, affected rat lipid metabolism. Rats were fed one of four diets [control diet (C), AIN-76 diet with 5 g/100 g rapeseed oil; wakame diet (W) containing 19.1 g/100 g Undaria pinnatifida (wakame) dried powder in the C diet; fish oil diet (FO), AIN-76 diet with 4.1 g/100 g fish oil; wakame-fish oil diet (W + FO), the FO diet containing 19.1 g/100 g dried wakame powder] for 4 wk. We measured the concentration of lipids in serum and liver and hepatic activities of enzymes involved in fatty acid metabolism. The W diet, FO diet and W + FO diet significantly reduced the concentration of triacylglycerols in the serum and liver compared with the C diet. This decrease in the concentration of hepatic triacylglycerol was greatest in rats fed the W + FO diet. The activity of glucose-6-phosphate dehydrogenase, which is involved in fatty acid synthesis in the liver, of rats fed the W, FO and W + FO diets was lower than that in rats fed the C diet. However, the activities of malic enzyme and fatty acid synthetase did not differ among the four groups. In contrast, the W diet and W + FO diet increased the serum concentration of beta-hydroxybutyrate. Further, the activity of 3-hydroxyacyl-CoA dehydrogenase, which is involved in fatty acid beta-oxidation in the liver, was greater in rats fed the W diet (42%), the FO diet (154%) and the W + FO diet (381%) than in those fed the C diet. Because the decrease in the concentration of triacylglycerol in the liver was greatest when rats were fed wakame and fish oil at the same time (W + FO diet), we conclude that there was a synergistic process affecting fatty acid beta-oxidation in the liver. These results suggest that the simultaneous consumption of fish (fish oil) and wakame decreases the concentration of triacylglycerol in the serum and liver.
Collapse
Affiliation(s)
- Masakazu Murata
- Laboratory of Applied Microorganisms, Marine Biochemistry Division, National Research Institute of Fisheries Science, Fisheries Research Agency Kanazawa-ku, Yokohama 236-8648, Japan.
| | | | | | | |
Collapse
|
39
|
Abstract
Carnitine is indispensable for energy metabolism, since it enables activated fatty acids to enter the mitochondria, where they are broken down via beta-oxidation. Carnitine is probably present in all animal species, and in numerous micro-organisms and plants. In mammals, carnitine homoeostasis is maintained by endogenous synthesis, absorption from dietary sources and efficient tubular reabsorption by the kidney. This review aims to cover the current knowledge of the enzymological, molecular, metabolic and regulatory aspects of mammalian carnitine biosynthesis, with an emphasis on the human and rat.
Collapse
|
40
|
Abstract
Carnitine is indispensable for energy metabolism, since it enables activated fatty acids to enter the mitochondria, where they are broken down via beta-oxidation. Carnitine is probably present in all animal species, and in numerous micro-organisms and plants. In mammals, carnitine homoeostasis is maintained by endogenous synthesis, absorption from dietary sources and efficient tubular reabsorption by the kidney. This review aims to cover the current knowledge of the enzymological, molecular, metabolic and regulatory aspects of mammalian carnitine biosynthesis, with an emphasis on the human and rat.
Collapse
Affiliation(s)
- Frédéric M Vaz
- Laboratory for Genetic Metabolic Diseases, Departments of Clinical Chemistry and Paediatrics, Emma Children's Hospital, Academic Medical Centre, University of Amsterdam, P.O. Box 22700, 1100 DE Amsterdam, The Netherlands.
| | | |
Collapse
|
41
|
Regulation of Ketogenesis in Liver. Compr Physiol 2001. [DOI: 10.1002/cphy.cp070221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
42
|
Abstract
Hepatothermic therapy (HT) of obesity is rooted in the observation that the liver has substantial capacities for both fatty acid oxidation and for thermogenesis. When hepatic fatty acid oxidation is optimized, the newly available free energy may be able to drive hepatic thermogenesis, such that respiratory quotient declines while basal metabolic rate increases, a circumstance evidently favorable for fat loss. Effective implementation of HT may require activation of carnitine palmitoyl transferase-1 (rate-limiting for fatty acid beta-oxidation), an increase in mitochondrial oxaloacetate production (required for optimal Krebs cycle activity), and up-regulation of hepatic thermogenic pathways. The possible utility of various natural agents and drugs for achieving these objectives is discussed. Potential components of HT regimens include EPA-rich fish oil, sesamin, hydroxycitrate, pantethine, L-carnitine, pyruvate, aspartate, chromium, coenzyme Q10, green tea polyphenols, conjugated linoleic acids, DHEA derivatives, cilostazol, diazoxide, and fibrate drugs. Aerobic exercise training and very-low-fat, low-glycemic-index, high-protein or vegan food choices may help to establish the hormonal environment conducive to effective HT. High-dose biotin and/or metformin may help to prevent an excessive increase in hepatic glucose output. Since many of the agents contemplated as components of HT regimens are nutritional or food-derived compounds likely to be health protective, HT is envisioned as an on-going lifestyle rather than as a temporary 'quick fix'. Initial clinical efforts to evaluate the potential of HT are now in progress.
Collapse
Affiliation(s)
- M F McCarty
- Pantox Laboratories, San Diego, California 92109, USA
| |
Collapse
|
43
|
Koumantakis E, Sifakis S, Koumantaki Y, Hassan E, Matalliotakis I, Papadopoulou E, Evageliou A. Plasma carnitine levels of pregnant adolescents in labor. J Pediatr Adolesc Gynecol 2001; 14:65-9. [PMID: 11479102 DOI: 10.1016/s1083-3188(00)00089-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
STUDY OBJECTIVE To determine the concentration of plasma carnitine (total, free, and acylcarnitine) during the delivery of uncomplicated pregnancies of adolescent women. To investigate the relationship between maternal and neonatal levels of carnitine and to compare these carnitine levels between pregnant and nonpregnant adolescents. DESIGN Samples of maternal and umbilical blood were taken at the time of delivery and examined for the determination of the carnitine-total, free, and acylcarnitine-concentration by the use of an enzymatic-radioisotope method. Twenty-two cases of uncomplicated adolescent pregnancies with a normal labor and without perinatal complications were examined. The plasma level of carnitine was also examined in 17 healthy nonpregnant adolescent women, which constituted the control group. RESULTS The concentrations of plasma carnitine in adolescent pregnancies at the time of delivery were calculated at 19.6 +/- 2.15 microMol/L (total), 12.62 +/- 1.31 microMol/L (free), and 6.98 +/- 1.55 microMol/L (acylcarnitine). The corresponding mean values in umbilical plasma were 30.31 +/- 2.06 microMol/L, 22.39 +/- 1.64 microMol/L, and 7.92 +/-.96 mucroMol/L. There is a statistically significant difference between the mean values in maternal and umbilical plasma (P <.0001 for total and free carnitine and P <.012 for acylcarnitine). The correlations between adolescent pregnant women and their infants as regards total, free, and acylcarnitine were 0.137, 0.018, and 0.33, respectively. Neither of these parameters was statistically significant. The corresponding mean values of carnitine in nonpregnant adolescent women were statistically significantly higher than in adolescent pregnant women (total carnitine: 41.61 +/- 3.09 microMol/L, free: 31.39 +/- 2.81 microMol/L, acylcarnitine: 10.22 +/- 1.88 microMol/L, P <.0001). CONCLUSIONS The concentration of plasma carnitine at the end of adolescent pregnancy is low compared to the levels of umbilical carnitine at birth and that found in nonpregnant adolescent women. It may not have an obvious impact on the utilization of fatty acids in an uncomplicated full-term pregnancy; however, it suggests the potential risk for neonatal fatty-acid oxidation in a preterm or complicated pregnancy.
Collapse
Affiliation(s)
- E Koumantakis
- Department of Obstetrics and Gynecology, Medical School, University of Crete, Heraklion, Greece
| | | | | | | | | | | | | |
Collapse
|
44
|
al-Amin AN, Ahrén B. Relation between malnutrition and development of diabetes mellitus. INTERNATIONAL JOURNAL OF PANCREATOLOGY : OFFICIAL JOURNAL OF THE INTERNATIONAL ASSOCIATION OF PANCREATOLOGY 1999; 26:125-30. [PMID: 10732288 DOI: 10.1385/ijgc:26:3:125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- A N al-Amin
- Department of Medicine, Lund University, Malmö, Sweden
| | | |
Collapse
|
45
|
Costa CC, de Almeida IT, Jakobs C, Poll-The BT, Duran M. Dynamic changes of plasma acylcarnitine levels induced by fasting and sunflower oil challenge test in children. Pediatr Res 1999; 46:440-4. [PMID: 10509365 DOI: 10.1203/00006450-199910000-00013] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The dynamic changes of plasma acylcarnitine levels in 1- to 7-y-old children during fasting and after the ingestion of sunflower oil were studied. Glucose, 3-hydroxybutyrate, acetoacetate, FFA, and individual plasma acylcarnitine levels were monitored in both conditions. Fasting experiments lasted for 20 h, and acylcarnitine concentrations were analyzed at 0, 15, and 20 h of fasting. During the fat load, acylcarnitine levels were analyzed at 0, 60, 120, and 180 min. In both tests, a generalized increase of all plasma straight-chain acylcarnitines was observed. Acetylcarnitine contributed the most to the increase of total esterified carnitine. In addition, we demonstrated that the relative increase of each individual acylcarnitine during enhanced fatty acid oxidation is tightly related to its molecular structure and chain length. Fasting as well as the fat load primarily resulted in an increase of unsaturated acylcarnitines. During fasting, C(12:1) and C(14:1) showed a relatively high increase, whereas after the fat load C(16:2) and C(14:2), metabolites of linoleic acid (66% of the fat load), were the main acylcarnitines that increased.
Collapse
Affiliation(s)
- C C Costa
- Centro de Metabolismos e Genética, Univ. Lisboa, Portugal
| | | | | | | | | |
Collapse
|
46
|
Jovanovic-Peterson L, Peterson CM. Review of Gestational Diabetes Mellitus and Low-calorie Diet and Physical Exercise as Therapy. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1099-0895(199612)12:4<287::aid-dmr171>3.0.co;2-e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
47
|
McCarty MF, Gustin JC. Pyruvate and hydroxycitrate/carnitine may synergize to promote reverse electron transport in hepatocyte mitochondria, effectively 'uncoupling' the oxidation of fatty acids. Med Hypotheses 1999; 52:407-16. [PMID: 10416948 DOI: 10.1054/mehy.1997.0683] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In a recent pilot study, joint administration of pyruvate, hydroxycitrate (HCA), and carnitine to obese subjects was associated with a remarkable rate of body-fat loss and thermogenesis, strongly suggestive of uncoupled fatty-acid oxidation. Hepatocytes possess an uncoupling mechanism--reverse electron transport--that enables fasting ketogenesis to proceed independent of respiratory control. Electrons entering the respiratory chain at the coenzyme Q (CoQ) level via FAD-dependent acyl coA dehydrogenase, can be driven 'up' the chain by the electrochemical proton gradient to reduce NAD+; if these electrons are then shuttled to the cytoplasm, returning to the respiratory chain at the CoQ level, the net result is heat generation at the expense of the proton gradient, enabling the uncoupled flow of electrons to oxygen. Pyruvate's bariatric utility may stem from its ability to catalyze the rapid transport of high-energy electrons from mitochondria to the cytoplasm, thus stimulating electron shuttle mechanisms. By enabling rapid mitochondrial uptake of fatty acids and thus disinhibiting hepatocyte ketogenesis, HCA/carnitine should initiate reverse electron transport: concurrent amplification of electron shuttle mechanisms by pyruvate can be expected to accelerate this reverse electron transport, thereby decreasing the electrochemical proton gradient. As a result, hepatocytes may be able to convert fatty acids to CO2 and heat with little net generation of ATP. These considerations suggest that it may be feasible to render hepatocytes functionally equivalent to activated brown fat, such that stored fat can be selectively oxidized in the absence of caloric restriction. Other measures which enhance the efficiency of hepatocyte electron shuttle mechanisms may increase the efficacy of this strategy.
Collapse
Affiliation(s)
- M F McCarty
- NutriGuard Research, Encinitas, CA 92024, USA
| | | |
Collapse
|
48
|
Mingrone G, Greco AV, Capristo E, Benedetti G, Giancaterini A, De Gaetano A, Gasbarrini G. L-carnitine improves glucose disposal in type 2 diabetic patients. J Am Coll Nutr 1999; 18:77-82. [PMID: 10067662 DOI: 10.1080/07315724.1999.10718830] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Aim of the present study is to evaluate the effects of L-carnitine on insulin-mediated glucose uptake and oxidation in type II diabetic patients and compare the results with those in healthy controls. DESIGN Fifteen type II diabetic patients and 20 healthy volunteers underwent a short-term (2 hours) euglycemic hyperinsulinemic clamp with simultaneous constant infusion of L-carnitine (0.28 micromole/kg bw/minute) or saline solution. Respiratory gas exchange was measured by an open-circuit ventilated hood system. Plasma glucose, insulin, non-esterified fatty acids (NEFA) and lactate levels were analyzed. Nitrogen urinary excretion was calculated to evaluate protein oxidation. RESULTS Whole body glucose uptake was significantly (p<0.001) higher with L-carnitine than with saline solution in the two groups investigated (48.66+/-4.73 without carnitine and 52.75+/-5.19 micromoles/kg(ffm)/minute with carnitine in healthy controls, and 35.90+/-5.00 vs. 38.90+/-5.16 micromoles/kg(ffm)/minute in diabetic patients). Glucose oxidation significantly increased only in the diabetic group (17.61+/-3.33 vs. 16.45+/-2.95 micromoles/kg(ffm)/minute, p<0.001). On the contrary, glucose storage increased in both groups (controls: 26.36+/-3.25 vs. 22.79+/-3.46 micromoles/kg(ffm)/minute, p<0.001; diabetics: 21.28+/-3.18 vs. 19.66+/-3.04 micromoles/kg(ffm)/minute, p<0.001). In type II diabetic patients, plasma lactate significantly decreased during L-carnitine infusion compared to saline, going from the basal period to the end-clamp period (0.028+/-0.0191 without carnitine and 0.0759+/-0.0329 with carnitine, p<0.0003). CONCLUSIONS L-carnitine constant infusion improves insulin sensitivity in insulin resistant diabetic patients; a significant effect on whole body insulin-mediated glucose uptake is also observed in normal subjects. In diabetics, glucose, taken up by the tissues, appears to be promptly utilized as fuel since glucose oxidation is increased during L-carnitine administration. The significantly reduced plasma levels of lactate suggest that this effect might be exerted through the activation of pyruvate dehydrogenase, whose activity is depressed in the insulin resistant status.
Collapse
Affiliation(s)
- G Mingrone
- Istituto di Medicina Interna, Catholic University, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Gustafson B, Ransnäs LA. Gsalpha-mediated regulation of the carnitine carrier in S49 lymphoma cells. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1372:255-60. [PMID: 9675303 DOI: 10.1016/s0005-2736(98)00063-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Carnitine is essential for mitochondrial oxidation of long-chain fatty acids. Peripheral cells rely on plasma transport of carnitine which is taken up by an active mechanism in the plasma membrane. This project investigated the plasma membrane bound carnitine carrier in cultured S49 lymphoma cells. We investigated wild-type cells and two mutant cells lines showing deficient activity of adenylyl cyclase, cyc- lacking and H21a containing a deficient Gsalpha. Plasma membranes derived from cyc- cells displayed six times more carnitine binding sites and a 1.35 times faster uptake rate than plasma membranes from wild-type cells. In vitro mixing of plasma membranes from cyc- and wild-type cells transferred a factor reducing the number of expected carnitine binding sites by about 30%. Cyclic AMP could not substitute for wild-type membranes as the inhibitor of carnitine binding to plasma membranes derived from cyc- cells. Cholera toxin induced ADP-ribosylation of Gsalpha causing activation of Gsalpha present in wild-type but not in cyc- cells, further reducing carnitine uptake and carnitine binding to plasma membranes. Our findings thus supported the notion that Gsalpha by a mechanism not involving cyclic AMP inhibited cellular uptake of carnitine by reducing the number of available carnitine binding sites in plasma membranes.
Collapse
Affiliation(s)
- B Gustafson
- Department of Allergology, Sahlgren's University Hospital, S-413 45 Göteborg, Sweden
| | | |
Collapse
|
50
|
al-Shurbaji A, Berglund L, Berge RK, Cederblad G, Humble E. On the interrelationship between hepatic carnitine, fatty acid oxidation, and triglyceride biosynthesis in nephrosis. Lipids 1997; 32:847-52. [PMID: 9270976 DOI: 10.1007/s11745-997-0108-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The nephrotic syndrome is associated with disturbances in plasma lipid pattern and metabolism. However, the reason for these perturbations is poorly understood. In the present study, we have investigated hepatic triglyceride metabolism in puromycin aminonucleoside-induced nephrotic syndrome in rats. Nephrotic rats displayed a 70% increase in hepatic triglyceride levels compared to controls (16.9 +/- 1.6 vs. 9.8 +/- 0.6 mumol/g liver; means +/- SEM, P < 0.01). The capacity for hepatic mitochondrial beta-oxidation of fatty acids was substantially elevated (80%). This was associated with a rise in the liver content of the fatty acid carrier carnitine (1.24 +/- 0.06 vs. 0.85 +/- 0.07 mumol/g dry weight, P < 0.05). A positive correlation between the levels of acetylcarnitine and acetyl-CoA was found in normal as well as in nephrotic rats, implying that carnitine plays an important role as an acetyl group acceptor in the liver under normo- and hyperlipidemic conditions. Changes in carnitine levels seem to be tightly coupled to the rate of fatty acid oxidation. There was a significant elevation in the activity of phosphatidate phosphohydrolase (E.C. 3.1.3.4) in liver microsomes from nephrotic rats (1.07 +/- 0.09 vs. 0.81 +/- 0.04 nmol/min.mg protein, P < 0.02). Hepatic very low density lipoprotein (VLDL)-triglyceride secretion rate was 18% higher in nephrotic rats than in controls. The results demonstrate a deranged hepatic triglyceride metabolism in nephrosis, with an increased hepatic triglyceride biosynthesis, a sizable accumulation of hepatic triglycerides, and only a modest increase in VLDL triglyceride secretion. In addition, mitochondrial beta-oxidation of fatty acids was enhanced, associated with an increased availability of carnitine.
Collapse
Affiliation(s)
- A al-Shurbaji
- Department of Medical Laboratory Sciences and Technology, Karolinska Institutet, Huddinge University Hospital, Sweden
| | | | | | | | | |
Collapse
|