1
|
Gillet R, Cerda-Drago TG, Brañes MC, Valenzuela R. Submicron Dispersions of Phytosterols Reverse Liver Steatosis with Higher Efficacy than Phytosterol Esters in a Diet Induced-Fatty Liver Murine Model. Int J Mol Sci 2025; 26:564. [PMID: 39859279 PMCID: PMC11766071 DOI: 10.3390/ijms26020564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Consumption of phytosterols is a nutritional strategy employed to reduce cholesterol absorption, but recent research shows that their biological activity might go beyond cholesterol reduction for the treatment of metabolic dysfunction-associated fatty liver disease (MAFLD), and novel phytosterol formulations, such as submicron dispersions, could improve these effects. We explored the therapeutic activity of phytosterols, either formulated as submicron dispersions of phytosterols (SDPs) or conventional phytosterol esters (PEs), in a mouse model of MAFLD. MAFLD was induced in mice by atherogenic diet (AD) feeding. The reversion of distorted serum and liver parameter values after a period of AD feeding was investigated after supplementation of the AD with SDPs, PEs, or a placebo (PT). Additionally, the metabolic parameters of fatty acid synthesis, fatty acid oxidation, and inflammation were studied to understand the mechanism of action of phytosterols. AD supplementation with SDPs was shown to reduce liver fat, along with showing a significant improvement in liver triglycerides (TGs), free fatty acids (FFAs), and liver cholesterol levels. These results were reinforced by the analyses of the liver steatosis scores, and liver histologies, where SDP intervention showed a consistent improvement. Treatment with PEs showed slighter effects in the same analyses, and no effects were observed with the PT treatment. Additionally, SDP intervention reversed, with a higher efficacy than PEs, the effect of AD on the serum levels of TGs, total- and LDL-cholesterol levels, and glucose levels. And, exceptionally, while SDP improved HDL-cholesterol serum levels, PEs did not show any effect on this parameter. We provide evidence for the therapeutical activity of phytosterols in MAFLD beyond the regulation of cholesterol levels, which is increased when the phytosterols are formulated as submicron dispersions compared to ester formulations.
Collapse
Affiliation(s)
- Raimundo Gillet
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Tomás G. Cerda-Drago
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - María C. Brañes
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| |
Collapse
|
2
|
Zúñiga-Hernández J, Farias C, Espinosa A, Mercado L, Dagnino-Subiabre A, Campo AD, Illesca P, Videla LA, Valenzuela R. Modulation of Δ5- and Δ6-desaturases in the brain-liver axis. Nutrition 2024; 131:112629. [PMID: 39642695 DOI: 10.1016/j.nut.2024.112629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/04/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Obesity is associated with liver depletion of ω-3 polyunsaturated fatty acids (ω-3 PUFAS) promoting steatosis and inflammation, whose levels are maintained by diet or biosynthesis involving Δ-5D, Δ-6D desaturases and elongases. METHOD We aimed to assess Δ-5D and Δ-6D activities in liver and brain from mice fed a control diet (CD) or high-fat diet (HFD) for four to sixteen weeks. RESULTS HFD led to (1) an early (4 weeks) enhancement in liver Δ-5D, Δ-6D, and PPAR-α activities, without changes in oxidative stress, liver damage or fat accumulation; (2) a latter progressive loss in hepatic desaturation with insufficient compensatory increases in mRNA and protein expression, leading to ω-3 PUFA depletion, PPAR-α down-regulation reducing FA oxidation, and liver steatosis with enhancement in lipogenesis; and (3) brain ω-3 PUFA depletion after 12 to 16 weeks of HFD feeding. CONCLUSION In conclusion, the brain-liver axis is drastically affected by obesity in a time dependent fashion.
Collapse
Affiliation(s)
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Escuela de Medicina, Campus San Felipe, Universidad de Valparaíso, San Felipe, Chile; Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Lorena Mercado
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Direccion de postgrado, Facultad Medicina, Universidad Andres Bello, Santiago, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, CIESAL, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Illesca
- Laboratorio de Estudio de Enfermedades Metabólicas Relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
3
|
Regina-Ferreira L, Valdivieso-Rivera F, Angelim MKSC, Menezes Dos Reis L, Furino VO, Morari J, Maia de Sousa L, Consonni SR, Sponton CH, Moraes-Vieira PM, Velloso LA. Inhibition of Crif1 protects fatty acid-induced POMC neuron-like cell-line damage by increasing CPT-1 function. Am J Physiol Endocrinol Metab 2024; 326:E681-E695. [PMID: 38597829 DOI: 10.1152/ajpendo.00420.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/07/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Hypothalamic proopiomelanocortin (POMC) neurons are sensors of signals that reflect the energy stored in the body. Inducing mild stress in proopiomelanocortin neurons protects them from the damage promoted by the consumption of a high-fat diet, mitigating the development of obesity; however, the cellular mechanisms behind these effects are unknown. Here, we induced mild stress in a proopiomelanocortin neuron cell line by inhibiting Crif1. In proopiomelanocortin neurons exposed to high levels of palmitate, the partial inhibition of Crif1 reverted the defects in mitochondrial respiration and ATP production; this was accompanied by improved mitochondrial fusion/fission cycling. Furthermore, the partial inhibition of Crif1 resulted in increased reactive oxygen species production, increased fatty acid oxidation, and reduced dependency on glucose for mitochondrial respiration. These changes were dependent on the activity of CPT-1. Thus, we identified a CPT-1-dependent metabolic shift toward greater utilization of fatty acids as substrates for respiration as the mechanism behind the protective effect of mild stress against palmitate-induced damage of proopiomelanocortin neurons.NEW & NOTEWORTHY Saturated fats can damage hypothalamic neurons resulting in positive energy balance, and this is mitigated by mild cellular stress; however, the mechanisms behind this protective effect are unknown. Using a proopiomelanocortin cell line, we show that under exposure to a high concentration of palmitate, the partial inhibition of the mitochondrial protein Crif1 results in protection due to a metabolic shift warranted by the increased expression and activity of the mitochondrial fatty acid transporter CPT-1.
Collapse
Affiliation(s)
| | - Fernando Valdivieso-Rivera
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Monara K S C Angelim
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | - Larissa Menezes Dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | | | - Joseane Morari
- Obesity and Comorbidities Research Center, São Paulo, Brazil
| | - Lizandra Maia de Sousa
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Sílvio Roberto Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Carlos H Sponton
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | - Lício A Velloso
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, São Paulo, Brazil
| |
Collapse
|
4
|
Guo R, Huang K, Yu K, Li J, Huang J, Wang D, Li Y. Effects of Fat and Carnitine on the Expression of Carnitine Acetyltransferase and Enoyl-CoA Hydratase Short-Chain 1 in the Liver of Juvenile GIFT ( Oreochromis niloticus). Genes (Basel) 2024; 15:480. [PMID: 38674414 PMCID: PMC11050330 DOI: 10.3390/genes15040480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Carnitine acetyltransferase (CAT) and Enoyl-CoA hydratase short-chain 1 (ECHS1) are considered key enzymes that regulate the β-oxidation of fatty acids. However, very few studies have investigated their full length and expression in genetically improved farmed tilapia (GIFT, Oreochromis niloticus), an important aquaculture species in China. Here, we cloned CAT and ECHS1 full-length cDNA via the rapid amplification of cDNA ends, and the expressions of CAT and ECHS1 in the liver of juvenile GIFT were detected in different fat and carnitine diets, as were the changes in the lipometabolic enzymes and serum biochemical indexes of juvenile GIFT in diets with different fat and carnitine levels. CAT cDNA possesses an open reading frame (ORF) of 2167 bp and encodes 461 amino acids, and the ECHS1 cDNA sequence is 1354 bp in full length, the ORF of which encodes a peptide of 391 amino acids. We found that juvenile GIFT had higher lipometabolic enzyme activity and lower blood CHOL, TG, HDL-C, and LDL-C contents when the dietary fat level was 2% or 6% and when the carnitine level was 500 mg/kg. We also found that the expression of ECHS1 and CAT genes in the liver of juvenile GIFT can be promoted by a 500 mg/kg carnitine level and 6% fat level feeding. These results suggested that CAT and ECHS1 may participate in regulating lipid metabolism, and when 2% or 6% fat and 500 mg/kg carnitine are added to the feed, it is the most beneficial to the liver and lipid metabolism of juvenile GIFT. Our results may provide a theoretical basis for GIFT feeding and treating fatty liver disease.
Collapse
Affiliation(s)
- Ruijie Guo
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| | - Kai Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| | - Kai Yu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| | - Jinghua Li
- Fisheries Research and Technology Extension Center of Shaanxi, Xi’an 710086, China;
| | - Jiao Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| | - Dandan Wang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| | - Yuda Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (R.G.); (K.Y.); (J.H.); (D.W.); (Y.L.)
| |
Collapse
|
5
|
Yong HEJ, Watkins OC, Mah TKL, Cracknell-Hazra VKB, Pillai RA, Selvam P, Islam MO, Sharma N, Cazenave-Gassiot A, Bendt AK, Wenk MR, Godfrey KM, Lewis RM, Chan SY. Increasing maternal age associates with lower placental CPT1B mRNA expression and acylcarnitines, particularly in overweight women. Front Physiol 2023; 14:1166827. [PMID: 37275238 PMCID: PMC10232777 DOI: 10.3389/fphys.2023.1166827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Older pregnant women have increased risks of complications including gestational diabetes and stillbirth. Carnitine palmitoyl transferase (CPT) expression declines with age in several tissues and is linked with poorer metabolic health. Mitochondrial CPTs catalyze acylcarnitine synthesis, which facilitates fatty acid oxidization as fuel. We hypothesized that the placenta, containing maternally-inherited mitochondria, shows an age-related CPT decline that lowers placental acylcarnitine synthesis, increasing vulnerability to pregnancy complications. We assessed CPT1A, CPT1B, CPT1C and CPT2 mRNA expression by qPCR in 77 placentas and quantified 10 medium and long-chain acylcarnitines by LC-MS/MS in a subset of 50 placentas. Older maternal age associated with lower expression of placental CPT1B, but not CPT1A, CPT1C or CPT2. CPT1B expression positively associated with eight acylcarnitines and CPT1C with three acylcarnitines, CPT1A negatively associated with nine acylcarnitines, while CPT2 did not associate with any acylcarnitine. Older maternal age associated with reductions in five acylcarnitines, only in those with BMI≥ 25 kg/m2, and not after adjusting for CPT1B expression. Our findings suggest that CPT1B is the main transferase for placental long-chain acylcarnitine synthesis, and age-related CPT1B decline may underlie decreased placental metabolic flexibility, potentially contributing to pregnancy complications in older women, particularly if they are overweight.
Collapse
Affiliation(s)
- Hannah E. J. Yong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Oliver C. Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tania K. L. Mah
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Victoria K. B. Cracknell-Hazra
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mohammad O. Islam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anne K. Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R. Wenk
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, United Kingdom
| | - Rohan M. Lewis
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Raghuvaran N, Sardar P, Sahu NP, Shamna N, Jana P, Paul M, Bhusare S, Bhavatharaniya U. Effect of L-carnitine supplemented diets with varying protein and lipid levels on growth, body composition, antioxidant status and physio-metabolic changes of white shrimp, Penaeus vannamei juveniles reared in inland saline water. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
7
|
Chang JL, Gong J, Rizal S, Peterson AL, Chang J, Yao C, Dennery PA, Yao H. Upregulating carnitine palmitoyltransferase 1 attenuates hyperoxia-induced endothelial cell dysfunction and persistent lung injury. Respir Res 2022; 23:205. [PMID: 35964084 PMCID: PMC9375342 DOI: 10.1186/s12931-022-02135-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants that may cause long-term lung dysfunction. Accumulating evidence supports the vascular hypothesis of BPD, in which lung endothelial cell dysfunction drives this disease. We recently reported that endothelial carnitine palmitoyltransferase 1a (Cpt1a) is reduced by hyperoxia, and that endothelial cell-specific Cpt1a knockout mice are more susceptible to developing hyperoxia-induced injury than wild type mice. Whether Cpt1a upregulation attenuates hyperoxia-induced endothelial cell dysfunction and lung injury remains unknown. We hypothesized that upregulation of Cpt1a by baicalin or L-carnitine ameliorates hyperoxia-induced endothelial cell dysfunction and persistent lung injury. METHODS Lung endothelial cells or newborn mice (< 12 h old) were treated with baicalin or L-carnitine after hyperoxia (50% and 95% O2) followed by air recovery. RESULTS We found that incubation with L-carnitine (40 and 80 mg/L) and baicalin (22.5 and 45 mg/L) reduced hyperoxia-induced apoptosis, impaired cell migration and angiogenesis in cultured lung endothelial cells. This was associated with increased Cpt1a gene expression. In mice, neonatal hyperoxia caused persistent alveolar and vascular simplification in a concentration-dependent manner. Treatment with L-carnitine (150 and 300 mg/kg) and baicalin (50 and 100 mg/kg) attenuated neonatal hyperoxia-induced alveolar and vascular simplification in adult mice. These effects were diminished in endothelial cell-specific Cpt1a knockout mice. CONCLUSIONS Upregulating Cpt1a by baicalin or L-carnitine ameliorates hyperoxia-induced lung endothelial cell dysfunction, and persistent alveolar and vascular simplification. These findings provide potential therapeutic avenues for using L-carnitine and baicalin as Cpt1a upregulators to prevent persistent lung injury in premature infants with BPD.
Collapse
Affiliation(s)
- Jason L Chang
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Jiannan Gong
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
- Department of Respiratory Medicine, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Salu Rizal
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Abigail L Peterson
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Julia Chang
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Chenrui Yao
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
| | - Phyllis A Dennery
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongwei Yao
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, SFH, Providence, RI, 02912, USA.
| |
Collapse
|
8
|
Zhang L, Wang Z, Wu H, Gao Y, Zheng J, Zhang J. Maternal High-Fat Diet Impairs Placental Fatty Acid β-Oxidation and Metabolic Homeostasis in the Offspring. Front Nutr 2022; 9:849684. [PMID: 35495939 PMCID: PMC9050107 DOI: 10.3389/fnut.2022.849684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 12/30/2022] Open
Abstract
Maternal overnutrition can affect fetal growth and development, thus increasing susceptibility to obesity and diabetes in later life of the offspring. Placenta is the central organ connecting the developing fetus with the maternal environment. It is indicated placental fatty acid metabolism plays an essential role in affecting the outcome of the pregnancy and fetus. However, the role of placental fatty acid β-oxidation (FAO) in maternal overnutrition affecting glucose metabolism in the offspring remains unclear. In this study, C57BL/6J female mice were fed with normal chow or high-fat diet before and during pregnancy and lactation. The placenta and fetal liver were collected at gestation day 18.5, and the offspring's liver was collected at weaning. FAO-related genes and AMP-activated protein kinase (AMPK) signaling pathway were examined both in the placenta and in the human JEG-3 trophoblast cells. FAO-related genes were further examined in the liver of the fetuses and in the offspring at weaning. We found that dams fed with high-fat diet showed higher fasting blood glucose, impaired glucose tolerance at gestation day 14.5 and higher serum total cholesterol (T-CHO) at gestation day 18.5. The placental weight and lipid deposition were significantly increased in maternal high-fat diet group. At weaning, the offspring mice of high-fat diet group exhibited higher body weight, impaired glucose tolerance, insulin resistance and increased serum T-CHO, compared with control group. We further found that maternal high-fat diet downregulated mRNA and protein expressions of carnitine palmitoyltransferase 2 (CPT2), a key enzyme in FAO, by suppressing the AMPK/Sirt1/PGC1α signaling pathway in the placenta. In JEG-3 cells, protein expressions of CPT2 and CPT1b were both downregulated by suppressing the AMPK/Sirt1/PGC1α signaling pathway under glucolipotoxic condition, but were later restored by the AMPK agonist 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR). However, there was no difference in CPT2 and CPT1 gene expression in the liver of fetuses and offspring at weaning age. In conclusion, maternal high-fat diet can impair gene expression involved in FAO in the placenta by downregulating the AMPK signaling pathway, and can cause glucose and lipid dysfunction of offspring at weaning, indicating that placental FAO may play a crucial role in regulating maternal overnutrition and metabolic health in the offspring.
Collapse
|
9
|
In vitro absorption and lipid-lowering activity of baicalin esters synthesized by whole-cell catalyzed esterification. Bioorg Chem 2022; 120:105628. [DOI: 10.1016/j.bioorg.2022.105628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/21/2022]
|
10
|
Wang M, Wang K, Liao X, Hu H, Chen L, Meng L, Gao W, Li Q. Carnitine Palmitoyltransferase System: A New Target for Anti-Inflammatory and Anticancer Therapy? Front Pharmacol 2021; 12:760581. [PMID: 34764874 PMCID: PMC8576433 DOI: 10.3389/fphar.2021.760581] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Lipid metabolism involves multiple biological processes. As one of the most important lipid metabolic pathways, fatty acid oxidation (FAO) and its key rate-limiting enzyme, the carnitine palmitoyltransferase (CPT) system, regulate host immune responses and thus are of great clinical significance. The effect of the CPT system on different tissues or organs is complex: the deficiency or over-activation of CPT disrupts the immune homeostasis by causing energy metabolism disorder and inflammatory oxidative damage and therefore contributes to the development of various acute and chronic inflammatory disorders and cancer. Accordingly, agonists or antagonists targeting the CPT system may become novel approaches for the treatment of diseases. In this review, we first briefly describe the structure, distribution, and physiological action of the CPT system. We then summarize the pathophysiological role of the CPT system in chronic obstructive pulmonary disease, bronchial asthma, acute lung injury, chronic granulomatous disease, nonalcoholic fatty liver disease, hepatic ischemia–reperfusion injury, kidney fibrosis, acute kidney injury, cardiovascular disorders, and cancer. We are also concerned with the current knowledge in either preclinical or clinical studies of various CPT activators/inhibitors for the management of diseases. These compounds range from traditional Chinese medicines to novel nanodevices. Although great efforts have been made in studying the different kinds of CPT agonists/antagonists, only a few pharmaceuticals have been applied for clinical uses. Nevertheless, research on CPT activation or inhibition highlights the pharmacological modulation of CPT-dependent FAO, especially on different CPT isoforms, as a promising anti-inflammatory/antitumor therapeutic strategy for numerous disorders.
Collapse
Affiliation(s)
- Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ximing Liao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiyang Hu
- Department of Vascular Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liangzhi Chen
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linlin Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Palombo V, Alharthi A, Batistel F, Parys C, Guyader J, Trevisi E, D'Andrea M, Loor JJ. Unique adaptations in neonatal hepatic transcriptome, nutrient signaling, and one-carbon metabolism in response to feeding ethyl cellulose rumen-protected methionine during late-gestation in Holstein cows. BMC Genomics 2021; 22:280. [PMID: 33865335 PMCID: PMC8053294 DOI: 10.1186/s12864-021-07538-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Methionine (Met) supply during late-pregnancy enhances fetal development in utero and leads to greater rates of growth during the neonatal period. Due to its central role in coordinating nutrient and one-carbon metabolism along with immune responses of the newborn, the liver could be a key target of the programming effects induced by dietary methyl donors such as Met. To address this hypothesis, liver biopsies from 4-day old calves (n = 6/group) born to Holstein cows fed a control or the control plus ethyl-cellulose rumen-protected Met for the last 28 days prepartum were used for DNA methylation, transcriptome, metabolome, proteome, and one-carbon metabolism enzyme activities. RESULTS Although greater withers and hip height at birth in Met calves indicated better development in utero, there were no differences in plasma systemic physiological indicators. RNA-seq along with bioinformatics and transcription factor regulator analyses revealed broad alterations in 'Glucose metabolism', 'Lipid metabolism, 'Glutathione', and 'Immune System' metabolism due to enhanced maternal Met supply. Greater insulin sensitivity assessed via proteomics, and efficiency of transsulfuration pathway activity suggested beneficial effects on nutrient metabolism and metabolic-related stress. Maternal Met supply contributed to greater phosphatidylcholine synthesis in calf liver, with a role in very low density lipoprotein secretion as a mechanism to balance metabolic fates of fatty acids arising from the diet or adipose-depot lipolysis. Despite a lack of effect on hepatic amino acid (AA) transport, a reduction in metabolism of essential AA within the liver indicated an AA 'sparing effect' induced by maternal Met. CONCLUSIONS Despite greater global DNA methylation, maternal Met supply resulted in distinct alterations of hepatic transcriptome, proteome, and metabolome profiles after birth. Data underscored an effect on maintenance of calf hepatic Met homeostasis, glutathione, phosphatidylcholine and taurine synthesis along with greater efficiency of nutrient metabolism and immune responses. Transcription regulators such as FOXO1, PPARG, E2F1, and CREB1 appeared central in the coordination of effects induced by maternal Met. Overall, maternal Met supply induced better immunometabolic status of the newborn liver, conferring the calf a physiologic advantage during a period of metabolic stress and suboptimal immunocompetence.
Collapse
Affiliation(s)
- Valentino Palombo
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, via De Sanctis snc, 86100, Campobasso, Italy
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA
| | - Abdulrahman Alharthi
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fernanda Batistel
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Claudia Parys
- Evonik Operations GmbH, Hanau-Wolfgang, 63457, Essen, Germany
| | - Jessie Guyader
- Evonik Operations GmbH, Hanau-Wolfgang, 63457, Essen, Germany
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - Mariasilvia D'Andrea
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, via De Sanctis snc, 86100, Campobasso, Italy
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA.
| |
Collapse
|
12
|
Xue Y, Liu H, Yang XX, Pang L, Liu J, Ng KTP, Yeung OWH, Lam YF, Zhang WY, Lo CM, Man K. Inhibition of Carnitine Palmitoyltransferase 1A Aggravates Fatty Liver Graft Injury via Promoting Mitochondrial Permeability Transition. Transplantation 2021; 105:550-560. [PMID: 32890136 DOI: 10.1097/tp.0000000000003437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Hepatic steatosis is a major risk factor for graft failure due to increased susceptibility of fatty liver to ischemia-reperfusion injury (IRI) during transplantation. Here, we aimed to investigate the role of carnitine palmitoyltransferase 1A (CPT1A) in fatty liver graft injury and to explore the underlying mechanism and therapeutic potential on attenuating hepatic IRI. METHODS Intragraft CPT1A expression profile and the association with fatty graft injury were investigated in human and rat liver transplantation samples. The underlying mechanism and therapeutic potential of CPT1A activator against IRI were also explored in mouse hepatic ischemia-reperfusion plus major hepatectomy model and in in vitro. RESULTS CPT1A expression was significantly reduced (P = 0.0019; n = 96) in human fatty liver graft compared with normal one at early phase after transplantation. Low expression of CPT1A was significantly associated with high serum alanine aminotransferase (P = 0.0144) and aspartate aminotransferase (P = 0.0060) levels. The inhibited CPT1A and poor liver function were consistently observed in rat and mouse models with fatty livers. Furthermore, inhibition of CPT1A significantly promoted the translocation of chloride intracellular channel 1 to form chloride ion channel. The dysregulation of chloride ion channel activity subsequently triggered mitochondrial permeability transition (MPT) pore opening, exacerbated cellular oxidative stress, and energy depletion. Importantly, our intravital confocal imaging showed that CPT1A activation attenuated hepatic injury through preventing MPT after reperfusion in fatty mice. CONCLUSIONS CPT1A inhibition triggered MPT contributed to severe IRI in fatty liver graft. CPT1A restoration may offer therapeutic potential on attenuating hepatic IRI.
Collapse
Affiliation(s)
- Yan Xue
- Department of Surgery, HKU-SZH &LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
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
|
14
|
Li Z, Wu YY, Yu BX. Methylophiopogonanone A, an Ophiopogon homoisoflavonoid, alleviates high-fat diet-induced hyperlipidemia: assessment of its potential mechanism. ACTA ACUST UNITED AC 2020; 53:e9201. [PMID: 32130294 PMCID: PMC7057930 DOI: 10.1590/1414-431x20199201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 12/04/2019] [Indexed: 11/22/2022]
Abstract
Methylophiopogonanone A (MO-A), a homoisoflavonoid extracted from Ophiopogon japonicus, has been shown to attenuate myocardial apoptosis and improve cerebral ischemia/reperfusion injury. However, the hypolipidemic effects remain unknown. This study was performed to investigate a potential hypolipidemic effect of MO-A in hyperlipidemia rats, as well as its underlying mechanism of action. A rat model of hyperlipidemia was induced by a high-fat diet (HFD). Animals were randomly divided into three groups (n=8/group): normal control group (NC), HFD group, and HFD+MO-A (10 mg·kg-1·d-1) treatment group. The effects of MO-A on serum lipids, body weight, activity of lipoprotein metabolism enzyme, and gene expression of lipid metabolism were evaluated in HFD-induced rats. In HFD-induced rats, pretreatment with MO-A decreased the body weight gain and reduced serum and hepatic lipid levels. In addition, pretreatment with MO-A improved the activities of lipoprotein lipase and hepatic lipase in serum and liver, down-regulated mRNA expression of acetyl CoA carboxylase and sterol regulatory element-binding protein 1c, and up-regulated mRNA expression of low-density lipoprotein receptor and peroxisome proliferator-activated receptor α in the liver. Our results indicated that MO-A showed strong ability to ameliorate the hyperlipidemia in HFD-induced rats. MO-A might be a potential candidate for prevention of overweight and dyslipidemia induced by HFD.
Collapse
Affiliation(s)
- Zhao Li
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| | - Ying-Ying Wu
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| | - Bei-Xin Yu
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| |
Collapse
|
15
|
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: 361] [Impact Index Per Article: 72.2] [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
|
16
|
Ruan D, Fouad AM, Zhang YN, Wang S, Chen W, Xia WG, Jiang SQ, Yang L, Zheng CT. Effects of dietary lysine on productivity, reproductive performance, protein and lipid metabolism-related gene expression in laying duck breeders. Poult Sci 2020; 98:5734-5745. [PMID: 31265113 DOI: 10.3382/ps/pez361] [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: 01/29/2019] [Accepted: 06/09/2019] [Indexed: 02/01/2023] Open
Abstract
This study investigated whether dietary lysine (Lys) affects productive performance and expression of genes related to protein and lipid metabolism in laying duck breeders. Longyan duck breeders (n = 540, 19 wk of age) were randomly assigned to 6 groups with 6 replicates of 15 birds each. Breeders were fed diets with 6 total Lys levels (6.4, 7.2, 8.0, 8.8, 9.6, and 10.4 g/kg) for 26 wk duration. Egg production, egg weight, egg mass, feed conversion ratio, hatchability, hatchling weight, albumen weight, eggshell weight, yolk weight, and yolk proportion increased with dietary Lys levels (P < 0.05). Dietary Lys level had a linear (P < 0.05) and quadratic (P < 0.05) effects on maternal hepatic expression of mechanistic target of rapamycin, eukaryotic translation initiation factor 4E binding protein 1, ubiquitin conjugating enzyme E2K (UBE2K), cathepsin B (CTSB), and quadratically (P < 0.05) increased the concentrations of plasma Lys, leucine, threonine, and tryptophan in duck breeders. In contrast, maternal dietary Lys suppressed expression of proteasome 26S subunit, UBE2K, and CTSB in the liver of hatchlings. Moreover, relative expression of peroxisome proliferator-activated receptors alpha, carnitine palmitoyltransferase 1A, and very low density apolipoprotein-II increased linearly (P < 0.05) and quadratically (P < 0.05), and that of VLDL receptor (VLDLR) decreased quadratically (P < 0.05) in the liver of duck breeders with increasing dietary Lys levels; hepatic triglyceride and cholesterol contents were reduced. Maternal dietary Lys suppressed hepatic expression of VLDLR in the hatchlings. A diet containing 8.6 g Lys/kg promoted protein turnover and lipid metabolism in laying duck breeders, which positively reflected in the productivity and reproductive performance.
Collapse
Affiliation(s)
- D Ruan
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China.,College of Animal Science, South China Agricultural University, Guangzhou 510640, P. R. China
| | - A M Fouad
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China.,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Y N Zhang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - S Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - W Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - W G Xia
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - S Q Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| | - L Yang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, P. R. China
| | - C T Zheng
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture; State Key Laboratory of Livestock and Poultry Breeding; Guangdong Pubic Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China
| |
Collapse
|
17
|
Soto-Alarcón SA, Ortiz M, Orellana P, Echeverría F, Bustamante A, Espinosa A, Illesca P, Gonzalez-Mañán D, Valenzuela R, Videla LA. Docosahexaenoic acid and hydroxytyrosol co-administration fully prevents liver steatosis and related parameters in mice subjected to high-fat diet: A molecular approach. Biofactors 2019; 45:930-943. [PMID: 31454114 DOI: 10.1002/biof.1556] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022]
Abstract
Attenuation of high-fat diet (HFD)-induced liver steatosis is accomplished by different nutritional interventions. Considering that the n-3 PUFA docosahexaenoic acid (DHA) modulates lipid metabolism and the antioxidant hydroxytyrosol (HT) diminishes oxidative stress underlying fatty liver, it is hypothesized that HFD-induced steatosis is suppressed by DHA and HT co-administration. Male C57BL/6J mice were fed a control diet (CD; 10% fat, 20% protein, 70% carbohydrates) or a HFD (60% fat, 20% protein, 20% carbohydrates) for 12 weeks, without and with supplementation of DHA (50 mg/kg/day), HT (5 mg/kg/day) or both. The combined DHA + HT protocol fully prevented liver steatosis and the concomitant pro-inflammatory state induced by HFD, with suppression of lipogenic and oxidative stress signaling, recovery of fatty acid oxidation capacity and enhancement in resolvin availability affording higher inflammation resolution capability. Abrogation of HFD-induced hepatic steatosis by DHA and HT co-administration represents a crucial therapeutic strategy eluding disease progression into stages lacking efficacious handling at present time.
Collapse
Affiliation(s)
| | - Macarena Ortiz
- Nutrition and Dietetics School, Faculty of Health Sciences, Catholic University of Maule, Curicó, Chile
| | - Paula Orellana
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Andrés Bustamante
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Illesca
- Biochemistry Department, Faculty of Biochemistry, University of Litoral, Santa Fe, Argentina
| | | | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
18
|
Echeverría F, Valenzuela R, Bustamante A, Álvarez D, Ortiz M, Espinosa A, Illesca P, Gonzalez-Mañan D, Videla LA. High-fat diet induces mouse liver steatosis with a concomitant decline in energy metabolism: attenuation by eicosapentaenoic acid (EPA) or hydroxytyrosol (HT) supplementation and the additive effects upon EPA and HT co-administration. Food Funct 2019; 10:6170-6183. [PMID: 31501836 DOI: 10.1039/c9fo01373c] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High-fat-diet (HFD) feeding is associated with liver oxidative stress (OS), n-3 long-chain polyunsaturated fatty acid (n-3 LCPUFA) depletion, hepatic steatosis and mitochondrial dysfunction. Our hypothesis is that the HFD-induced liver injury can be attenuated by the combined supplementation of n-3 LCPUFA eicosapentaenoic acid (EPA) and the antioxidant hydroxytyrosol (HT). The C57BL/6J mice were administered an HFD (60% fat, 20% protein, 20% carbohydrates) or control diet (CD; 10% fat, 20% protein, 70% carbohydrates), with or without EPA (50 mg kg-1 day-1), HT (5 mg kg-1 day-1), or EPA + HT (50 and 5 mg kg-1 day-1, respectively) for 12 weeks. We measured the body and liver weights and dietary and energy intakes along with liver histology, FA composition, steatosis score and associated transcription factors, mitochondrial functions and metabolic factors related to energy sensing through the AMP-activated protein kinase (AMPK) and PPAR-γ coactivator-1α (PGC-1α) cascade. It was found that the HFD significantly induced liver steatosis, with a 66% depletion of n-3 LCPUFAs and a 100% increase in n-6/n-3 LCPUFA ratio as compared to the case of CD (p < 0.05). These changes were concomitant with (i) a 95% higher lipogenic and 70% lower FA oxidation signaling, (ii) a 40% diminution in mitochondrial respiratory capacity and (iii) a 56% lower ATP content. HFD-induced liver steatosis was also associated with (iv) a depressed mRNA expression of AMPK-PGC-1α signaling components, nuclear respiratory factor-2 (NRF-2) and β-ATP synthase. These HFD effects were significantly attenuated by the combined EPA + HT supplementation in an additive manner. These results suggested that EPA and HT co-administration partly prevented HFD-induced liver steatosis, thus strengthening the importance of combined interventions in hepatoprotection in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Francisca Echeverría
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Andrés Bustamante
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Daniela Álvarez
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Macarena Ortiz
- Nutrition and Dietetics School, Faculty of Health Sciences, Catholic University of Maule, Curicó, Chile
| | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Illesca
- Biochemistry Department, Faculty of Biochemistry, University of Litoral, Santa Fe, Argentina
| | | | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
19
|
Gong J, Zhao H, Liu T, Li L, Cheng E, Zhi S, Kong L, Yao HW, Li J. Cigarette Smoke Reduces Fatty Acid Catabolism, Leading to Apoptosis in Lung Endothelial Cells: Implication for Pathogenesis of COPD. Front Pharmacol 2019; 10:941. [PMID: 31555131 PMCID: PMC6727183 DOI: 10.3389/fphar.2019.00941] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/24/2019] [Indexed: 11/13/2022] Open
Abstract
Endothelial cell (EC) apoptosis contributes to cigarette smoke (CS)-induced pulmonary emphysema. Metabolism of glucose, glutamine, and fatty acid is dysregulated in patients with chronic obstructive pulmonary disease (COPD). Whether CS causes metabolic dysregulation in ECs leading to development of COPD remains elusive. We hypothesized that CS alters metabolism, resulting in apoptosis in lung ECs. To test this hypothesis, we treated primary mouse pulmonary microvascular ECs (PMVECs) with CS extract (CSE) and employed PMVECs from healthy subjects and COPD patients. We found that mitochondrial respiration was reduced in CSE-treated PMVECs and in PMVECs from COPD patients. Specifically, oxidation of fatty acids (FAO) was reduced in these cells, which linked to reduced carnitine palmitoyltransferase 1a (Cpt1a), an essential enzyme for carnitine shuttle. CSE-induced apoptosis was further increased when cells were treated with a specific Cpt1 inhibitor etomoxir or transfected with Cpt1a siRNA. L-Carnitine treatment augmented FAO but attenuated CSE-induced apoptosis by upregulating Cpt1a. CSE treatment increased palmitate-derived ceramide synthesis, which was reduced by L-carnitine. Although CSE treatment increased glycolysis, inhibiting glycolysis with 2-deoxy-d-glucose had no effects on CSE-mediated apoptosis in lung ECs. Conclusively, FAO reduction increases ceramide and apoptosis in lung ECs treated with CSE, which may contribute to the pathogenesis of COPD/emphysema.
Collapse
Affiliation(s)
- Jiannan Gong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Tanzhen Liu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lifang Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Erjing Cheng
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuyin Zhi
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lufei Kong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hong-Wei Yao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianqiang Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
20
|
Gu L, Larson Casey JL, Andrabi SA, Lee JH, Meza-Perez S, Randall TD, Carter AB. Mitochondrial calcium uniporter regulates PGC-1α expression to mediate metabolic reprogramming in pulmonary fibrosis. Redox Biol 2019; 26:101307. [PMID: 31473487 PMCID: PMC6831865 DOI: 10.1016/j.redox.2019.101307] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease with an increased mortality. Metabolic reprogramming has a critical role in multiple chronic diseases. Lung macrophages expressing the mitochondrial calcium uniporter (MCU) have a critical role in fibrotic repair, but the contribution of MCU in macrophage metabolism is not known. Here, we show that MCU regulates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and metabolic reprogramming to fatty acid oxidation (FAO) in macrophages. MCU regulated PGC-1α expression by increasing the phosphorylation of ATF-2 by the p38 MAPK in a redox-dependent manner. The expression and activation of PGC-1α via the p38 MAPK was regulated by MCU-mediated mitochondrial calcium uptake, which is linked to increased mitochondrial ROS (mtROS) production. Mice harboring a conditional expression of dominant-negative MCU in macrophages had a marked reduction in mtROS and FAO and were protected from pulmonary fibrosis. Moreover, IPF lung macrophages had evidence of increased MCU and mitochondrial calcium, increased phosphorylation of ATF2 and p38, as well as increased expression of PGC-1α. These observations suggest that macrophage MCU-mediated metabolic reprogramming contributes to fibrotic repair after lung injury.
Collapse
Affiliation(s)
- Linlin Gu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jennifer L Larson Casey
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Shaida A Andrabi
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jun Hee Lee
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - A Brent Carter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Birmingham VAMC, Birmingham, AL, 35294, USA.
| |
Collapse
|
21
|
Geisler CE, Ghimire S, Bogan RL, Renquist BJ. Role of ketone signaling in the hepatic response to fasting. Am J Physiol Gastrointest Liver Physiol 2019; 316:G623-G631. [PMID: 30767679 PMCID: PMC6580236 DOI: 10.1152/ajpgi.00415.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ketosis is a metabolic adaptation to fasting, nonalcoholic fatty liver disease (NAFLD), and prolonged exercise. β-OH butyrate acts as a transcriptional regulator and at G protein-coupled receptors to modulate cellular signaling pathways in a hormone-like manner. While physiological ketosis is often adaptive, chronic hyperketonemia may contribute to the metabolic dysfunction of NAFLD. To understand how β-OH butyrate signaling affects hepatic metabolism, we compared the hepatic fasting response in control and 3-hydroxy-3-methylglutaryl-CoA synthase II (HMGCS2) knockdown mice that are unable to elevate β-OH butyrate production. To establish that rescue of ketone metabolic/endocrine signaling would restore the normal hepatic fasting response, we gave intraperitoneal injections of β-OH butyrate (5.7 mmol/kg) to HMGCS2 knockdown and control mice every 2 h for the final 9 h of a 16-h fast. In hypoketonemic, HMGCS2 knockdown mice, fasting more robustly increased mRNA expression of uncoupling protein 2 (UCP2), a protein critical for supporting fatty acid oxidation and ketogenesis. In turn, exogenous β-OH butyrate administration to HMGCS2 knockdown mice decreased fasting UCP2 mRNA expression to that observed in control mice. Also supporting feedback at the transcriptional level, β-OH butyrate lowered the fasting-induced expression of HMGCS2 mRNA in control mice. β-OH butyrate also regulates the glycemic response to fasting. The fast-induced fall in serum glucose was absent in HMGCS2 knockdown mice but was restored by β-OH butyrate administration. These data propose that endogenous β-OH butyrate signaling transcriptionally regulates hepatic fatty acid oxidation and ketogenesis, while modulating glucose tolerance. NEW & NOTEWORTHY Ketogenesis regulates whole body glucose metabolism and β-OH butyrate produced by the liver feeds back to inhibit hepatic β-oxidation and ketogenesis during fasting.
Collapse
Affiliation(s)
- Caroline E. Geisler
- School of Animal and Comparative Biomedical Science, University of Arizona, Tucson, Arizona
| | - Susma Ghimire
- School of Animal and Comparative Biomedical Science, University of Arizona, Tucson, Arizona
| | - Randy L. Bogan
- School of Animal and Comparative Biomedical Science, University of Arizona, Tucson, Arizona
| | - Benjamin J. Renquist
- School of Animal and Comparative Biomedical Science, University of Arizona, Tucson, Arizona
| |
Collapse
|
22
|
Reis LMD, Adamoski D, Ornitz Oliveira Souza R, Rodrigues Ascenção CF, Sousa de Oliveira KR, Corrêa-da-Silva F, Malta de Sá Patroni F, Meira Dias M, Consonni SR, Mendes de Moraes-Vieira PM, Silber AM, Dias SMG. Dual inhibition of glutaminase and carnitine palmitoyltransferase decreases growth and migration of glutaminase inhibition-resistant triple-negative breast cancer cells. J Biol Chem 2019; 294:9342-9357. [PMID: 31040181 DOI: 10.1074/jbc.ra119.008180] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/25/2019] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancers (TNBCs) lack progesterone and estrogen receptors and do not have amplified human epidermal growth factor receptor 2, the main therapeutic targets for managing breast cancer. TNBCs have an altered metabolism, including an increased Warburg effect and glutamine dependence, making the glutaminase inhibitor CB-839 therapeutically promising for this tumor type. Accordingly, CB-839 is currently in phase I/II clinical trials. However, not all TNBCs respond to CB-839 treatment, and the tumor resistance mechanism is not yet fully understood. Here we classified cell lines as CB-839-sensitive or -resistant according to their growth responses to CB-839. Compared with sensitive cells, resistant cells were less glutaminolytic and, upon CB-839 treatment, exhibited a smaller decrease in ATP content and less mitochondrial fragmentation, an indicator of poor mitochondrial health. Transcriptional analyses revealed that the expression levels of genes linked to lipid metabolism were altered between sensitive and resistant cells and between breast cancer tissues (available from The Cancer Genome Atlas project) with low versus high glutaminase (GLS) gene expression. Of note, CB-839-resistant TNBC cells had increased carnitine palmitoyltransferase 2 (CPT2) protein and CPT1 activity levels. In agreement, CB-839-resistant TNBC cells mobilized more fatty acids into mitochondria for oxidation, which responded to AMP-activated protein kinase and acetyl-CoA carboxylase signaling. Moreover, chemical inhibition of both glutaminase and CPT1 decreased cell proliferation and migration of CB-839-resistant cells compared with single inhibition of each enzyme. We propose that dual targeting of glutaminase and CPT1 activities may have therapeutic relevance for managing CB-839-resistant tumors.
Collapse
Affiliation(s)
- Larissa Menezes Dos Reis
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil.,the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Douglas Adamoski
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil.,the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Rodolpho Ornitz Oliveira Souza
- the Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Science, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
| | - Carolline Fernanda Rodrigues Ascenção
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil.,the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Krishina Ratna Sousa de Oliveira
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil.,the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Felipe Corrêa-da-Silva
- the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil.,the Department of Genetics, Evolution, Microbiology, and Immunology, Laboratory of Immunometabolism, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil, and
| | - Fábio Malta de Sá Patroni
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil.,the Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Marília Meira Dias
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil
| | - Sílvio Roberto Consonni
- the Department of Biochemistry and Tissue Biology, Laboratory of Cytochemistry and Immunocytochemistry, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil
| | - Pedro Manoel Mendes de Moraes-Vieira
- the Department of Genetics, Evolution, Microbiology, and Immunology, Laboratory of Immunometabolism, Institute of Biology, University of Campinas, 13083-970 Campinas, São Paulo, Brazil, and
| | - Ariel Mariano Silber
- the Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Science, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
| | - Sandra Martha Gomes Dias
- From the Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil,
| |
Collapse
|
23
|
Davani-Davari D, Karimzadeh I, Sagheb MM, Khalili H. The Renal Safety of L-Carnitine, L-Arginine, and Glutamine in Athletes and Bodybuilders. J Ren Nutr 2018; 29:221-234. [PMID: 30341034 DOI: 10.1053/j.jrn.2018.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/07/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023] Open
Abstract
One of the major concerns about taking amino acid supplements is their potential adverse effects on the kidney as a major organ involved in the metabolism and excretion of exogenous substances. The aim of this study is to review available data about renal safety of the most prominent amino acid supplements including L-arginine, glutamine and also L-carnitine as well as creatine (as amino acid derivatives) in athletes and bodybuilders. The literature was searched by keywords such as "L-carnitine", "L-arginine", "glutamine", and "kidney injury" in databases such as Scopus, Medline, Embase, and ISI Web of Knowledge. Articles published from 1950 to December 2017 were included. Among 3171, 5740, and 1608 records after primary search in the relevant databases, 8, 7, and 5 studies have been finally included, respectively, for L-carnitine, L-arginine, and glutamine in this review. Arginine appears to have both beneficial and detrimental effects on kidney function. However, adverse effects are unlikely to occur with the routine doses (from 3 to >100 g/day). The risks and benefits of L-carnitine on the athletes' and bodybuilders' kidney have not been evaluated yet. However, L-carnitine up to 6000 mg/day is generally considered to be a safe supplement at least in healthy adults. Both short-term (20-30 g within a few hours) and long-term (0.1 g/kg four times daily for 2 weeks) glutamine supplementation in healthy athletes were associated with no significant adverse effects, but it can cause glomerulosclerosis and serum creatinine level elevation in the setting of diabetic nephropathy. Creatine supplementation (ranged from 5 to 30 g/day) also appears to have no detrimental effects on kidney function of individuals without underlying renal diseases. More clinical data are warranted to determine the optimal daily dose and intake duration of common supplemental amino acids associated with the lowest renal adverse effects in sportsmen and sports women.
Collapse
Affiliation(s)
- Dorna Davani-Davari
- Pharmaceutical Biotechnology Incubator, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Karimzadeh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Mahdi Sagheb
- Nephrology-Urology Research Center and Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Khalili
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Barrera C, Valenzuela R, Rincón MÁ, Espinosa A, Echeverria F, Romero N, Gonzalez-Mañan D, Videla LA. Molecular mechanisms related to the hepatoprotective effects of antioxidant-rich extra virgin olive oil supplementation in rats subjected to short-term iron administration. Free Radic Biol Med 2018; 126:313-321. [PMID: 30153476 DOI: 10.1016/j.freeradbiomed.2018.08.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/27/2022]
Abstract
Enhanced iron levels in liver are associated with oxidative stress development and damage with increased fat accumulation. The aim of this work was to assess the hypothesis that antioxidant-rich extra virgin olive oil (AR-EVOO) counteracts iron-rich diet (IRD)-induced oxidative stress hindering hepatic steatosis. Male Wistar rats were fed and IRD (200 mg iron/kg diet) versus a control diet (CD; 50 mg iron/kg diet) with alternate AR-EVOO supplementation (100 mg/day) for 21 days. IRD induced liver steatosis and oxidative stress (higher levels of protein oxidation and lipid peroxidation with glutathione depletion), mitochondrial dysfunction (decreased citrate synthase and complex I and II activities) and loss of polyunsaturated fatty acids (PUFAs), with a drastic enhancement in the sterol regulatory element-binding protein-1c (SREBP-1c)/peroxisome proliferator-activated receptor-α (PPAR-α) ratio upregulating the expression of lipogenic enzymes (acetyl-CoA carboxylase, fatty acid (FA) synthase and stearoyl desaturase 2) and downregulating those involved in FA oxidation (carnitine palmitoyl transferase and acyl-CoA oxidase) over values in the CD group. IRD also upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) and its target genes. AR-EVOO supplementation alone did not modify the studied parameters, however, IRD combined with AR-EVOO administration returned IRD-induced changes to baseline levels of the CD group. It is concluded that IRD-induced non-alcoholic fatty liver disease (NAFLD) is prevented by AR-EVOO supplementation, which might be related to the protective effects of its components such as hydroxytyrosol, oleic acid, tocopherols and/or PUFAs, thus representing a suitable anti-steatotic strategy to avoid progression into more severe stages of the disease, underlying NAFLD associated with iron overloading pathologies or obesity.
Collapse
Affiliation(s)
- Cynthia Barrera
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Lipid Center, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile.
| | - Miguel Ángel Rincón
- Lipid Center, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Francisca Echeverria
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nalda Romero
- Department of Food Science and Chemical Technology, Faculty of Chemical Sciences and Pharmacy, University of Chile, Santiago, Chile
| | | | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago-7, Chile
| |
Collapse
|
25
|
Tan Z, Xiao L, Tang M, Bai F, Li J, Li L, Shi F, Li N, Li Y, Du Q, Lu J, Weng X, Yi W, Zhang H, Fan J, Zhou J, Gao Q, Onuchic JN, Bode AM, Luo X, Cao Y. Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy. Am J Cancer Res 2018; 8:2329-2347. [PMID: 29721083 PMCID: PMC5928893 DOI: 10.7150/thno.21451] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 02/28/2018] [Indexed: 12/17/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has a particularly high prevalence in southern China, southeastern Asia and northern Africa. Radiation resistance remains a serious obstacle to successful treatment in NPC. This study aimed to explore the metabolic feature of radiation-resistant NPC cells and identify new molecular-targeted agents to improve the therapeutic effects of radiotherapy in NPC. Methods: Radiation-responsive and radiation-resistant NPC cells were used as the model system in vitro and in vivo. Metabolomics approach was used to illustrate the global metabolic changes. 13C isotopomer tracing experiment and Seahorse XF analysis were undertaken to determine the activity of fatty acid oxidation (FAO). qRT-PCR was performed to evaluate the expression of essential FAO genes including CPT1A. NPC tumor tissue microarray was used to investigate the prognostic role of CPT1A. Either RNA interference or pharmacological blockade by Etomoxir were used to inhibit CPT1A. Radiation resistance was evaluated by colony formation assay. Mitochondrial membrane potential, apoptosis and neutral lipid content were measured by flow cytometry analysis using JC-1, Annexin V and LipidTOX Red probe respectively. Molecular markers of mitochondrial apoptosis were detected by western blot. Xenografts were treated with Etomoxir, radiation, or a combination of Etomoxir and radiation. Mitochondrial apoptosis and lipid droplets content of tumor tissues were detected by cleaved caspase 9 and Oil Red O staining respectively. Liquid chromatography coupled with tandem mass spectrometry approach was used to identify CPT1A-binding proteins. The interaction of CPT1A and Rab14 were detected by immunoprecipitation, immunofluorescence and in situ proximity ligation analysis. Fragment docking and direct coupling combined computational protein-protein interaction prediction method were used to predict the binding interface. Fatty acid trafficking was measured by pulse-chase assay using BODIPY C16 and MitoTracker Red probe. Results: FAO was active in radiation-resistant NPC cells, and the rate-limiting enzyme of FAO, carnitine palmitoyl transferase 1 A (CPT1A), was consistently up-regulated in these cells. The protein level of CPT1A was significantly associated with poor overall survival of NPC patients following radiotherapy. Inhibition of CPT1A re-sensitized NPC cells to radiation therapy by activating mitochondrial apoptosis both in vitro and in vivo. In addition, we identified Rab14 as a novel CPT1A binding protein. The CPT1A-Rab14 interaction facilitated fatty acid trafficking from lipid droplets to mitochondria, which decreased radiation-induced lipid accumulation and maximized ATP production. Knockdown of Rab14 attenuated CPT1A-mediated fatty acid trafficking and radiation resistance. Conclusion: An active FAO is a vital signature of NPC radiation resistance. Targeting CPT1A could be a beneficial regimen to improve the therapeutic effects of radiotherapy in NPC patients. Importantly, the CPT1A-Rab14 interaction plays roles in CPT1A-mediated radiation resistance by facilitating fatty acid trafficking. This interaction could be an attractive interface for the discovery of novel CPT1A inhibitors.
Collapse
|
26
|
Meng Q, Sun S, Sun Y, Li J, Wu D, Shan A, Shi B, Cheng B. Effects of dietary lecithin and l-carnitine on fatty acid composition and lipid-metabolic genes expression in subcutaneous fat and longissimus thoracis of growing-finishing pigs. Meat Sci 2018; 136:68-78. [DOI: 10.1016/j.meatsci.2017.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 01/22/2023]
|
27
|
Valenzuela R, Rincón-Cervera MÁ, Echeverría F, Barrera C, Espinosa A, Hernández-Rodas MC, Ortiz M, Valenzuela A, Videla LA. Iron-induced pro-oxidant and pro-lipogenic responses in relation to impaired synthesis and accretion of long-chain polyunsaturated fatty acids in rat hepatic and extrahepatic tissues. Nutrition 2018; 45:49-58. [DOI: 10.1016/j.nut.2017.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022]
|
28
|
Hernández-Rodas MC, Valenzuela R, Echeverría F, Rincón-Cervera MÁ, Espinosa A, Illesca P, Muñoz P, Corbari A, Romero N, Gonzalez-Mañan D, Videla LA. Supplementation with Docosahexaenoic Acid and Extra Virgin Olive Oil Prevents Liver Steatosis Induced by a High-Fat Diet in Mice through PPAR-α and Nrf2 Upregulation with Concomitant SREBP-1c and NF-kB Downregulation. Mol Nutr Food Res 2017; 61. [PMID: 28940752 DOI: 10.1002/mnfr.201700479] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/29/2017] [Indexed: 12/15/2022]
Abstract
SCOPE Nonalcoholic fatty liver disease is the most common cause of liver disease, for which there is no validated drug therapy at present time. In this respect, the PUFA docosahexaenoic acid (DHA; C22:6 n-3) modulate lipid metabolism in the liver, and extra virgin olive oil (EVOO) has hepatoprotective effects. METHODS AND RESULTS The effect of combined DHA (C22:6 n-3) and EVOO administration to mice on oxidative stress and metabolic disturbances induced by high-fat diet (HFD) is evaluated. Male C57BL/6J mice are fed with a control diet (10% fat, 20% protein, and 70% carbohydrates) or an HFD (60% fat, 20% protein, and 20% carbohydrates) for 12 weeks. Animals are supplemented with DHA (50 mg/kg/day), EVOO (50 mg/kg/day), or DHA + EVOO through oral route. DHA + EVOO cosupplementation results in greater protection (p < 0.05) over that elicited by DHA or EVOO supply alone, when compared to the damage induced by HFD. DHA + EVOO significantly reduces hepatic steatosis, oxidative stress, systemic inflammation, and insulin resistance. CONCLUSION Synergistic beneficial effects of DHA + EVOO supplementation are associated with the activation/inactivation of key transcription factors involved in the above-mentioned processes. Data presented indicate that dietary supplementation with DHA + EVOO drastically reduces the development of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Illesca
- Department of Biochemistry, School of Biochemistry, University of Litoral, Santa Fe, Argentina
| | - Patricio Muñoz
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alicia Corbari
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nalda Romero
- Faculty of Chemical Sciences and Pharmacy, Department of Food Science and Chemical Technology, University of Chile, Santiago, Chile
| | - Daniel Gonzalez-Mañan
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
29
|
Valenzuela R, Illesca P, Echeverría F, Espinosa A, Rincón-Cervera MÁ, Ortiz M, Hernandez-Rodas MC, Valenzuela A, Videla LA. Molecular adaptations underlying the beneficial effects of hydroxytyrosol in the pathogenic alterations induced by a high-fat diet in mouse liver: PPAR-α and Nrf2 activation, and NF-κB down-regulation. Food Funct 2017; 8:1526-1537. [PMID: 28386616 DOI: 10.1039/c7fo00090a] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SCOPE Non-alcoholic fatty liver disease (NAFLD) is a condition characterized by an increment in the liver fat content, with a concomitant reduction in the content of n-3-long chain polyunsaturated fatty acids (n-3 LCPUFAs), downregulation of PPAR-α activity, and upregulation of NF-κB activity, effects that induce pro-lipogenic and pro-inflammatory responses. Hydroxytyrosol (HT), a polyphenol with cytoprotective effects present in extra virgin olive oil, improves the cellular antioxidant capacity for activation of transcription factor Nrf2. The objective of this work is to evaluate the molecular adaptations involved in the anti-lipogenic, anti-inflammatory, and anti-oxidant effects of HT supplementation in high-fat diet (HFD)-fed mice. METHODS AND RESULTS Male C57BL/6J mice received (i) control diet (10% fat); (ii) control diet + HT (daily doses of 5 mg per kg body weight), (iii) HFD (60% fat); or (iv) HFD + HT for 12 weeks. HFD-fed mice exhibited (i) liver steatosis; (ii) inflammation; (iii) oxidative stress; and (iv) depletion of n-3 LCPUFAs, together with down-regulation of PPAR-α and Nrf2, and up-regulation of NF-κB. HT supplementation attenuated the metabolic alterations produced by HFD, normalizing the activity of Nrf2, reducing the drop in activity of PPAR-α, and attenuating increment of NF-κB activation. CONCLUSION Supplementation with HT activating transcription factors PPAR-α and Nrf2, along with the deactivation of NF-κB, may reduce the liver alterations induced in HFD-fed mice.
Collapse
Affiliation(s)
- Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Fortino MA, Oliva ME, Rodriguez S, Lombardo YB, Chicco A. Could post-weaning dietary chia seed mitigate the development of dyslipidemia, liver steatosis and altered glucose homeostasis in offspring exposed to a sucrose-rich diet from utero to adulthood? Prostaglandins Leukot Essent Fatty Acids 2017; 116:19-26. [PMID: 28088290 DOI: 10.1016/j.plefa.2016.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/20/2022]
Abstract
The present work analyzes the effects of dietary chia seeds during postnatal life in offspring exposed to a sucrose-rich diet (SRD) from utero to adulthood. At weaning, chia seed (rich in α-linolenic acid) replaced corn oil (rich in linoleic acid) in the SRD. At 150 days of offspring life, anthropometrical parameters, blood pressure, plasma metabolites, hepatic lipid metabolism and glucose homeostasis were analyzed. Results showed that chia was able to prevent the development of hypertension, liver steatosis, hypertriglyceridemia and hypercholesterolemia. Normal triacylglycerol secretion and triacylglycerol clearance were accompanied by an improvement of de novo hepatic lipogenic and carnitine-palmitoyl transferase-1 enzymatic activities, associated with an accretion of n-3 polyunsaturated fatty acids in the total composition of liver homogenate. Glucose homeostasis and plasma free fatty acid levels were improved while visceral adiposity was slightly decreased. These results confirm that the incorporation of chia seed in the diet in postnatal life may provide a viable therapeutic option for preventing/mitigating adverse outcomes induced by an SRD from utero to adulthood.
Collapse
Affiliation(s)
- M A Fortino
- Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, El Pozo, CC 242, 3000 Santa Fe, Argentina
| | - M E Oliva
- Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, El Pozo, CC 242, 3000 Santa Fe, Argentina
| | - S Rodriguez
- Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, El Pozo, CC 242, 3000 Santa Fe, Argentina
| | - Y B Lombardo
- Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, El Pozo, CC 242, 3000 Santa Fe, Argentina
| | - A Chicco
- Departamento de Ciencias Biológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, El Pozo, CC 242, 3000 Santa Fe, Argentina.
| |
Collapse
|
31
|
Sayed-Ahmed MM, Darweesh AQ, Fatani AJ. Carnitine deficiency and oxidative stress provoke cardiotoxicity in an ifosfamide-induced Fanconi Syndrome rat model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 3:266-74. [PMID: 20972373 PMCID: PMC2952087 DOI: 10.4161/oxim.3.4.12859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In addition to hemorrhagic cystitis, Fanconi Syndrome is a serious clinical side effect during ifosfamide (IFO) therapy. Fanconi syndrome is a generalized dysfunction of the proximal tubule which is characterized by excessive urinary excretion of glucose, phosphate, bicarbonate, amino acids and other solutes excreted by this segment of the nephron including L-carnitine. Carnitine is essential cofactor for β-oxidation of long-chain fatty acids in the myocardium. IFO therapy is associated with increased urinary carnitine excretion with subsequent secondary deficiency of the molecule. Cardiac abnormalities in IFO-treated cancer patients were reported as isolated clinical cases. This study examined whether carnitine deficiency and oxidative stress, secondary to Fanconi Syndrome, provoke IFO-induced cardiomyopathy as well as exploring if carnitine supplementation using Propionyl-L-carnitine (PLC) could offer protection against this toxicity. In the current study, an animal model of carnitine deficiency was developed in rats by D-carnitine-mildronate treatment Adult male Wistar albino rats were assigned to one of six treatment groups: the first three groups were injected intraperitoneally with normal saline, D-carnitine (DC, 250 mg/kg/day) combined with mildronate (MD, 200 mg/kg/day) and PLC (250 mg/kg/day), respectively, for 10 successive days. The 4th, 5th and 6th groups were injected with the same doses of normal saline, DC-MD and PLC, respectively for 5 successive days before and 5 days concomitant with IFO (50 mg/kg/day). IFO significantly increased serum creatinine, blood urea nitrogen (BUN), urinary carnitine excretion and clearance, creatine phosphokinase isoenzyme (CK-MB), lactate dehydrogenase (LDH), intramitochondrial acetyl-CoA/CoA-SH and thiobarbituric acid reactive substances (TBARS) in cardiac tissues and significantly decreased adenosine triphosphate (ATP) and total carnitine and reduced glutathione (GSH) content in cardiac tissues. In carnitine-depleted rats, IFO induced dramatic increase in serum creatinine, BUN, CK-MB, LDH, carnitine clearance and intramitochondrial acetyl-CoA/CoA-SH, as well as progressive reduction in total carnitine and ATP in cardiac tissues. Interestingly, PLC supplementation completely reversed the biochemical changes-induced by IFO to the control values. In conclusion, data from the present study suggest that: Carnitine deficiency and oxidative stress, secondary to Fanconi Syndrome, constitute risk factors and should be viewed as mechanisms during development of IFO-induced cardiotoxicity. Carnitine supplementation, using PLC, prevents the development of IFO-induced cardiotoxicity through antioxidant signalling and improving mitochondrial function.
Collapse
Affiliation(s)
- Mohamed M Sayed-Ahmed
- Department of Pharmacology; College of Pharmacy; King Saud University; Riyadh, Kingdom of Saudi Arabia.
| | - Amal Q Darweesh
- Department of Pharmacology; College of Pharmacy; King Saud University; Riyadh, Kingdom of Saudi Arabia
| | - Amal J Fatani
- Department of Pharmacology; College of Pharmacy; King Saud University; Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Sung GJ, Choi HK, Kwak S, Song JH, Ko H, Yoon HG, Kang HB, Choi KC. Targeting CPT1A enhances metabolic therapy in human melanoma cells with the BRAF V600E mutation. Int J Biochem Cell Biol 2016; 81:76-81. [PMID: 27793752 DOI: 10.1016/j.biocel.2016.10.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/10/2016] [Accepted: 10/21/2016] [Indexed: 11/26/2022]
Abstract
Cancer cells are characterized by altered metabolic processes. Recent evidence of metabolic alterations has indicated that the fatty acid oxidation (FAO) pathway is used as a carbon source for anabolic processes in some tumors, thus making this pathway a potential target for therapy. The carnitine palmitoyltransferase (CPT; EC 2.3.1.21) enzyme transfers long-chain fatty acids from the cytosol to the mitochondrial matrix for β-oxidation. Because carnitine palmitoyl transferase 1a (CPT1a) is the rate-limiting enzyme for FAO, the authors evaluated the effects of CPT1A knock-down in BRAF V600E melanoma cell lines. The results showed that knock-down of CPT1A inhibited FAO and that CPT1A is critical for malignant V600E melanoma cells, particularly BRAF V600E melanoma cells. The proliferation and tumorigenesis in V600E melanoma were decrease after CPT1A knockdown. These results suggest that therapy for BRAF V600E melanoma can include targeting metabolic alterations. CPT1A is more important for lipid synthesis in V600E mutant melanoma cells than in wild-type BRAF melanoma cells.
Collapse
Affiliation(s)
- Gi-Jun Sung
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo-Kyoung Choi
- Division of Nutrition and Metabolism Research Group, Korea Food Research Institute, Gyeonggi-do, South Korea
| | - Sungmin Kwak
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji-Hye Song
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyeonseok Ko
- Laboratory of Molecular Oncology, Cheil General Hospital & Women's Healthcare Center, Dankuk University College of Medicine, Seoul, South Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Center for Chronic Metabolic Disease Research, Brain Korea 21 Project for Medical Sciences, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee-Bum Kang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
33
|
Chicco A, Creus A, Illesca P, Hein GJ, Rodriguez S, Fortino A. Effects of post-suckling n-3 polyunsaturated fatty acids: prevention of dyslipidemia and liver steatosis induced in rats by a sucrose-rich diet during pre- and post-natal life. Food Funct 2016; 7:445-54. [PMID: 26511757 DOI: 10.1039/c5fo00705d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The interaction between fetal programming and the post-natal environment suggests that the post-natal diet could amplify or attenuate programmed outcomes. We investigated whether dietary n-3 long-chain polyunsaturated fatty acids (n-3 PUFAs) at weaning resulted in an amelioration of dyslipidemia, adiposity and liver steatosis that was induced by a sucrose-rich diet (SRD; where the fat source is corn oil) from the onset of pregnancy up to adulthood. During pregnancy and lactation, dams were fed an SRD or the standard powdered rodent commercial diet (RD). At weaning and until 150 days of life, male offspring from SRD-dams were divided into two groups and fed an SRD or SRD-with-fish oil [where 6% of the corn oil was partially replaced by fish oil (FO) 5% and corn oil (CO) 1%], forming SRD-SRD or SRD-FO groups. Male offspring from RD-dams continued with RD up to the end of the experimental period, forming an RD-RD group. The presence of FO in the weaning diet showed the following: prevention of hypertriglyceridemia and liver steatosis, together with increased lipogenic enzyme activity caused by a maternal SRD; the complete normalization of CPT I activity and PPARα protein mass levels; a slight but not statistically significant accretion of visceral adiposity; and limited body fat content and reduced plasma free fatty acid levels. All of these results were observed even in the presence of a high-sucrose diet challenge after weaning. SRD-dams' breast milk showed a more saturated fatty acid composition. These results suggest the capacity of n-3 PUFAs to overcome some adverse outcomes induced by a maternal and post-weaning sucrose-rich diet.
Collapse
Affiliation(s)
- Adriana Chicco
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| | - Agustina Creus
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| | - Paola Illesca
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| | - Gustavo Juan Hein
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| | - Silvia Rodriguez
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| | - Alejandra Fortino
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo CC 242, (3000) Santa Fe, Argentina.
| |
Collapse
|
34
|
Valenzuela R, Espinosa A, Llanos P, Hernandez-Rodas MC, Barrera C, Vergara D, Romero N, Pérez F, Ruz M, Videla LA. Anti-steatotic effects of an n-3 LCPUFA and extra virgin olive oil mixture in the liver of mice subjected to high-fat diet. Food Funct 2016; 7:140-50. [PMID: 26471014 DOI: 10.1039/c5fo01086a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by liver steatosis, oxidative stress, and drastic depletion of n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA), namely, eicosapentaenoic acid (C20:5 n-3, EPA) and docosahexaenoic acid (C22:6 n-3, DHA), which trigger lipolysis stimulation and lipogenesis inhibition. Extra virgin olive oil (EVOO) has important antioxidant effects. This study evaluated the anti-steatotic effects of n-3 LCPUFA plus EVOO in the liver of male C57BL/6J mice subjected to a control diet (CD) (10% fat, 20% protein, 70% carbohydrate) or high fat diet (HFD) (60% fat, 20% protein, 20% carbohydrate), without and with supplementation with n-3 LCPUFA (100 mg per kg per day) plus EVOO (100 mg per kg per day) for 12 weeks. HFD induced (i) liver steatosis (increased total fat, triacylglycerols, and free fatty acid total contents), (ii) higher fasting serum glucose and insulin levels and HOMA index, total cholesterol, triacylglycerols and TNF-α and IL-6, (iii) liver and plasma oxidative stress enhancement, (iv) depletion of the n-3 LCPUFA hepatic content, and (v) increment in lipogenic enzyme activity and reduction in lipolytic enzyme activity. These changes were either reduced (p < 0.05) or normalized to control the values in animals subjected to HFD supplemented with n-3 LCPUFA plus EVOO. In conclusion, n-3 LCPUFA plus EVOO intervention exerts anti-steatotic effects underlying antioxidant and anti-inflammatory responses, improved insulin sensitivity, and recovery of the lipolytic/lipogenic status of the liver altered by HFD, and supports the potential therapeutic use of n-3 LCPUFA plus EVOO supplementation in the treatment of human liver steatosis induced by nutritional factors or other etiologies.
Collapse
Affiliation(s)
- Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Espinosa
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Faculty of Dentistry, University of Chile, Santiago, Chile
| | | | - Cynthia Barrera
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Daniela Vergara
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Nalda Romero
- Faculty of Chemical Sciences and Pharmacy, Department of Food Science and Chemical Technology, University of Chile, Santiago, Chile
| | - Francisco Pérez
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Manuel Ruz
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
35
|
Pooyandjoo M, Nouhi M, Shab-Bidar S, Djafarian K, Olyaeemanesh A. The effect of (L-)carnitine on weight loss in adults: a systematic review and meta-analysis of randomized controlled trials. Obes Rev 2016; 17:970-6. [PMID: 27335245 DOI: 10.1111/obr.12436] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/21/2016] [Accepted: 05/09/2016] [Indexed: 11/27/2022]
Abstract
This study provides a systematic review and meta-analysis of randomized controlled trials, which have examined the effect of the carnitine on adult weight loss. Relevant studies were identified by systematic search of PubMed, Embase, Cochrane Central Register of Controlled Trials and reference lists of relevant marker studies. Nine studies (total n = 911) of adequate methodological quality were included in the review. Trials with mean difference (MD) of 95% confidence interval (CI) were pooled using random effect model. Results from meta-analysis of eligible trials revealed that subjects who received carnitine lost significantly more weight (MD: -1.33 kg; 95% CI: -2.09 to -0.57) and showed a decrease in body mass index (MD: -0.47 kg m(-2) ; 95% CI: -0.88 to -0.05) compared with the control group. The results of meta-regression analysis of duration of consumption revealed that the magnitude of weight loss resulted by carnitine supplementation significantly decreased over time (p = 0.002). We conclude that receiving the carnitine resulted in weight loss. Using multiple-treatments meta-analysis of the drugs and non-pharmacotherapy options seem to be insightful areas for research. © 2016 World Obesity.
Collapse
Affiliation(s)
- M Pooyandjoo
- School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - M Nouhi
- Health Management and Economics Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - S Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - K Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - A Olyaeemanesh
- National institute of health research, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Rincón-Cervera MÁ, Valenzuela R, Hernandez-Rodas MC, Barrera C, Espinosa A, Marambio M, Valenzuela A. Vegetable oils rich in alpha linolenic acid increment hepatic n-3 LCPUFA, modulating the fatty acid metabolism and antioxidant response in rats. Prostaglandins Leukot Essent Fatty Acids 2016; 111:25-35. [PMID: 26995676 DOI: 10.1016/j.plefa.2016.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 11/29/2022]
Abstract
Alpha-linolenic acid (C18:3 n-3, ALA) is an essential fatty acid and the metabolic precursor of long-chain polyunsaturated fatty acids (LCPUFA) from the n-3 family with relevant physiological and metabolic roles: eicosapentaenoic acid (C20:5 n-3, EPA) and docosahexaenoic acid (C22:6 n-3, DHA). Western diet lacks of suitable intake of n-3 LCPUFA and there are recommendations to increase the dietary supply of such nutrients. Seed oils rich in ALA such as those from rosa mosqueta (Rosa rubiginosa), sacha inchi (Plukenetia volubis) and chia (Salvia hispanica) may constitute an alternative that merits research. This study evaluated hepatic and epididymal accretion and biosynthesis of n-3 LCPUFA, the activity and expression of Δ-5 and Δ-6 desaturase enzymes, the expression and DNA-binding activity of PPAR-α and SREBP-1c, oxidative stress parameters and the activity of antioxidative enzymes in rats fed sunflower oil (SFO, 1% ALA) as control group, canola oil (CO, 10% ALA), rosa mosqueta oil (RMO, 33% ALA), sacha inchi oil (SIO, 49% ALA) and chia oil (ChO, 64% ALA) as single lipid source. A larger supply of ALA increased the accretion of n-3 LCPUFA, the activity and expression of desaturases, the antioxidative status, the expression and DNA-binding of PPAR-α, the oxidation of fatty acids and the activity of antioxidant enzymes, whereas the expression and DNA-binding activity of SREBP-1c transcription factor and the biosynthetic activity of fatty acids declined. Results showed that oils rich in ALA such as SIO and ChO may trigger metabolic responses in rats such as those produced by n-3 PUFA.
Collapse
Affiliation(s)
| | - Rodrigo Valenzuela
- Lipid Center, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile; Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | - Cynthia Barrera
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Macarena Marambio
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alfonso Valenzuela
- Lipid Center, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile; Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| |
Collapse
|
37
|
Rincón-Cervera MA, Valenzuela R, Hernandez-Rodas MC, Marambio M, Espinosa A, Mayer S, Romero N, Barrera M Sc C, Valenzuela A, Videla LA. Supplementation with antioxidant-rich extra virgin olive oil prevents hepatic oxidative stress and reduction of desaturation capacity in mice fed a high-fat diet: Effects on fatty acid composition in liver and extrahepatic tissues. Nutrition 2016; 32:1254-67. [PMID: 27346714 DOI: 10.1016/j.nut.2016.04.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/30/2016] [Accepted: 04/22/2016] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The aim of this study was to assess the effect of dietary supplementation with extra virgin olive oil (EVOO) in mice on the reduction of desaturase and antioxidant enzymatic activities in liver, concomitantly with long-chain polyunsaturated fatty acids (LCPUFA) profiles in liver and extrahepatic tissues induced by a high-fat diet (HFD). METHODS Male mice C57 BL/6 J were fed with a control diet (CD; 10% fat, 20% protein, 70% carbohydrates) or an HFD (60% fat, 20% protein, 20% carbohydrates) for 12 wk. Animals were supplemented with 100 mg/d EVOO with different antioxidant contents (EVOO I, II, and III). RESULTS After the intervention, blood and several tissues were analyzed. Dietary supplementation with EVOO with the highest antioxidant content and antioxidant capacity (EVOO III) significantly reduced fat accumulation in liver and the plasmatic metabolic alterations caused by HFD and produced a normalization of oxidative stress-related parameters, desaturase activities, and LCPUFA content in tissues. CONCLUSIONS Data suggest that dietary supplementation with EVOO III may prevent oxidative stress and reduction of biosynthesis and accretion of ω-3 LCPUFA in the liver of HFD-fed mice.
Collapse
Affiliation(s)
| | - Rodrigo Valenzuela
- Lipid Center, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile; Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | - Macarena Marambio
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Susana Mayer
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nalda Romero
- Faculty of Chemical Sciences and Pharmacy, Department of Food Science and Chemical Technology, University of Chile, Santiago, Chile
| | | | - Alfonso Valenzuela
- Lipid Center, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
38
|
Liu P, Liu X, Hu J, Han Z, Li F, Wang Y, Song L, Chen Z. Carnitine palmitoyl transferase 2 polymorphism may be associated with enterovirus 71 severe infection in a Chinese population. Arch Virol 2016; 161:1217-27. [PMID: 26874509 DOI: 10.1007/s00705-016-2785-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 01/31/2016] [Indexed: 12/13/2022]
Abstract
Genetic polymorphism in the carnitine palmitoyl transferase 2 (CPT2) gene has been reported to be a susceptibility factor in a number of syndromes of acute encephalopathy with various infectious diseases, but evidence of its effect on enterovirus 71 (EV71) infection is lacking. The goal of this study was to examine the relationship between genetic polymorphism of CPT2 and severity of EV71 infection in a Chinese population. PCR of five exons of the CPT2 gene was carried out to identify single-nucleotide polymorphisms (SNPs) in EV71-infected subjects (n = 333), including mild cases (n = 271) and severe cases (n = 62) as well as healthy controls (n = 328). Blood ATP levels were measured within 24 h of admission. The frequency of the A allele of rs1799821 (P = 0.023) and the G allele of rs2229291 (P = 0.009) in the CPT2 gene was higher in patients with severe EV71 infection. The A-G haplotype of rs1799821and rs2229291 was directly linked to EV71 severe infection risk when compared to all other haplotypes (OR = 2.005, 95 % CI = 1.087-3.700, P = 0.024). The blood ATP levels of severe cases were significantly lower than in mild cases (P < 0.01) and controls (P < 0.01). A significant negative correlation was observed in haplotype A-G between ATP levels and physical findings in severe cases (P < 0.05). These findings suggest that CPT2 polymorphism may be associated with severity of EV71 infection and that the A-G haplotype of the CPT2 gene is involved in the inflammatory process of EV71 infection.
Collapse
Affiliation(s)
- Peipei Liu
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China
| | - Xiangping Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingfei Hu
- NICU, Qingdao Women & Children's Hospital, Qingdao, China
| | - Zhenliang Han
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China
| | - Fei Li
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China
| | - Yuanyuan Wang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China
| | - Long Song
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China
| | - Zongbo Chen
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, NO. 59, Haier Road, Qingdao, 266000, China.
| |
Collapse
|
39
|
Panchal SK, Poudyal H, Ward LC, Waanders J, Brown L. Modulation of tissue fatty acids by L-carnitine attenuates metabolic syndrome in diet-induced obese rats. Food Funct 2015; 6:2496-506. [PMID: 26190559 DOI: 10.1039/c5fo00480b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity and dyslipidaemia are metabolic defects resulting from impaired lipid metabolism. These impairments are associated with the development of cardiovascular disease and non-alcoholic fatty liver disease. Correcting the defects in lipid metabolism may attenuate obesity and dyslipidaemia, and reduce cardiovascular risk and liver damage. L-Carnitine supplementation was used in this study to enhance fatty acid oxidation so as to ameliorate diet-induced disturbances in lipid metabolism. Male Wistar rats (8-9 weeks old) were fed with either corn starch or high-carbohydrate, high-fat diets for 16 weeks. Separate groups were supplemented with L-carnitine (1.2% in food) on either diet for the last 8 weeks of the protocol. High-carbohydrate, high-fat diet-fed rats showed central obesity, dyslipidaemia, hypertension, impaired glucose tolerance, hyperinsulinaemia, cardiovascular remodelling and non-alcoholic fatty liver disease. L-Carnitine supplementation attenuated these high-carbohydrate, high-fat diet-induced changes, together with modifications in lipid metabolism including the inhibition of stearoyl-CoA desaturase-1 activity, reduced storage of short-chain monounsaturated fatty acids in the tissues with decreased linoleic acid content and trans fatty acids stored in retroperitoneal fat. Thus, L-carnitine supplementation attenuated the signs of metabolic syndrome through inhibition of stearoyl-CoA desaturase-1 activity, preferential β-oxidation of some fatty acids and increased storage of saturated fatty acids and relatively inert oleic acid in the tissues.
Collapse
Affiliation(s)
- Sunil K Panchal
- Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD 4350, Australia.
| | | | | | | | | |
Collapse
|
40
|
Chronic inflammation aggravates metabolic disorders of hepatic fatty acids in high-fat diet-induced obese mice. Sci Rep 2015; 5:10222. [PMID: 25974206 PMCID: PMC4431481 DOI: 10.1038/srep10222] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/07/2015] [Indexed: 02/06/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) increases with increasing body mass index (BMI). However, approximately 40–50% of obese adults do not develop hepatic steatosis. The level of inflammatory biomarkers is higher in obese subjects with NAFLD compared to BMI-matched subjects without hepatic steatosis. We used a casein injection in high-fat diet (HFD)-fed C57BL/6J mice to induce inflammatory stress. Although mice on a HFD exhibited apparent phenotypes of obesity and hyperlipidemia regardless of exposure to casein injection, only the HFD+Casein mice showed increased hepatic vacuolar degeneration accompanied with elevated inflammatory cytokines in the liver and serum, compared to mice on a normal chow diet. The expression of genes related to hepatic fatty acid synthesis and oxidation were upregulated in the HFD-only mice. The casein injection further increased baseline levels of lipogenic genes and decreased the levels of oxidative genes in HFD-only mice. Inflammatory stress induced both oxidative stress and endoplasmic reticulum stress in HFD-fed mice livers. We conclude that chronic inflammation precedes hepatic steatosis by disrupting the balance between fatty acid synthesis and oxidation in the livers of HFD-fed obese mice. This mechanism may operate in obese individuals with chronic inflammation, thus making them more prone to NAFLD.
Collapse
|
41
|
Targeting prostate cancer cell metabolism: impact of hexokinase and CPT-1 enzymes. Tumour Biol 2014; 36:2893-905. [PMID: 25501281 DOI: 10.1007/s13277-014-2919-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/28/2014] [Indexed: 10/24/2022] Open
Abstract
Glycolysis has been shown to be required for the cell growth and proliferation in several cancer cells. However, prostate cancer cells were accused of using more fatty acid than glucose to meet their bioenergetic demands. The present study was designed to evaluate the involvement of hexokinase and CPT-1 in the cell growth and proliferation of human prostate cancer cell lines, PC3, and LNCaP-FGC-10. Hexokinase and CPT-1 activities were examined in the presence of different concentrations of their inhibitors, lonidamine and etomoxir, to find the concentration of maximum inhibition ([I max]). To assess cell viability and proliferation, dimethylthiazol (MTT) assay was carried out using [I max] for 24, 48, and 72 h on PC3 and LNCaP cells. Apoptosis was determined using annexin-V, caspase-3 activity assay, Hoechst 33258 staining, and evaluation of mitochondrial membrane potential (MMP). Moreover, ATP levels were measured following lonidamine and etomoxir exposure. In addition, to define the impact of exogenous fatty acid on the cell growth and proliferation, CPT-1 activity was evaluated in the presence of palmitate (50 μM). Hexokinase and CPT-1 activities were significantly inhibited by lonidamine [600 μM] and etomoxir [100 μM] in both cell lines. Treatment of the cells with lonidamine [600 μM] resulted in a significant ATP reduction, cell viability and apoptosis, caspase-3 activity elevation, MMP reduction, and appearance of apoptosis-related morphological changes in the cells. In contrast, etomoxir [100 μM] just decreased ATP levels in both cell lines without significant cell death and apoptosis. Compared with glucose (2 g/L), palmitate intensified CPT-1 activity in both cell lines, especially in LNCaP cells. In addition, activity of CPT-1 was higher in LNCaP than PC3 cells. Our results suggest that prostate cancer cells may metabolize glucose as a source of bioenergetic pathways. ATP could also be produced by long-chain fatty acid oxidation. In addition, these data might suggest that LNCaP is more compatible with palmitate.
Collapse
|
42
|
Wong HS, Chen J, Leong PK, Leung HY, Chan WM, Ko KM. Cistanches Herba reduces the weight gain in high fat diet-induced obese mice possibly through mitochondrial uncoupling. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
43
|
Mechanisms underlying the beneficial effect of soy protein in improving the metabolic abnormalities in the liver and skeletal muscle of dyslipemic insulin resistant rats. Eur J Nutr 2014; 54:407-19. [DOI: 10.1007/s00394-014-0721-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/13/2014] [Indexed: 01/18/2023]
|
44
|
D'Alessandro ME, Oliva ME, Fortino MA, Chicco A. Maternal sucrose-rich diet and fetal programming: changes in hepatic lipogenic and oxidative enzymes and glucose homeostasis in adult offspring. Food Funct 2014; 5:446-53. [DOI: 10.1039/c3fo60436e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
45
|
Dionne S, Elimrani I, Roy MJ, Qureshi IA, Sarma DR, Levy E, Seidman EG. Studies on the chemopreventive effect of carnitine on tumorigenesis in vivo, using two experimental murine models of colon cancer. Nutr Cancer 2013; 64:1279-87. [PMID: 23163856 DOI: 10.1080/01635581.2012.722247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Carnitine is known for its essential role in intermediary metabolism. In vitro studies suggest that its antioxidant and anti-inflammatory properties are potentially beneficial toward cancer prevention. This study tested effects of carnitine on the development of colon cancer in vivo using 2 murine models: azoxymethane (AOM) treatment as a model of carcinogen-induced colon cancer and a genetically induced model using Apc (Min/+) mice. AOM and Apc (Min/+) mice divided into dietary groups varying in lipid content, with or without carnitine supplementation (0.08%). AOM-exposed mice on a high butterfat diet had significantly increased aberrant crypts (ACF) (9.3 ± 0.88 vs. 6.3 ± 0.65), and macroscopic tumors (3.8 ± 0.95 vs. 2.0 ± 0.25) compared to mice on a control diet. In AOM mice fed the high butterfat diet, carnitine supplementation inhibited ACF (4.9 ± 0.7 vs. 9.3 ± 0.88, P < 0.001), crypt multiciplicity (1.6 ± 0.08 vs. 1.92 ± 0.1, P < 0.01) and tumors (1.5 ± 0.38 vs. 3.8 ± 0.95, P < 0.001). Carnitine supplementation resulted in significantly increased tissue carnitine and acylcarnitine levels. Carnitine inhibited the development of precancerous lesions and macroscopic colonic tumors in AOM-treated mice. However, carnitine did not exert protective effects on intestinal tumors in Apc (Min/+) mice.
Collapse
Affiliation(s)
- Serge Dionne
- Division of Gastroenterology, Research Institute, McGill University Health Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
Inhibition of p53 attenuates steatosis and liver injury in a mouse model of non-alcoholic fatty liver disease. J Hepatol 2013; 58:785-91. [PMID: 23211317 PMCID: PMC3612370 DOI: 10.1016/j.jhep.2012.11.042] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 10/26/2012] [Accepted: 11/23/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS p53 and its transcriptional target miRNA34a have been implicated in the pathogenesis of fatty liver. We tested the efficacy of a p53 inhibitor, pifithrin-α p-nitro (PFT) in attenuating steatosis, associated oxidative stress and apoptosis in a murine model of non-alcoholic fatty liver disease (NAFLD). METHODS C57BL/6 mice were fed a high-fat (HFD) or control diet for 8 weeks; PFT or DMSO (vehicle) was administered three times per week. Markers of oxidative stress and apoptosis as well as mediators of hepatic fatty acid metabolism were assessed by immunohistochemistry, Western blot, real-time PCR, and biochemical assays. RESULTS PFT administration suppressed HFD-induced weight gain, ALT elevation, steatosis, oxidative stress, and apoptosis. PFT treatment blunted the HFD-induced upregulation of miRNA34a and increased SIRT1 expression. In the livers of HFD-fed, PFT-treated mice, activation of the SIRT1/PGC1α/PPARα axis increased the expression of malonyl-CoA decarboxylase (MLYCD), an enzyme responsible for malonyl-CoA (mCoA) degradation. Additionally, the SIRT1/LKB1/AMPK pathway (upstream activator of MLYCD) was promoted by PFT. Thus, induction of these two pathways by PFT diminished the hepatic mCoA content by enhancing MLYCD expression and function. Since mCoA inhibits carnitine palmitoyltransferase 1 (CPT1), the decrease of hepatic mCoA in the PFT-treated, HFD-fed mice increased CPT1 activity, favored fatty acid oxidation, and decreased steatosis. Additionally, we demonstrated that PFT abrogated steatosis and promoted MLYCD expression in palmitoleic acid-treated human HepaRG cells. CONCLUSIONS The p53 inhibitor PFT diminished hepatic triglyceride accumulation and lipotoxicity in mice fed a HFD, by depleting mCoA and favoring the β-oxidation of fatty acids.
Collapse
|
47
|
Ferreira MR, Camberos MDC, Selenscig D, Martucci LC, Chicco A, Lombardo YB, Cresto JC. Changes in hepatic lipogenic and oxidative enzymes and glucose homeostasis induced by an acetyl-l-carnitine and nicotinamide treatment in dyslipidaemic insulin-resistant rats. Clin Exp Pharmacol Physiol 2013; 40:205-11. [DOI: 10.1111/1440-1681.12050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 12/11/2012] [Accepted: 12/18/2012] [Indexed: 01/06/2023]
Affiliation(s)
- Maria R Ferreira
- Department of Biochemistry; School of Biochemistry; Litoral University; Santa Fe; Argentina
| | - Maria del C Camberos
- Endocrinology Research Centre (CEDIE); Ricardo Gutierrez Hospital; Buenos Aires; Argentina
| | - Dante Selenscig
- Department of Biochemistry; School of Biochemistry; Litoral University; Santa Fe; Argentina
| | - Lucía C Martucci
- Endocrinology Research Centre (CEDIE); Ricardo Gutierrez Hospital; Buenos Aires; Argentina
| | - Adriana Chicco
- Department of Biochemistry; School of Biochemistry; Litoral University; Santa Fe; Argentina
| | - Yolanda B Lombardo
- Department of Biochemistry; School of Biochemistry; Litoral University; Santa Fe; Argentina
| | - Juan C Cresto
- Endocrinology Research Centre (CEDIE); Ricardo Gutierrez Hospital; Buenos Aires; Argentina
| |
Collapse
|
48
|
Shaum KM, Polotsky AJ. Nutrition and reproduction: is there evidence to support a "Fertility Diet" to improve mitochondrial function? Maturitas 2013; 74:309-12. [PMID: 23376023 DOI: 10.1016/j.maturitas.2013.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 11/29/2022]
Abstract
Normal function of mitochondria plays an essential role in enabling reproductive capacity. To date, few studies have investigated the role of promoting mitochondrial health in relation to fertility in humans. Selected nutritional interventions have demonstrated a potential to enhance mitochondrial function, suggesting a promise for future research for fertility treatment. This review summarizes the extant literature and highlights a putative role of particular nutrients in promotion of mitochondrial function, including in vitro, animal and human studies. Strong basis exists to advocate for further investigation of nutritional treatments for infertility patients.
Collapse
|
49
|
Leong PK, Leung HY, Wong HS, Chen J, Ma CW, Yang Y, Ko KM. Long-Term Treatment with an Herbal Formula MCC Reduces the Weight Gain in High Fat Diet-Induced Obese Mice. Chin Med 2013. [DOI: 10.4236/cm.2013.43010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
50
|
Fat body dSir2 regulates muscle mitochondrial physiology and energy homeostasis nonautonomously and mimics the autonomous functions of dSir2 in muscles. Mol Cell Biol 2012; 33:252-64. [PMID: 23129806 DOI: 10.1128/mcb.00976-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Sir2 is an evolutionarily conserved NAD(+)-dependent deacetylase which has been shown to play a critical role in glucose and fat metabolism. In this study, we have perturbed Drosophila Sir2 (dSir2) expression, bidirectionally, in muscles and the fat body. We report that dSir2 plays a critical role in insulin signaling, glucose homeostasis, and mitochondrial functions. Importantly, we establish the nonautonomous functions of fat body dSir2 in regulating mitochondrial physiology and insulin signaling in muscles. We have identified a novel interplay between dSir2 and dFOXO at an organismal level, which involves Drosophila insulin-like peptide (dILP)-dependent insulin signaling. By genetic perturbations and metabolic rescue, we provide evidence to illustrate that fat body dSir2 mediates its effects on the muscles via free fatty acids (FFA) and dILPs (from the insulin-producing cells [IPCs]). In summary, we show that fat body dSir2 is a master regulator of organismal energy homeostasis and is required for maintaining the metabolic regulatory network across tissues.
Collapse
|