1
|
Pike B, Zhao J, Hicks JA, Wang F, Hagen R, Liu HC, Odle J, Lin X. Intestinal Carnitine Status and Fatty Acid Oxidation in Response to Clofibrate and Medium-Chain Triglyceride Supplementation in Newborn Pigs. Int J Mol Sci 2023; 24:ijms24076066. [PMID: 37047049 PMCID: PMC10094207 DOI: 10.3390/ijms24076066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
To investigate the role of peroxisome proliferator-activated receptor alpha (PPARα) in carnitine status and intestinal fatty acid oxidation in neonates, a total of 72 suckled newborn piglets were assigned into 8 dietary treatments following a 2 (±0.35% clofibrate) × 4 (diets with: succinate+glycerol (Succ), tri-valerate (TC5), tri-hexanoate (TC6), or tri-2-methylpentanoate (TMPA)) factorial design. All pigs received experimental milk diets with isocaloric energy for 5 days. Carnitine statuses were evaluated, and fatty acid oxidation was measured in vitro using [1-14C]-palmitic acid (1 mM) as a substrate in absence or presence of L659699 (1.6 µM), iodoacetamide (50 µM), and carnitine (1 mM). Clofibrate increased concentrations of free (41%) and/or acyl-carnitine (44% and 15%) in liver and plasma but had no effects in the intestine. The effects on carnitine status were associated with the expression of genes involved in carnitine biosynthesis, absorption, and transportation. TC5 and TMPA stimulated the increased fatty acid oxidation rate induced by clofibrate, while TC6 had no effect on the increased fatty acid oxidation induced by clofibrate (p > 0.05). These results suggest that dietary clofibrate improved carnitine status and increased fatty acid oxidation. Propionyl-CoA, generated from TC5 and TMPA, could stimulate the increased fatty acid oxidation rate induced by clofibrate as anaplerotic carbon sources.
Collapse
Affiliation(s)
- Brandon Pike
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Jinan Zhao
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Julie A Hicks
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Feng Wang
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Rachel Hagen
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Hsiao-Ching Liu
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Jack Odle
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Xi Lin
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
2
|
Zhou S, Shu Y. Transcriptional Regulation of Solute Carrier (SLC) Drug Transporters. Drug Metab Dispos 2022; 50:DMD-MR-2021-000704. [PMID: 35644529 PMCID: PMC9488976 DOI: 10.1124/dmd.121.000704] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 05/02/2022] [Accepted: 05/16/2022] [Indexed: 09/03/2023] Open
Abstract
Facilitated transport is necessitated for large size, charged, and/or hydrophilic drugs to move across the membrane. The drug transporters in the solute carrier (SLC) superfamily, mainly including organic anion-transporting polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), organic cation/carnitine transporters (OCTNs), peptide transporters (PEPTs), and multidrug and toxin extrusion proteins (MATEs), are critical facilitators of drug transport and distribution in human body. The expression of these SLC drug transporters is found in tissues throughout the body, with high abundance in the epithelial cells of major organs for drug disposition, such as intestine, liver, and kidney. These SLC drug transporters are clinically important in drug absorption, metabolism, distribution, and excretion. The mechanisms underlying their regulation have been revealing in recent years. Epigenetic and nuclear receptor-mediated transcriptional regulation of SLC drug transporters have particularly attracted much attention. This review focuses on the transcriptional regulation of major SLC drug transporter genes. Revealing the mechanisms underlying the transcription of those critical drug transporters will help us understand pharmacokinetics and pharmacodynamics, ultimately improving drug therapeutic effectiveness while minimizing drug toxicity. Significance Statement It has become increasingly recognized that solute carrier (SLC) drug transporters play a crucial, and sometimes determinative, role in drug disposition and response, which is reflected in decision-making during not only clinical drug therapy but also drug development. Understanding the mechanisms accounting for the transcription of these transporters is critical to interpret their abundance in various tissues under different conditions, which is necessary to clarify the pharmacological response, adverse effects, and drug-drug interactions for clinically used drugs.
Collapse
Affiliation(s)
- Shiwei Zhou
- Pharmaceutical Sciences, University of Maryland, United States
| | - Yan Shu
- Pharmaceutical Sciences, University of Maryland, United States
| |
Collapse
|
3
|
Upadhyay A, Boyle KE, Broderick TL. The Effects of Streptozotocin-Induced Diabetes and Insulin Treatment on Carnitine Biosynthesis and Renal Excretion. Molecules 2021; 26:6872. [PMID: 34833964 PMCID: PMC8620001 DOI: 10.3390/molecules26226872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Carnitine insufficiency is reported in type 1 diabetes mellitus. To determine whether this is accompanied by defects in biosynthesis and/or renal uptake, liver and kidney were obtained from male Sprague-Dawley rats with streptozotocin-induced diabetes. Diabetic rats exhibited the metabolic consequences of type 1 diabetes, including hypoinsulinemia, hyperglycemia, and increased urine output. Systemic hypocarnitinemia, expressed as free carnitine levels, was evident in the plasma, liver, and kidney of diabetic rats. Compared to control rats, the low free carnitine in the plasma of diabetic rats was accompanied by decreased expression of γ-butyrobetaine hydroxylase in liver and kidney, suggesting impaired carnitine biosynthesis. Expression of organic cation transporter-2 in kidney was also reduced, indicating impaired renal reabsorption, and confirmed by the presence of elevated levels of free carnitine in the urine of diabetic rats. Insulin treatment of diabetic rats reversed the plasma hypocarnitinemia, increased the free carnitine content in both kidney and liver, and prevented urinary losses of free carnitine. This was associated with increased expression of γ-butyrobetaine hydroxylase and organic cation transporter-2. The results of our study indicate that type 1 diabetes induced with streptozotocin disrupts carnitine biosynthesis and renal uptake mechanisms, leading to carnitine insufficiency. These aberrations in carnitine homeostasis are prevented with daily insulin treatment.
Collapse
Affiliation(s)
- Aman Upadhyay
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - Kate E. Boyle
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
| | - Tom L. Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
4
|
Acevedo-Giraldo J, Sánchez J, Romero M. Effects of feed withdrawal times prior to slaughter on some animal welfare indicators and meat quality traits in commercial pigs. Meat Sci 2020; 167:107993. [PMID: 32388087 DOI: 10.1016/j.meatsci.2019.107993] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/10/2019] [Accepted: 10/28/2019] [Indexed: 11/26/2022]
|
5
|
Lin X, Lyvers Peffer PA, Woodworth J, Odle J. Ontogeny of carnitine biosynthesis in Sus scrofa domesticus, inferred from γ-butyrobetaine hydroxylase (dioxygenase) activity and substrate inhibition. Am J Physiol Regul Integr Comp Physiol 2020; 319:R43-R49. [PMID: 32432915 DOI: 10.1152/ajpregu.00051.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
γ-Butyrobetaine hydroxylase (γ-BBH) is the last limiting enzyme of the l-carnitine biosynthesis pathway and plays an important role in catalyzing the hydroxylation of γ-butyrobetaine (γ-BB) to l-carnitine. To study the developmental effect of substrate concentration on the enzyme's specific activity, kinetics of γ-BBH were measured in liver and kidney from newborn and 1-, 7-, 21-, 35-, 56-, and 210-day-old domestic pigs. Fresh tissue homogenates were assayed under nine concentrations of γ-BB from 0 to 1.5 mM. Substrate inhibition associated with age was observed at ≥0.6 mM of γ-BB. Hepatic activity was low at birth but increased after 1 day. By 21 days, the activity rose by 6.6-fold (P < 0.05) and remained constant after 56 days. Renal activity was higher than in liver at birth but remained constant through 35 days. By 56 days, the velocity increased by 44% over the activity at birth (P < 0.05). The apparent Km for γ-BB at birth on average was 2.8-fold higher than at 1 day. The Km value was 60% higher in kidney than liver during development but showed no difference in adult pigs. The total organ enzyme activity increased by 130-fold for liver and 18-fold for kidney as organ weight increased from birth to 56 days. In conclusion, age and substrate affect γ-BBH specific activity and Km for γ-BB in liver and kidney. Whereas the predominant organ for carnitine synthesis is likely the kidney at birth, the liver appears to predominate after the pig exceeds 7 days of age.
Collapse
Affiliation(s)
- Xi Lin
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, North Carolina
| | - Pasha A Lyvers Peffer
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, North Carolina
| | | | - Jack Odle
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
6
|
Mooren FC, Krueger K, Ringseis R, Eder K, Liebisch G, Conrad K, Alack K, Hajizadeh Maleki B. Combined effects of moderate exercise and short-term fasting on markers of immune function in healthy human subjects. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1103-R1115. [PMID: 32401626 DOI: 10.1152/ajpregu.00341.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This study aimed to investigate the effects of a short-term (36 h) fasting period combined with an acute bout of exercise on markers of immune function and inflammation in healthy human subjects. Fourteen moderately trained male subjects (aged 19-39 yr) participated in a 36-h fasting trial (FA-T), followed by an acute bout of moderate exercise (60% V̇o2max). After 1 wk, the same subjects, as their own control, participated in a nonfasting trial (NFA-T) in which they performed an exercise trial of the same duration and intensity. Blood samples were taken before, immediately after, and 1 h after each exercise bout and analyzed for several immunological and metabolic markers. At baseline, fasting subjects showed lower levels of T cell apoptosis, lymphocyte-proliferative responses, IL-6, monocyte chemoattractant protein-1 (MCP-1), insulin, and leptin (P < 0.05) as well as higher levels of neutrophil oxidative burst and thiobarbituric acid reactive substances (TBARS) than those in the NFA-T (P < 0.05). After the exercise protocol, fasted subjects revealed higher T cell apoptosis, neutrophil oxidative burst, TBARS, TNFα, and MCP-1 levels as well as lower levels of lymphocyte-proliferative response, IL-6, insulin, and leptin than those in the NFA-T (P < 0.05). Short-term fasting aggravates perturbations in markers of immune function, and inflammation was induced by an acute moderate-intensity exercise protocol.
Collapse
Affiliation(s)
- Frank C Mooren
- Witten/Herdecke University, Faculty of Health/School of Medicine, Witten, Germany
| | - Karsten Krueger
- Department of Exercise Physiology and Sports Therapy, Justus-Liebig-University, Giessen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University, Giessen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University, Giessen, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Regensburg, Germany
| | - Kerstin Conrad
- Department of Exercise Physiology and Sports Therapy, Justus-Liebig-University, Giessen, Germany
| | - Katharina Alack
- Department of Exercise Physiology and Sports Therapy, Justus-Liebig-University, Giessen, Germany
| | - Behzad Hajizadeh Maleki
- Department of Exercise Physiology and Sports Therapy, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
7
|
Effects of Exercise Training on Renal Carnitine Biosynthesis and Uptake in the High-Fat and High-Sugar-Fed Mouse. Molecules 2020; 25:molecules25092100. [PMID: 32365864 PMCID: PMC7248909 DOI: 10.3390/molecules25092100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Diet-induced obesity inhibits hepatic carnitine biosynthesis. Herein, the effects of high-fat (HF) and high-sugar (HFHS) feeding and exercise training (ET) on renal carnitine biosynthesis and uptake were determined. (2) Methods: Male C57BL/6J mice were assigned to the following groups: lean control (standard chow), HFHS diet, and HFHS diet with ET. ET consisted of 150 min of treadmill running per week for 12 weeks. Protein levels of γ-butyrobetaine hydroxylase (γ-BBH) and organic cation transporter-2 (OCTN2) were measured as markers of biosynthesis and uptake, respectively. (3) Results: HFHS feeding induced an obese diabetic state with accompanying hypocarnitinemia, reflected by decreased free carnitine levels in plasma and kidney. This hypocarnitinemia was associated with decreased γ-BBH (~30%) and increased OCTN2 levels (~50%). ET failed to improve the obesity and hyperglycemia, but improved insulin levels and prevented the hypocarnitinemia. ET increased protein levels of γ-BBH, whereas levels of OCTN2 were decreased. Peroxisome proliferator-activated receptor-alpha content was not changed by the HFHS diet or ET. (4) Conclusions: Our results indicate that ET prevents the hypocarnitinemia induced by HFHS feeding by increasing carnitine biosynthesis in kidney. Increased expression of OCTN2 with HFHS feeding suggests that renal uptake was stimulated to prevent carnitine loss.
Collapse
|
8
|
Ribot J, Arreguín A, Kuda O, Kopecky J, Palou A, Bonet ML. Novel Markers of the Metabolic Impact of Exogenous Retinoic Acid with A Focus on Acylcarnitines and Amino Acids. Int J Mol Sci 2019; 20:E3640. [PMID: 31349613 PMCID: PMC6696161 DOI: 10.3390/ijms20153640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/21/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
Treatment with all-trans retinoic acid (ATRA), the carboxylic form of vitamin A, lowers body weight in rodents by promoting oxidative metabolism in multiple tissues including white and brown adipose tissues. We aimed to identify novel markers of the metabolic impact of ATRA through targeted blood metabolomics analyses, with a focus on acylcarnitines and amino acids. Blood was obtained from mice treated with a high ATRA dose (50 mg/kg body weight/day, subcutaneous injection) or placebo (controls) during the 4 days preceding collection. LC-MS/MS analyses with a focus on acylcarnitines and amino acids were conducted on plasma and PBMC. Main results showed that, relative to controls, ATRA-treated mice had in plasma: increased levels of carnitine, acetylcarnitine, and longer acylcarnitine species; decreased levels of citrulline, and increased global arginine bioavailability ratio for nitric oxide synthesis; increased levels of creatine, taurine and docosahexaenoic acid; and a decreased n-6/n-3 polyunsaturated fatty acids ratio. While some of these features likely reflect the stimulation of lipid mobilization and oxidation promoted by ATRA treatment systemically, other may also play a causal role underlying ATRA actions. The results connect ATRA to specific nutrition-modulated biochemical pathways, and suggest novel mechanisms of action of vitamin A-derived retinoic acid on metabolic health.
Collapse
Affiliation(s)
- Joan Ribot
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain.
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain.
| | - Andrea Arreguín
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
| | - Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Andreu Palou
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Maria Luisa Bonet
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| |
Collapse
|
9
|
Shi B, Zhao X, Wang C, Wang N, Tian M, Shan A. l-carnitine and fat type in the maternal diet during gestation and lactation modify the fatty acid composition and expression of lipid metabolism-related genes in piglets. J Anim Physiol Anim Nutr (Berl) 2019; 103:1207-1217. [PMID: 30994244 DOI: 10.1111/jpn.13099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 12/01/2022]
Abstract
This study examined the influence of adding different amounts of maternal dietary l-carnitine and two fat types on fatty acid (FA) composition and the expression of lipid metabolism-related genes in piglets. The experiment was designed as a 2 × 2 factorial with two fat types (3.5% soyabean oil, SO, and 3.5% fish oil, FO) and two levels of l-carnitine (0 and 100 mg/kg) added to the sows' diets. A higher proportion of n-3 polyunsaturated fatty acids (PUFA) and a lower ratio of n-6/n-3 PUFA in sow milk and piglet tissues were observed in the FO groups than in the SO groups. Adding l-carnitine increased the proportion of C16:1 in sow milk and decreased n-3 PUFA in piglet subcutaneous fat. Hepatic peroxisome proliferator-activated receptor α (PPAR-α) was more abundantly expressed in piglets from the FO groups than from the SO groups (p < 0.05), whereas stearoyl-CoA-desaturase (SCD), sterol regulatory element binding protein-1 (SREBP1) and ∆6-desaturase (D6D) genes were less expressed in the FO groups compared with piglets from the SO groups. The expression of fatty acid synthase (FAS) genes was decreased in the SO groups with l-carnitine compared to that of the other dietary treatments. No differences among dietary treatments were observed with regard to the expression of acetyl-CoA carboxylase (ACC). In conclusion, FO and l-carnitine supplementation in sows affect FA composition and hepatic gene expression in piglets.
Collapse
Affiliation(s)
- Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Xuan Zhao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chuanqi Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Na Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Meili Tian
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
10
|
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: 3.8] [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
|
11
|
Mendes C, Meirelles GC, Silva MA, Ponchel G. Intestinal permeability determinants of norfloxacin in Ussing chamber model. Eur J Pharm Sci 2018; 121:236-242. [DOI: 10.1016/j.ejps.2018.05.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 12/18/2022]
|
12
|
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: 1.8] [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
|
13
|
Zeitz JO, Weber A, Most E, Windisch W, Bolduan C, Geyer J, Romberg FJ, Koch C, Eder K. Effects of supplementing rumen-protected niacin on fiber composition and metabolism of skeletal muscle in dairy cows during early lactation. J Dairy Sci 2018; 101:8004-8020. [PMID: 29960772 DOI: 10.3168/jds.2018-14490] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/13/2018] [Indexed: 01/01/2023]
Abstract
Nicotinic acid (NA) has been shown to induce muscle fiber switching toward oxidative type I fibers and a muscle metabolic phenotype that favors fatty acid (FA) utilization in growing rats, pigs, and lambs. The hypothesis of the present study was that supplementation of NA in cows during the periparturient phase also induces muscle fiber switching from type II to type I fibers in skeletal muscle and increases the capacity of the muscle to use free FA, which may help to reduce nonesterified fatty acid (NEFA) flow to the liver, liver triglyceride (TG) accumulation, and ketogenesis. Thirty multiparous Holstein dairy cows were allocated to 2 groups and fed a total mixed ration without (control group) or with ∼55 g of rumen-protected NA per cow per day (NA group) from 21 d before expected calving until 3 wk postpartum (p.p.). Blood samples were collected on d -21, -14, -7, 7, 14, 21, 35, and 63 relative to parturition for analysis of TG, NEFA, and β-hydroxybutyrate. Muscle and liver biopsies were collected on d 7 and 21 for gene expression analysis and to determine muscle fiber composition in the musculus semitendinosus, semimembranosus, and longissimus lumborum by immunohistochemistry, and liver TG concentrations. Supplementation of NA did not affect the proportions of type I (oxidative) or the type II:type I ratio in the 3 muscles considered. A slight shift from glycolytic IIx fibers toward oxidative-glycolytic fast-twitch IIa fibers was found in the semitendinosus, and a tendency in the longissimus lumborum, but not in the semimembranosus. The transcript levels of the genes encoding the muscle fiber type isoforms and involved in FA uptake and oxidation, carnitine transport, tricarboxylic acid cycle, oxidative phosphorylation, and glucose utilization were largely unaffected by NA supplementation in all 3 muscles. Supplementation of NA had no effect on plasma TG and NEFA concentrations, liver TG concentrations, and hepatic expression of genes involved in hepatic FA utilization and lipogenesis. However, it reduced plasma β-hydroxybutyrate concentrations in wk 2 and 3 p.p. by 18 and 26% and reduced hepatic gene expression of fibroblast growth factor 21, a stress hormone involved in the regulation of ketogenesis, by 74 and 56%. In conclusion, a high dosage of rumen-protected NA reduced plasma β-hydroxybutyrate concentrations in cows during early lactation, but failed to cause an alteration in muscle fiber composition and muscle metabolic phenotype.
Collapse
Affiliation(s)
- J O Zeitz
- Institute of Animal Nutrition and Nutritional Physiology, University of Giessen, Heinrich-Buff-Ring 26-32 (IFZ), 35392 Giessen, Germany.
| | - A Weber
- Institute of Animal Nutrition and Nutritional Physiology, University of Giessen, Heinrich-Buff-Ring 26-32 (IFZ), 35392 Giessen, Germany
| | - E Most
- Institute of Animal Nutrition and Nutritional Physiology, University of Giessen, Heinrich-Buff-Ring 26-32 (IFZ), 35392 Giessen, Germany
| | - W Windisch
- Chair of Animal Nutrition, Technische Universität München, Liesel-Beckmann-Strasse 2, 85354 Freising, Germany
| | - C Bolduan
- Chair of Animal Nutrition, Technische Universität München, Liesel-Beckmann-Strasse 2, 85354 Freising, Germany
| | - J Geyer
- Institute of Pharmacology and Toxicology, University of Giessen, Schubertstraße 81 (BFS), 35392 Giessen, Germany
| | - F-J Romberg
- Educational and Research Centre for Animal Husbandry, Hofgut Neumuehle, 67728 Muenchweiler an der Alsenz, Germany
| | - C Koch
- Educational and Research Centre for Animal Husbandry, Hofgut Neumuehle, 67728 Muenchweiler an der Alsenz, Germany
| | - K Eder
- Institute of Animal Nutrition and Nutritional Physiology, University of Giessen, Heinrich-Buff-Ring 26-32 (IFZ), 35392 Giessen, Germany
| |
Collapse
|
14
|
Broderick TL, Cusimano FA, Carlson C, Babu JR. Biosynthesis of the Essential Fatty Acid Oxidation Cofactor Carnitine Is Stimulated in Heart and Liver after a Single Bout of Exercise in Mice. J Nutr Metab 2018; 2018:2785090. [PMID: 30002928 PMCID: PMC5996426 DOI: 10.1155/2018/2785090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/13/2018] [Accepted: 04/04/2018] [Indexed: 11/19/2022] Open
Abstract
We determined whether one single bout of exercise stimulates carnitine biosynthesis and carnitine uptake in liver and heart. Free carnitine (FC) in plasma was assayed using acetyltransferase and [14C]acetyl-CoA in Swiss Webster mice after 1 hour of moderate-intensity treadmill running or 4 hours and 8 hours into recovery. Liver and heart were removed under the same conditions for measurement of carnitine biosynthesis enzymes (liver butyrobetaine hydroxylase, γ-BBH; heart trimethyllysine dioxygenase, TMLD), organic cation transporter-2 (OCTN2, carnitine transporter), and liver peroxisome proliferator-activated receptor-alpha (PPARα, transcription factor for γ-BBH and OCTN2 synthesis). In exercised mice, FC levels in plasma decreased while heart and liver OCTN2 protein expressed increased, reflecting active uptake of FC. During recovery, the rise in FC to control levels was associated with increased liver γ-BBH expression. Protein expression of PPARα was stimulated in liver after exercise and during recovery. Interestingly, heart TMLD protein was also detected after exercise. Acute exercise stimulates carnitine uptake in liver and heart. The rapid return of FC levels in plasma after exercise indicates carnitine biosynthesis by liver is stimulated to establish carnitine homeostasis. Our results suggest that exercise may benefit patients with carnitine deficiency syndromes.
Collapse
Affiliation(s)
- Tom L. Broderick
- Department of Physiology and Laboratory of Diabetes and Exercise Metabolism, Midwestern University, Glendale, AZ, USA
| | - Frank A. Cusimano
- Department of Physiology and Laboratory of Diabetes and Exercise Metabolism, Midwestern University, Glendale, AZ, USA
| | - Chelsea Carlson
- Department of Physiology and Laboratory of Diabetes and Exercise Metabolism, Midwestern University, Glendale, AZ, USA
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, USA
| |
Collapse
|
15
|
Broderick T, Cusimano F, Carlson C, Tamura L. Acute Exercise Stimulates Carnitine Biosynthesis and OCTN2 Expression in Mouse Kidney. Kidney Blood Press Res 2017; 42:398-405. [DOI: 10.1159/000478737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/19/2017] [Indexed: 11/19/2022] Open
|
16
|
Plasma Acylcarnitines and Amino Acid Levels As an Early Complex Biomarker of Propensity to High-Fat Diet-Induced Obesity in Mice. PLoS One 2016; 11:e0155776. [PMID: 27183228 PMCID: PMC4868278 DOI: 10.1371/journal.pone.0155776] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/04/2016] [Indexed: 02/08/2023] Open
Abstract
Obesity is associated with insulin resistance and impaired glucose tolerance, which represent characteristic features of the metabolic syndrome. Development of obesity is also linked to changes in fatty acid and amino acid metabolism observed in animal models of obesity as well as in humans. The aim of this study was to explore whether plasma metabolome, namely the levels of various acylcarnitines and amino acids, could serve as a biomarker of propensity to obesity and impaired glucose metabolism. Taking advantage of a high phenotypic variation in diet-induced obesity in C57BL/6J mice, 12-week-old male and female mice (n = 155) were fed a high-fat diet (lipids ~32 wt%) for a period of 10 weeks, while body weight gain (BWG) and changes in insulin sensitivity (ΔHOMA-IR) were assessed. Plasma samples were collected before (week 4) and after (week 22) high-fat feeding. Both univariate and multivariate statistical analyses were then used to examine the relationships between plasma metabolome and selected phenotypes including BWG and ΔHOMA-IR. Partial least squares-discrimination analysis was able to distinguish between animals selected either for their low or high BWG (or ΔHOMA-IR) in male but not female mice. Among the metabolites that differentiated male mice with low and high BWG, and which also belonged to the major discriminating metabolites when analyzed in plasma collected before and after high-fat feeding, were amino acids Tyr and Orn, as well as acylcarnitines C16-DC and C18:1-OH. In general, the separation of groups selected for their low or high ΔHOMA-IR was less evident and the outcomes of a corresponding multivariate analysis were much weaker than in case of BWG. Thus, our results document that plasma acylcarnitines and amino acids could serve as a gender-specific complex biomarker of propensity to obesity, however with a limited predictive value in case of the associated impairment of insulin sensitivity.
Collapse
|
17
|
Schooneman MG, Ten Have GAM, van Vlies N, Houten SM, Deutz NEP, Soeters MR. Transorgan fluxes in a porcine model reveal a central role for liver in acylcarnitine metabolism. Am J Physiol Endocrinol Metab 2015; 309:E256-64. [PMID: 26037250 DOI: 10.1152/ajpendo.00503.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/10/2015] [Indexed: 01/08/2023]
Abstract
Acylcarnitines are derived from mitochondrial acyl-CoA metabolism and have been associated with diet-induced insulin resistance. However, plasma acylcarnitine profiles have been shown to poorly reflect whole body acylcarnitine metabolism. We aimed to clarify the individual role of different organ compartments in whole body acylcarnitine metabolism in a fasted and postprandial state in a porcine transorgan arteriovenous model. Twelve cross-bred pigs underwent surgery where intravascular catheters were positioned before and after the liver, gut, hindquarter muscle compartment, and kidney. Before and after a mixed meal, we measured acylcarnitine profiles at several time points and calculated net transorgan acylcarnitine fluxes. Fasting plasma acylcarnitine concentrations correlated with net hepatic transorgan fluxes of free and C2- and C16-carnitine. Transorgan acylcarnitine fluxes were small, except for a pronounced net hepatic C2-carnitine production. The peak of the postprandial acylcarnitine fluxes was between 60 and 90 min. Acylcarnitine production or release was seen in the gut and liver and consisted mostly of C2-carnitine. Acylcarnitines were extracted by the kidney. No significant net muscle acylcarnitine flux was observed. We conclude that liver has a key role in acylcarnitine metabolism, with high net fluxes of C2-carnitine both in the fasted and fed state, whereas the contribution of skeletal muscle is minor. These results further clarify the role of different organ compartments in the metabolism of different acylcarnitine species.
Collapse
Affiliation(s)
- Marieke G Schooneman
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Gabriella A M Ten Have
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A & M University, College Station, Texas; and
| | - Naomi van Vlies
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A & M University, College Station, Texas; and
| | - Maarten R Soeters
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands;
| |
Collapse
|
18
|
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: 2.0] [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
|
19
|
Intestinal drug transporters: an overview. Adv Drug Deliv Rev 2013; 65:1340-56. [PMID: 23041352 DOI: 10.1016/j.addr.2012.09.042] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/21/2012] [Accepted: 09/24/2012] [Indexed: 02/07/2023]
Abstract
The importance of drug transporters as one of the determinants of pharmacokinetics has become increasingly evident. While much research has been conducted focusing the role of drug transporters in the liver and kidney less is known about the importance of uptake and efflux transporters identified in the intestine. Over the past years the effects of intestinal transporters have been studied using in vivo models, in situ organ perfusions, in vitro tissue preparations and cell lines. This review aims to describe up to date findings regarding the importance of intestinal transporters on drug absorption and bioavailability, highlighting areas in need of further research. Wu and Benet proposed a Biopharmaceutics Drug Disposition Classification System (BDDCS) that allows the prediction of transporter effects on the drug disposition of orally administered drugs. This review also discusses BDDCS predictions with respect to the role of intestinal transporters and intestinal transporter-metabolizing enzyme interplay on oral drug pharmacokinetics.
Collapse
|
20
|
Augmentation of normal and glutamate-impaired neuronal respiratory capacity by exogenous alternative biofuels. Transl Stroke Res 2013; 4:643-51. [PMID: 24323418 DOI: 10.1007/s12975-013-0275-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/21/2013] [Indexed: 01/07/2023]
Abstract
Mitochondrial respiratory capacity is critical for responding to changes in neuronal energy demand. One approach toward neuroprotection is the administration of alternative energy substrates ("biofuels") to overcome brain injury-induced inhibition of glucose-based aerobic energy metabolism. This study tested the hypothesis that exogenous pyruvate, lactate, β-hydroxybutyrate, and acetyl-L-carnitine each increase neuronal respiratory capacity in vitro either in the absence of or following transient excitotoxic glutamate receptor stimulation. Compared to the presence of 5 mM glucose alone, the addition of pyruvate, lactate, or β-hydroxybutyrate (1.0-10.0 mM) to either day in vitro (DIV) 14 or 7 rat cortical neurons resulted in significant, dose-dependent stimulation of respiratory capacity, measured by cell respirometry as the maximal O2 consumption rate in the presence of the respiratory uncoupler carbonyl cyanide-p-trifluoromethoxyphenylhydrazone. A 30-min exposure to 100 μM glutamate impaired respiratory capacity for DIV 14, but not DIV 7, neurons. Glutamate reduced the respiratory capacity for DIV 14 neurons with glucose alone by 25 % and also reduced respiratory capacity with glucose plus pyruvate, lactate, or β-hydroxybutyrate. However, respiratory capacity in glutamate-exposed neurons following pyruvate or β-hydroxybutyrate addition was still, at least, as high as that obtained with glucose alone in the absence of glutamate exposure. These results support the interpretation that previously observed neuroprotection by exogenous pyruvate, lactate, or β-hydroxybutyrate is at least partially mediated by their preservation of neuronal respiratory capacity.
Collapse
|
21
|
Rosenbaum S, Ringseis R, Most E, Hillen S, Becker S, Erhardt G, Reiner G, Eder K. Genes involved in carnitine synthesis and carnitine uptake are up-regulated in the liver of sows during lactation. Acta Vet Scand 2013; 55:24. [PMID: 23497718 PMCID: PMC3608077 DOI: 10.1186/1751-0147-55-24] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 03/07/2013] [Indexed: 11/13/2022] Open
Abstract
Background Convincing evidence exist that carnitine synthesis and uptake of carnitine into cells is regulated by peroxisome proliferator-activated receptor α (PPARA), a transcription factor which is physiologically activated during fasting or energy deprivation. Sows are typically in a negative energy balance during peak lactation. We investigated the hypothesis that genes involved in carnitine synthesis and uptake in the liver of sows are up-regulated during peak lactation. Findings Transcript levels of several PPARα target genes involved in fatty acid uptake (FABP4, SLC25A20), fatty acid oxidation (ACOX1, CYP4A24) and ketogenesis (HMGCS2, FGF21) were elevated in the liver of lactating compared to non-lactating sows (P < 0.05). In addition, transcript levels of genes involved in carnitine synthesis (ALDH9A1, TMLHE, BBOX1) and carnitine uptake (SLC22A5) in the liver were greater in lactating than in non-lactating sows (P < 0.05). Carnitine concentrations in liver and plasma were about 20% and 50%, respectively, lower in lactating than in non-lactating sows (P < 0.05), which is likely due to an increased loss of carnitine via the milk. Conclusions The results of the present study show that PPARα is activated in the liver of sows during lactation which leads to an up-regulation of genes involved in carnitine synthesis and carnitine uptake. The PPARα mediated up-regulation of genes involved in carnitine synthesis and uptake in the liver of lactating sows may be regarded as an adaptive mechanism to maintain hepatic carnitine levels at a level sufficient to transport excessive amounts of fatty acids into the mitochondrion.
Collapse
|
22
|
Rigault C, Le Borgne F, Tazir B, Benani A, Demarquoy J. A high-fat diet increases L-carnitine synthesis through a differential maturation of the Bbox1 mRNAs. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1831:370-7. [PMID: 23127966 DOI: 10.1016/j.bbalip.2012.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/15/2012] [Accepted: 10/26/2012] [Indexed: 12/30/2022]
Abstract
l-carnitine is a key molecule in both mitochondrial and peroxisomal lipid metabolisms. l-carnitine is biosynthesized from gamma-butyrobetaine by a reaction catalyzed by the gamma-butyrobetaine hydroxylase (Bbox1). The aim of this work was to identify molecular mechanisms involved in the regulation of l-carnitine biosynthesis and availability. Using 3' RACE, we identified four alternatively polyadenylated Bbox1 mRNAs in rat liver. We utilized a combination of in vitro experiments using hybrid constructs containing the Bbox1 3' UTR and in vivo experiments on rat liver mRNAs to reveal specificities in the different Bbox1 mRNA isoforms, especially in terms of polyadenylation efficiency, mRNA stability and translation efficiency. This complex maturation process of the Bbox1 mRNAs in the liver was studied on rats fed a high-fat diet. High-fat diet selectively increased the level of three Bbox1 mRNA isoforms in rat liver and the alternative use of polyadenylation sites contributed to the global increase in Bbox1 enzymatic activity and l-carnitine levels. Our results show that the maturation of Bbox1 mRNAs is nutritionally regulated in the liver through a selective polyadenylation process to adjust l-carnitine biosynthesis to the energy supply.
Collapse
Affiliation(s)
- Caroline Rigault
- Université de Bourgogne, BioperoxIL, EA 7270, Faculté Gabriel, 6 blvd Gabriel, 21000 Dijon, France
| | | | | | | | | |
Collapse
|
23
|
Goselink R, van Baal J, Widjaja H, Dekker R, Zom R, de Veth M, van Vuuren A. Effect of rumen-protected choline supplementation on liver and adipose gene expression during the transition period in dairy cattle. J Dairy Sci 2013. [DOI: 10.3168/jds.2012-5396] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Shibani M, Keller J, König B, Kluge H, Hirche F, Stangl G, Ringseis R, Eder K. Effects of fish oil and conjugated linoleic acids on carnitine homeostasis in laying hens. Br Poult Sci 2012; 53:431-8. [DOI: 10.1080/00071668.2012.713464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- M. Shibani
- a Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen , 35392 Gießen , Germany
| | - J. Keller
- a Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen , 35392 Gießen , Germany
| | - B. König
- b Institute of Agricultural and Nutritional Sciences, Martin-Luther-Universität Halle-Wittenberg , 06120 Halle (Saale) , Germany
| | - H. Kluge
- b Institute of Agricultural and Nutritional Sciences, Martin-Luther-Universität Halle-Wittenberg , 06120 Halle (Saale) , Germany
| | - F. Hirche
- b Institute of Agricultural and Nutritional Sciences, Martin-Luther-Universität Halle-Wittenberg , 06120 Halle (Saale) , Germany
| | - G.I. Stangl
- b Institute of Agricultural and Nutritional Sciences, Martin-Luther-Universität Halle-Wittenberg , 06120 Halle (Saale) , Germany
| | - R. Ringseis
- a Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen , 35392 Gießen , Germany
| | - K. Eder
- a Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen , 35392 Gießen , Germany
| |
Collapse
|
25
|
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: 1.7] [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
|
26
|
Schlegel G, Keller J, Hirche F, Geissler S, Schwarz FJ, Ringseis R, Stangl GI, Eder K. Expression of genes involved in hepatic carnitine synthesis and uptake in dairy cows in the transition period and at different stages of lactation. BMC Vet Res 2012; 8:28. [PMID: 22417075 PMCID: PMC3361467 DOI: 10.1186/1746-6148-8-28] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 03/14/2012] [Indexed: 12/14/2022] Open
Abstract
Background In rodents and pigs, it has shown that carnitine synthesis and uptake of carnitine into cells are regulated by peroxisome proliferator-activated receptor α (PPARA), a transcription factor which is physiologically activated during fasting or energy deprivation. Dairy cows are typically in a negative energy balance during early lactation. We investigated the hypothesis that genes of carnitine synthesis and uptake in dairy cows are enhanced during early lactation. Results mRNA abundances of PPARA and some of its classical target genes and genes involved in carnitine biosynthesis [trimethyllysine dioxygenase (TMLHE), 4-N-trimethylaminobutyraldehyde dehydrogenase (ALDH9A1), γ-butyrobetaine dioxygenase (BBOX1)] and uptake of carnitine [novel organic cation transporter 2 (SLC22A5)] as well as carnitine concentrations in liver biopsy samples of 20 dairy cows in late pregnancy (3 wk prepartum) and early lactation (1 wk, 5 wk, 14 wk postpartum) were determined. From 3 wk prepartum to 1 wk postpartum, mRNA abundances of PPARΑ and several PPARΑ target genes involved in fatty acid uptake, fatty acid oxidation and ketogenesis in the liver were strongly increased. Simultaneously, mRNA abundances of enzymes of carnitine synthesis (TMLHE: 10-fold; ALDH9A1: 6-fold; BBOX1: 1.8-fold) and carnitine uptake (SLC22A5: 13-fold) and the concentration of carnitine in the liver were increased from 3 wk prepartum to 1 wk postpartum (P < 0.05). From 1 wk to 5 and 14 wk postpartum, mRNA abundances of these genes and hepatic carnitine concentrations were declining (P < 0.05). There were moreover positive correlations between plasma concentrations of non-esterified fatty acids (NEFA) and hepatic carnitine concentrations at 1 wk, 5 wk and 14 wk postpartum (P < 0.05). Conclusions The results of this study show for the first time that the expression of hepatic genes of carnitine synthesis and cellular uptake of carnitine is enhanced in dairy cows during early lactation. These changes might provide an explanation for increased hepatic carnitine concentrations observed in 1 wk postpartum and might be regarded as a physiologic means to provide liver cells with sufficient carnitine required for transport of excessive amounts of NEFA during a negative energy balance.
Collapse
Affiliation(s)
- Gloria Schlegel
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Giessen, Giessen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
The mouse gene encoding the carnitine biosynthetic enzyme 4-N-trimethylaminobutyraldehyde dehydrogenase is regulated by peroxisome proliferator-activated receptor α. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:357-65. [PMID: 22285688 DOI: 10.1016/j.bbagrm.2012.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 01/09/2012] [Accepted: 01/10/2012] [Indexed: 11/23/2022]
Abstract
Genes involved in carnitine uptake and synthesis, such as organic cation transporter-2 (OCTN2) and γ-butyrobetaine dioxygenase (BBD), have been shown to be regulated by peroxisome proliferator-activated receptor (PPAR)α directly. Whether other genes encoding enzymes involved in the carnitine synthesis pathway, such as 4-N-trimethylaminobutyraldehyde dehydrogenase (TMABA-DH) and trimethyllysine dioxygenase (TMLD), are also direct PPARα target genes is less clear. In silico-analysis of the mouse TMLD promoter and first intron and the TMABA-DH promoter revealed several putative peroxisome proliferator response elements (PPRE) with high similarity to the consensus PPRE. Luciferase reporter gene assays using either a 2kb TMLD promoter or a 4kb TMLD first intron reporter constructs revealed no functional PPRE. In contrast, reporter gene assays using wild-type and mutated 5´-truncation TMABA-DH promoter reporter constructs showed that one PPRE located at position -132 in the proximal promoter is probably functional. Using gel shift assays we observed in vitro-binding of PPARα to this PPRE. Moreover, using chromatin immunoprecipitation assays we found that PPARα also binds in vivo to a nucleotide sequence spanning the PPRE at -132, which confirms that this PPRE is functional. In conclusion, the present study shows that the mouse TMABA-DH gene is a direct PPARα target gene. Together with the recent identification of the mouse BBD and the mouse OCTN2 genes as PPARα target genes this finding confirm that PPARα plays a key role in the regulation of carnitine homeostasis by controlling genes involved in carnitine synthesis and carnitine uptake.
Collapse
|
28
|
Role of carnitine in the regulation of glucose homeostasis and insulin sensitivity: evidence from in vivo and in vitro studies with carnitine supplementation and carnitine deficiency. Eur J Nutr 2011; 51:1-18. [PMID: 22134503 DOI: 10.1007/s00394-011-0284-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 11/17/2011] [Indexed: 10/15/2022]
Abstract
BACKGROUND Although carnitine is best known for its role in the import of long-chain fatty acids (acyl groups) into the mitochondrial matrix for subsequent β-oxidation, carnitine is also necessary for the efflux of acyl groups out of the mitochondria. Since intracellular accumulation of acyl-CoA derivatives has been implicated in the development of insulin resistance, carnitine supplementation has gained attention as a tool for the treatment of insulin resistance. More recent studies even point toward a causative role for carnitine insufficiency in developing insulin resistance during states of chronic metabolic stress, such as obesity, which can be reversed by carnitine supplementation. METHODS The present review provides an overview about data from both animal and human studies reporting effects of either carnitine supplementation or carnitine deficiency on parameters of glucose homeostasis and insulin sensitivity in order to establish the less well-recognized role of carnitine in regulating glucose homeostasis. RESULTS Carnitine supplementation studies in both humans and animals demonstrate an improvement of glucose tolerance, in particular during insulin-resistant states. In contrast, less consistent results are available from animal studies investigating the association between carnitine deficiency and glucose intolerance. The majority of studies dealing with this question could either find no association or even reported that carnitine deficiency lowers blood glucose and improves insulin sensitivity. CONCLUSIONS In view of the abovementioned beneficial effect of carnitine supplementation on glucose tolerance during insulin-resistant states, carnitine supplementation might be an effective tool for improvement of glucose utilization in obese type 2 diabetic patients. However, further studies are necessary to explain the conflicting observations from studies dealing with carnitine deficiency.
Collapse
|
29
|
Broderick TL, El Midaoui A, Chiasson JL, Wang D, Jankowski M, Gutkowska J. The effects of exercise training on γ-butyrobetaine hydroxylase and novel organic cation transporter-2 gene expression in the rat. Appl Physiol Nutr Metab 2011; 36:781-9. [DOI: 10.1139/h11-094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The concentration of carnitine in plasma is generally increased with exercise training, suggesting that either carnitine biosynthesis is stimulated or renal reabsorption of carnitine is enhanced, or both. Carnitine, an essential cofactor in the oxidation of fatty acids, is released into the plasma following hydroxylation by γ-butyrobetaine hydroxylase (BBH), the final enzyme in the biosynthetic pathway found primarily in the liver. The organic cation transporter (OCTN2), the carnitine transporter found in kidney, is important in the distribution of carnitine by facilitating its renal reabsorption from urine. In this study, we tested the hypothesis that exercise training increases gene and protein expression of BBH and OCTN2, resulting in enhanced plasma carnitine levels. Male Wistar rats were subjected to 2 daily exercise sessions of treadmill running, 5 days per week, for a 10-week period. The concentration of total carnitine in plasma was significantly increased in trained rats compared with sedentary rats. In trained rats, mRNA and protein expression of BBH were increased in liver, whereas only BBH mRNA expression was increased in kidney. Liver of trained rats demonstrated increased mRNA and protein expression of OCTN2 compared with sedentary rats. In kidney of trained rats, however, only an increase in mRNA expression of OCTN2 was observed. Our results suggest that the improved plasma carnitine status in the trained rat is associated with increased carnitine biosynthesis in liver and kidney. The observation that OCTN2 expression was increased in kidney suggests a potential role of the kidney in the reabsorption of carnitine from the urine.
Collapse
Affiliation(s)
- Tom L. Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, Midwestern University, 19555 North 59th Avenue, Glendale, AZ 85308, USA
| | - Adil El Midaoui
- Research Centre, Centre Hospitalier de l’Université de Montréal-Hôtel-Dieu, Montréal, QC H2W 1T7, Canada
| | - Jean-Louis Chiasson
- Research Centre, Centre Hospitalier de l’Université de Montréal-Hôtel-Dieu, Montréal, QC H2W 1T7, Canada
| | - Donghao Wang
- Laboratory of Cardiovascular Biochemistry, Research Centre, Centre Hospitalier de L’Université de Montréal-Hôtel-Dieu, Montréal, QC H2W 1T7, Canada
| | - Marek Jankowski
- Laboratory of Cardiovascular Biochemistry, Research Centre, Centre Hospitalier de L’Université de Montréal-Hôtel-Dieu, Montréal, QC H2W 1T7, Canada
| | - Jolanta Gutkowska
- Laboratory of Cardiovascular Biochemistry, Research Centre, Centre Hospitalier de L’Université de Montréal-Hôtel-Dieu, Montréal, QC H2W 1T7, Canada
| |
Collapse
|
30
|
Ringseis R, Mooren FC, Keller J, Couturier A, Wen G, Hirche F, Stangl GI, Eder K, Krüger K. Regular endurance exercise improves the diminished hepatic carnitine status in mice fed a high-fat diet. Mol Nutr Food Res 2011; 55 Suppl 2:S193-202. [DOI: 10.1002/mnfr.201100040] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/29/2011] [Accepted: 05/17/2011] [Indexed: 12/29/2022]
|
31
|
Wen G, Kühne H, Rauer C, Ringseis R, Eder K. Mouse γ-butyrobetaine dioxygenase is regulated by peroxisome proliferator-activated receptor α through a PPRE located in the proximal promoter. Biochem Pharmacol 2011; 82:175-83. [PMID: 21549104 DOI: 10.1016/j.bcp.2011.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 04/12/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
Abstract
Convincing evidence from studies with peroxisome proliferator-activated receptor (PPAR)α-deficient mice suggested that the carnitine biosynthetic enzyme γ-butyrobetaine dioxygenase (BBD) is regulated by PPARα. However, the identification of BBD as a direct PPARα target gene as well as its exact regulation remained to be demonstrated. In silico-analysis of the mouse BBD promoter revealed seven putative peroxisome proliferator response elements (PPRE) with high similarity to the consensus PPRE. Luciferase reporter gene assays using mutated and non-mutated serial 5'-truncation BBD promoter reporter constructs revealed that one PPRE located at -75 to -87 relative to the transcription start site in the proximal BBD promoter is probably functional. Using gel shift assays we observed in vitro-binding of PPARα/RXRα heterodimer to this PPRE confirming that it is functional. In conclusion, the present study clearly shows that mouse BBD is a direct PPARα target gene and that transcriptional up-regulation of mouse BBD by PPARα is likely mediated by binding of the PPARα/RXR heterodimer to one PPRE located in its proximal promoter region. The results confirm emerging evidence from recent studies that PPARα plays a key role in the regulation of carnitine homeostasis by controlling genes involved in both, carnitine synthesis and carnitine uptake.
Collapse
Affiliation(s)
- Gaiping Wen
- Institute of Animal Nutrition and Nutritional Physiology, Justus-Liebig-Universität, Heinrich-Buff-Ring Giessen, Germany
| | | | | | | | | |
Collapse
|
32
|
König B, Fischer S, Schlotte S, Wen G, Eder K, Stangl GI. Monocarboxylate transporter 1 and CD147 are up-regulated by natural and synthetic peroxisome proliferator-activated receptor alpha agonists in livers of rodents and pigs. Mol Nutr Food Res 2011; 54:1248-56. [PMID: 20306479 DOI: 10.1002/mnfr.200900432] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Monocarboxylate transporter (MCT)-1 mediates the transport of ketone bodies and other monocarboxylic acids across the plasma membrane. MCT1 is up-regulated by peroxisome proliferator-activated receptor (PPAR)-alpha, a transcription factor that mediates the adaptive response to fasting by up-regulation of genes involved in fatty acid oxidation and ketogenesis. Here, we show for the first time that MCT1 is up-regulated by dietary natural PPAR-alpha agonists. Both, an oxidized fat and conjugated linoleic acids increased MCT1 mRNA concentration in the liver of rats. Also, in the liver of pigs as non-proliferating species MCT1 was up-regulated upon PPAR-alpha activation by clofibrate, oxidized fat and fasting. Concomitant with up-regulation of MCT1, mRNA level of CD147 was increased in livers of rats and pigs. CD147 is a plasma membrane glycoprotein that is required for translocation and transport activity of MCT1. CD147 mRNA increase upon PPAR-alpha activation could not be observed in mice lacking PPAR-alpha, which also fail in up-regulation of MCT1 indicating a co-regulation of MCT1 and CD147. Analysis of the 5'-flanking region of mouse MCT1 gene by reporter gene assay revealed that promoter activity of mouse MCT1 was not induced by PPAR-alpha, indicating that the 5'-flanking region is not involved in MCT1 regulation by PPAR-alpha.
Collapse
Affiliation(s)
- Bettina König
- Institute of Agricultural and Nutritional Sciences, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.
| | | | | | | | | | | |
Collapse
|
33
|
Keller J, Ringseis R, Priebe S, Guthke R, Kluge H, Eder K. Dietary L-carnitine alters gene expression in skeletal muscle of piglets. Mol Nutr Food Res 2010; 55:419-29. [DOI: 10.1002/mnfr.201000293] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/24/2010] [Accepted: 09/03/2010] [Indexed: 01/21/2023]
|
34
|
Weikard R, Altmaier E, Suhre K, Weinberger KM, Hammon HM, Albrecht E, Setoguchi K, Takasuga A, Kühn C. Metabolomic profiles indicate distinct physiological pathways affected by two loci with major divergent effect on Bos taurus growth and lipid deposition. Physiol Genomics 2010; 42A:79-88. [DOI: 10.1152/physiolgenomics.00120.2010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Identifying trait-associated genetic variation offers new prospects to reveal novel physiological pathways modulating complex traits. Taking advantage of a unique animal model, we identified the I442M mutation in the non-SMC condensin I complex, subunit G ( NCAPG) gene and the Q204X mutation in the growth differentiation factor 8 ( GDF8) gene as substantial modulators of pre- and/or postnatal growth in cattle. In a combined metabolomic and genotype association approach, which is the first respective study in livestock, we surveyed the specific physiological background of the effects of both loci on body-mass gain and lipid deposition. Our data provided confirming evidence from two historically and geographically distant cattle populations that the onset of puberty is the key interval of divergent growth. The locus-specific metabolic patterns obtained from monitoring 201 plasma metabolites at puberty mirror the particular NCAPG I442M and GDF8 Q204X effects and represent biosignatures of divergent physiological pathways potentially modulating effects on proportional and disproportional growth, respectively. While the NCAPG I442M mutation affected the arginine metabolism, the 204X allele in the GDF8 gene predominantly raised the carnitine level and had concordant effects on glycerophosphatidylcholines and sphingomyelins. Our study provides a conclusive link between the well-described growth-regulating functions of arginine metabolism and the previously unknown specific physiological role of the NCAPG protein in mammalian metabolism. Owing to the confirmed effect of the NCAPG/LCORL locus on human height in genome-wide association studies, the results obtained for bovine NCAPG might add valuable, comparative information on the physiological background of genetically determined divergent mammalian growth.
Collapse
Affiliation(s)
- Rosemarie Weikard
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf
| | - Elisabeth Altmaier
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Klaus M. Weinberger
- Biocrates Life Sciences Aktiengesellschaft, Innsbruck, Austria; Research Units
| | | | - Elke Albrecht
- Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Kouji Setoguchi
- Cattle Breeding Development Institute of Kagoshima Prefecture, Osumi, So, Kagoshima; and
| | - Akiko Takasuga
- Shirikawa Institute of Animal Genetics, Japan Livestock Technology Association, Odakura, Nishigo, Fukushima, Japan
| | - Christa Kühn
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf
| |
Collapse
|
35
|
Abstract
PPARα is one of three members of the soluble nuclear receptor family called peroxisome proliferator-activated receptor (PPAR). It is a sensor for changes in levels of fatty acids and their derivatives that responds to ligand binding with PPAR target gene transcription, inasmuch as it can influence physiological homeostasis, including lipid and carbohydrate metabolism in various tissues. In this paper we summarize the involvement of PPARα in the metabolically active tissues liver and skeletal muscle and provide an overview of the risks and benefits of ligand activation of PPARα, with particular consideration to interspecies differences.
Collapse
|
36
|
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: 1.6] [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]
|
37
|
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: 3.4] [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
|
38
|
Abstract
The transition from the fed to the fasted resting state is characterized by, among other things, changes in lipid metabolism and peripheral insulin resistance. Acylcarnitines have been suggested to play a role in insulin resistance, as well as other long-chain fatty acid metabolites. Plasma levels of long-chain acylcarnitines increase during fasting, but this is unknown for muscle long-chain acylcarnitines. In the present study we investigated whether muscle long-chain acylcarnitines increase during fasting and we investigated their relationship with glucose/fat oxidation and insulin sensitivity in lean healthy humans. After 14 h and 62 h of fasting, glucose fluxes, substrate oxidation, and plasma and muscle acylcarnitines were measured before and during a hyperinsulinaemic–euglycaemic clamp. Hyperinsulinaemia decreased long-chain muscle acylcarnitines after 14 h of fasting, but not after 62 h of fasting. In both the basal state and during the clamp, glucose oxidation was lower and fatty acid oxidation was higher after 62 h compared with 14 h of fasting. Absolute changes in glucose and fatty acid oxidation in the basal compared with hyperinsulinaemic state were not different. Muscle long-chain acylcarnitines did not correlate with glucose oxidation, fatty acid oxidation or insulin-mediated peripheral glucose uptake. After 62 h of fasting, the suppression of muscle long-chain acylcarnitines by insulin was attenuated compared with 14 h of fasting. Muscle long-chain acylcarnitines do not unconditionally reflect fatty acid oxidation. The higher fatty acid oxidation during hyperinsulinaemia after 62 h compared with 14 h of fasting, although the absolute decrease in fatty acid oxidation was not different, suggests a different set point.
Collapse
|