1
|
Zhong W, Chen C, Tan S, He X, Wen X, Wang S, Tocher DR, Waiho K, Chen C. Identification and Functional Characterization of the FATP1 Gene from Mud Crab, Scylla paramamosain. Animals (Basel) 2024; 14:2969. [PMID: 39457899 PMCID: PMC11506284 DOI: 10.3390/ani14202969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
In mammals, fatty acid transport protein 1 (FATP1) plays important roles in cellular uptake and activation of long-chain fatty acid (LCFA), especially in processes of transportation, oxidation and triacylglycerol synthesis. However, the role of FATP1 in invertebrates, especially decapod crustaceans, is still poorly understood. In this study, the cDNA of a FATP1 gene from a decapod crustacean, mud crab Scylla paramamosain, was cloned and functionally characterized. The FATP1 gene encoded a polypeptide consisting of 643 amino acids that exhibits all the typical features of the FATP family and shares high homology with the other FATP orthologs of crustaceans. The relative mRNA expression levels of FATP1 were observed to be higher in metabolically active tissues such as hepatopancreas, stomach and gill than in other crab parts. Knockdown of the FATP1 mRNA in vivo significantly reduced triacylglycerols and total lipid levels in the hepatopancreas, accompanied by an increase in the expression of genes related to fatty acid transportation, allocation and hydrolysis, including long-chain acyl-CoA synthetase 3/4 (ACSL3/4) and carnitine palmitoyl transferase 1 (CPT1), and a decrease in the expression of genes related to fatty acid synthesis such as acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) in the hepatopancreas. Furthermore, increased dietary n-3 long-chain polyunsaturated fatty acid (LC-PUFA) levels resulted in the up-regulation of the FATP1 expression in the hepatopancreas, accompanied by an increase in LC-PUFA content, especially eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), in both polar (PLs) and neutral lipids (NLs) in the hepatopancreas and muscles of crabs. These findings suggested that the FATP1 gene identified in S. paramamosain might play important roles in regulating long-chain fatty acid metabolism and deposition in crustaceans.
Collapse
Affiliation(s)
- Wenjie Zhong
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| | - Chuangsi Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| | - Senyue Tan
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| | - Xianda He
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| | - Xiaobo Wen
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China;
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| | - Douglas R. Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Khor Waiho
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, University Malaysia Terengganu, Kuala Terengganu 21300, Malaysia;
| | - Cuiying Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; (W.Z.); (C.C.); (S.T.); (X.H.); (S.W.); (D.R.T.)
| |
Collapse
|
2
|
Mishra JS, Zhao H, Zheng J, Kumar S. Sex-Specific Dysregulation of Placental Lipid Metabolism in Preeclampsia. OBSTETRICS AND GYNECOLOGY RESEARCH 2024; 7:49-58. [PMID: 39131546 PMCID: PMC11315440 DOI: 10.26502/ogr0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Preeclampsia (PE) is a hypertensive disorder of pregnancy associated with adverse maternal and fetal outcomes. While placental dysfunction is implicated in PE pathogenesis, the impact of PE on placental lipid metabolism and its potential sexual dimorphism remains poorly understood. Methods We conducted a comprehensive analysis of term placentas from PE and normotensive pregnancies with male and female fetuses. Lipid profiles were quantified using mass spectrometry, and mRNA expression of genes involved in fatty acid oxidation, esterification, and transport was assessed using qPCR. Results Placentas from PE pregnancies exhibited elevated lipid levels, with male placentas showing a more pronounced increase in triacylglycerols, cholesteryl esters, and free cholesterol compared to female placentas. Gene expression analysis revealed sexually dimorphic alterations, with male PE placentas exhibiting upregulation of genes involved in fatty acid uptake, oxidation, and esterification, while female PE placentas showed a more complex response with both upregulation and downregulation of certain genes. Notably, peroxisomal fatty acid oxidation was upregulated in male PE placentas but suppressed in female PE placentas. Conclusions Our findings reveal sexually dimorphic alterations in placental lipid metabolism in PE, suggesting that male placentas may be more vulnerable to lipotoxicity. These insights may have implications for understanding the pathogenesis of PE and developing sex-specific interventions to improve maternal and fetal outcomes.
Collapse
Affiliation(s)
- Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Hanjie Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jing Zheng
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
3
|
Nie T, Wang X, Li A, Shan A, Ma J. The promotion of fatty acid β-oxidation by hesperidin via activating SIRT1/PGC1α to improve NAFLD induced by a high-fat diet. Food Funct 2024; 15:372-386. [PMID: 38099440 DOI: 10.1039/d3fo04348g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Reducing fat deposits in hepatocytes is a direct treatment for nonalcoholic fatty liver disease (NAFLD) and the fatty acid metabolic processes mediated by fatty acid β-oxidation are important for the prevention of NAFLD. In this study, we established high-fat-diet models in vitro and in vivo to investigate the mechanism by which hesperidin (HDN) prevents NAFLD by modulating fatty acid β oxidation. Based on LC-MS screening of differential metabolites, many metabolites involved in phospholipid and lipid metabolism were found to be significantly altered and closely associated with fatty acid β-oxidation. The results from COIP experiments indicated that HDN increased the deacetylation of PGC1α by SIRT1. In addition, the results of CETSA and molecular docking experiments suggest that HDN targeting of SIRT1 plays an important role in their stable binding. Meanwhile, it was found that HDN reduced fatty acid uptake and synthesis and promoted the expression of SIRT1/PGC1α and fatty acid β-oxidation, and the latter process was inhibited after transfection to knockdown SIRT1. The results suggest that HDN improves NAFLD by promoting fatty acid β-oxidation through activating SIRT1/PGC1α. Thus, the findings indicate that HDN may be a potential drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Tong Nie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Aqun Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China.
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin, 150030, P.R. China
| |
Collapse
|
4
|
López de Las Hazas MC, Del Saz-Lara A, Cedó L, Crespo MC, Tomé-Carneiro J, Chapado LA, Macià A, Visioli F, Escola-Gil JC, Dávalos A. Hydroxytyrosol Induces Dyslipidemia in an ApoB100 Humanized Mouse Model. Mol Nutr Food Res 2024; 68:e2300508. [PMID: 37933702 DOI: 10.1002/mnfr.202300508] [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: 07/18/2023] [Revised: 09/25/2023] [Indexed: 11/08/2023]
Abstract
SCOPE Extra virgin olive oil has numerous cardiopreventive effects, largely due to its high content of (poly)phenols such as hydroxytyrosol (HT). However, some animal studies suggest that its excessive consumption may alter systemic lipoprotein metabolism. Because human lipoprotein metabolism differs from that of rodents, this study examines the effects of HT in a humanized mouse model that approximates human lipoprotein metabolism. METHODS AND RESULTS Mice are treated as follows: control diet or diet enriched with HT. Serum lipids and lipoproteins are determined after 4 and 8 weeks. We also analyzed the regulation of various genes and miRNA by HT, using microarrays and bioinformatic analysis. An increase in body weight is found after supplementation with HT, although food intake was similar in both groups. In addition, HT induced the accumulation of triacylglycerols but not cholesterol in different tissues. Systemic dyslipidemia after HT supplementation and impaired glucose metabolism are observed. Finally, HT modulates the expression of genes related to lipid metabolism, such as Pltp or Lpl. CONCLUSION HT supplementation induces systemic dyslipidemia and impaired glucose metabolism in humanized mice. Although the numerous health-promoting effects of HT far outweigh these potential adverse effects, further carefully conducted studies are needed.
Collapse
Affiliation(s)
- María-Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
| | - Andrea Del Saz-Lara
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
- Laboratory of Functional Foods, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, 16171, Spain
| | - Lídia Cedó
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, 08041, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona, 43005, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - María Carmen Crespo
- Laboratory of Functional Foods, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
| | - João Tomé-Carneiro
- Laboratory of Functional Foods, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
| | - Luis A Chapado
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
| | - Alba Macià
- Department of Food Technology, Engineering and Science, XaRTA-TPV, Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, University of Lleida, Lleida, 25198, Spain
| | - Francesco Visioli
- Laboratory of Functional Foods, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, Padova, 35121, Italy
| | - Joan C Escola-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, 08041, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Departament de Bioquímica, Biología Molecular i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Spain
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM+CSIC, Madrid, 28049, Spain
- Consorcio CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| |
Collapse
|
5
|
Nusir A, Sinclair P, Kabbani N. Mitochondrial Proteomes in Neural Cells: A Systematic Review. Biomolecules 2023; 13:1638. [PMID: 38002320 PMCID: PMC10669788 DOI: 10.3390/biom13111638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Mitochondria are ancient endosymbiotic double membrane organelles that support a wide range of eukaryotic cell functions through energy, metabolism, and cellular control. There are over 1000 known proteins that either reside within the mitochondria or are transiently associated with it. These mitochondrial proteins represent a functional subcellular protein network (mtProteome) that is encoded by mitochondrial and nuclear genomes and significantly varies between cell types and conditions. In neurons, the high metabolic demand and differential energy requirements at the synapses are met by specific modifications to the mtProteome, resulting in alterations in the expression and functional properties of the proteins involved in energy production and quality control, including fission and fusion. The composition of mtProteomes also impacts the localization of mitochondria in axons and dendrites with a growing number of neurodegenerative diseases associated with changes in mitochondrial proteins. This review summarizes the findings on the composition and properties of mtProteomes important for mitochondrial energy production, calcium and lipid signaling, and quality control in neural cells. We highlight strategies in mass spectrometry (MS) proteomic analysis of mtProteomes from cultured cells and tissue. The research into mtProteome composition and function provides opportunities in biomarker discovery and drug development for the treatment of metabolic and neurodegenerative disease.
Collapse
Affiliation(s)
- Aya Nusir
- Interdisciplinary Program in Neuroscience, School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| | - Patricia Sinclair
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| | - Nadine Kabbani
- Interdisciplinary Program in Neuroscience, School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| |
Collapse
|
6
|
Mićić B, Djordjevic A, Veličković N, Kovačević S, Martić T, Macut D, Vojnović Milutinović D. AMPK Activation as a Protective Mechanism to Restrain Oxidative Stress in the Insulin-Resistant State in Skeletal Muscle of Rat Model of PCOS Subjected to Postnatal Overfeeding. Biomedicines 2023; 11:1586. [PMID: 37371678 DOI: 10.3390/biomedicines11061586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/21/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy in women of reproductive age, often associated with obesity and insulin resistance. Childhood obesity is an important predisposing factor for the development of PCOS later in life. Being particularly interested in the interplay between prepubertal obesity and hyperandrogenemia, we investigated the effects of early postnatal overfeeding, accomplished by reducing litter size during the period of suckling, on energy sensing and insulin signaling pathways in the gastrocnemius muscle of a rat model of PCOS-induced by 5α-dihydrotestosterone (DHT). The combination of overfeeding and DHT treatment caused hyperinsulinemia and decreased systemic insulin sensitivity. Early postnatal overfeeding induced defects at critical nodes of the insulin signaling pathway in skeletal muscle, which was associated with reduced glucose uptake in the presence of hyperandrogenemia. In this setting, under a combination of overfeeding and DHT treatment, skeletal muscle switched to mitochondrial β-oxidation of fatty acids, resulting in oxidative stress and inflammation that stimulated AMP-activated protein kinase (AMPK) activity and its downstream targets involved in mitochondrial biogenesis and antioxidant protection. Overall, a combination of overfeeding and hyperandrogenemia resulted in a prooxidative and insulin-resistant state in skeletal muscle. This was accompanied by the activation of AMPK, which could represent a potential therapeutic target in insulin-resistant PCOS patients.
Collapse
Affiliation(s)
- Bojana Mićić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Teodora Martić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Djuro Macut
- Clinic for Endocrinology, Diabetes and Metabolic Diseases University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Doktora Subotića 13, 11000 Belgrade, Serbia
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of the Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| |
Collapse
|
7
|
Korbecki J, Kojder K, Jeżewski D, Simińska D, Tomasiak P, Tarnowski M, Chlubek D, Baranowska-Bosiacka I. Reduced Expression of Very-Long-Chain Acyl-CoA Synthetases SLC27A4 and SLC27A6 in the Glioblastoma Tumor Compared to the Peritumoral Area. Brain Sci 2023; 13:brainsci13050771. [PMID: 37239243 DOI: 10.3390/brainsci13050771] [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: 04/12/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
This study aimed to analyze solute carrier family 27 (SLC27) in glioblastoma tumors. The investigation of these proteins will provide insight into how and to what extent fatty acids are taken up from the blood in glioblastoma tumors, as well as the subsequent fate of the up-taken fatty acids. Tumor samples were collected from a total of 28 patients and analyzed using quantitative real-time polymerase chain reaction (qRT-PCR). The study also sought to explore the relationship between SLC27 expression and patient characteristics (age, height, weight, body mass index (BMI), and smoking history), as well as the expression levels of enzymes responsible for fatty acid synthesis. The expression of SLC27A4 and SLC27A6 was lower in glioblastoma tumors compared to the peritumoral area. Men had a lower expression of SLC27A5. Notably, a positive correlation was observed between the expression of SLC27A4, SLC27A5, and SLC27A6 and smoking history in women, whereas men exhibited a negative correlation between these SLC27s and BMI. The expression of SLC27A1 and SLC27A3 was positively correlated with the expression of ELOVL6. In comparison to healthy brain tissue, glioblastoma tumors take up fewer fatty acids. The metabolism of fatty acids in glioblastoma is dependent on factors such as obesity and smoking.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Neurosurgery and Pediatric Neurosurgery, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, Unii Lubelska 1, 71-252 Szczecin, Poland
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Tomasiak
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
8
|
Ma D, Wu T, Qu Y, Yang J, Cai L, Li X, Wang Y. Astragalus polysaccharide prevents heart failure-induced cachexia by alleviating excessive adipose expenditure in white and brown adipose tissue. Lipids Health Dis 2023; 22:9. [PMID: 36670439 PMCID: PMC9863193 DOI: 10.1186/s12944-022-01770-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Astragalus polysaccharide (APS) is a key active ingredient isolated from Astragalus membranaceus that has been reported to be a potential treatment for obesity and diabetes by regulating lipid metabolism and adipogenesis, alleviating inflammation, and improving insulin resistance. However, whether APS regulates lipid metabolism in the context of cachexia remains unclear. Therefore, this study analysed the effects of APS on lipid metabolism and adipose expenditure in a heart failure (HF)-induced cardiac cachexia rat model. METHODS: A salt-sensitive hypertension-induced cardiac cachexia rat model was used in the present study. Cardiac function was detected by echocardiography. The histological features and fat droplets in fat tissue and liver were observed by H&E staining and Oil O Red staining. Immunohistochemical staining, Western blotting and RT‒qPCR were used to detect markers of lipolysis and adipose browning in white adipose tissue (WAT) and thermogenesis in brown adipose tissue (BAT). Additionally, sympathetic nerve activity and inflammation in adipose tissue were detected. RESULTS Rats with HF exhibited decreased cardiac function and reduced adipose accumulation as well as adipocyte atrophy. In contrast, administration of APS not only improved cardiac function and increased adipose weight but also prevented adipose atrophy and FFA efflux in HF-induced cachexia. Moreover, APS inhibited HF-induced lipolysis and browning of white adipocytes since the expression levels of lipid droplet enzymes, including HSL and perilipin, and beige adipocyte markers, including UCP-1, Cd137 and Zic-1, were suppressed after administration of APS. In BAT, treatment with APS inhibited PKA-p38 MAPK signalling, and these effects were accompanied by decreased thermogenesis reflected by decreased expression of UCP-1, PPAR-γ and PGC-1α and reduced FFA β-oxidation in mitochondria reflected by decreased Cd36, Fatp-1 and Cpt1. Moreover, sympathetic nerve activity and interleukin-6 levels were abnormally elevated in HF rats, and astragalus polysaccharide could inhibit their activity. CONCLUSION APS prevented lipolysis and adipose browning in WAT and decreased BAT thermogenesis. These effects may be related to suppressed sympathetic activity and inflammation. This study provides a potential approach to treat HF-induced cardiac cachexia.
Collapse
Affiliation(s)
- Dufang Ma
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China
| | - Tao Wu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiwei Qu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinlong Yang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China
| | - Lu Cai
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China
| | - Xiao Li
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China.
| |
Collapse
|
9
|
Fatty acid metabolism in aggressive B-cell lymphoma is inhibited by tetraspanin CD37. Nat Commun 2022; 13:5371. [PMID: 36100608 PMCID: PMC9470561 DOI: 10.1038/s41467-022-33138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
The importance of fatty acid (FA) metabolism in cancer is well-established, yet the mechanisms underlying metabolic reprogramming remain elusive. Here, we identify tetraspanin CD37, a prognostic marker for aggressive B-cell lymphoma, as essential membrane-localized inhibitor of FA metabolism. Deletion of CD37 on lymphoma cells results in increased FA oxidation shown by functional assays and metabolomics. Furthermore, CD37-negative lymphomas selectively deplete palmitate from serum in mouse studies. Mechanistically, CD37 inhibits the FA transporter FATP1 through molecular interaction. Consequently, deletion of CD37 induces uptake and processing of exogenous palmitate into energy and essential building blocks for proliferation, and inhibition of FATP1 reverses this phenotype. Large lipid deposits and intracellular lipid droplets are observed in CD37-negative lymphoma tissues of patients. Moreover, inhibition of carnitine palmitoyl transferase 1 A significantly compromises viability and proliferation of CD37-deficient lymphomas. Collectively, our results identify CD37 as a direct gatekeeper of the FA metabolic switch in aggressive B-cell lymphoma. Tetraspanin CD37 deficiency has been reported as a prognostic marker for aggressive B-cell lymphoma. Here, the authors show that CD37 interacts with the fatty acid transporter 1 to inhibit palmitate uptake and its deficiency leads to increased fatty acid metabolism which promotes tumorigenesis in B-cell lymphoma.
Collapse
|
10
|
Huang J, Guo D, Zhu R, Feng Y, Li R, Yang X, Shi D. FATP1 Exerts Variable Effects on Adipogenic Differentiation and Proliferation in Cells Derived From Muscle and Adipose Tissue. Front Vet Sci 2022; 9:904879. [PMID: 35898540 PMCID: PMC9310014 DOI: 10.3389/fvets.2022.904879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
In livestock, intramuscular adipose tissue is highly valued whereas adipose tissue in other depots is considered as waste. Thus, genetic factors that favor fat deposition in intramuscular compartments over that in other adipose depots are highly desirable in meat-producing animals. Fatty acid transport 1 (FATP1) has been demonstrated to promote cellular fatty acid uptake and metabolism; however, whether it also influences cellular lipid accumulation remains unclear. In the present study, we investigated the effects of FATP1 on the differentiation and proliferation of adipocytes in five types of cells derived from muscle and adipose tissue and estimated the effects of FATP1 on intramuscular fat (IMF) deposition. We showed that FATP1 is mainly expressed in heart and muscle tissue in buffaloes as well as cells undergoing adipogenic differentiation. Importantly, we found that FATP1 promoted the adipogenic differentiation of muscle-derived cells (buffalo myocytes and intramuscular preadipocytes and mouse C2C12 cells) but did not affect, or even inhibited, that of adipose-derived cells (buffalo subcutaneous preadipocytes and mouse 3T3-L1 cells, respectively). Correspondingly, our results further indicated that FATP1 promotes IMF deposition in mice in vivo. Meanwhile, FATP1 was found to enhance the proliferative activity of all the assessed cells, except murine 3T3-L1 cells. These results provide new insights into the potential effects of FATP1 on IMF deposition, especially regarding its positive effects on meat quality in buffaloes and other livestock.
Collapse
|
11
|
Lopez-Tello J, Schofield Z, Kiu R, Dalby MJ, van Sinderen D, Le Gall G, Sferruzzi-Perri AN, Hall LJ. Maternal gut microbiota Bifidobacterium promotes placental morphogenesis, nutrient transport and fetal growth in mice. Cell Mol Life Sci 2022; 79:386. [PMID: 35760917 PMCID: PMC9236968 DOI: 10.1007/s00018-022-04379-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 12/22/2022]
Abstract
The gut microbiota plays a central role in regulating host metabolism. While substantial progress has been made in discerning how the microbiota influences host functions post birth and beyond, little is known about how key members of the maternal gut microbiota can influence feto-placental growth. Notably, in pregnant women, Bifidobacterium represents a key beneficial microbiota genus, with levels observed to increase across pregnancy. Here, using germ-free and specific-pathogen-free mice, we demonstrate that the bacterium Bifidobacterium breve UCC2003 modulates maternal body adaptations, placental structure and nutrient transporter capacity, with implications for fetal metabolism and growth. Maternal and placental metabolome were affected by maternal gut microbiota (i.e. acetate, formate and carnitine). Histological analysis of the placenta confirmed that Bifidobacterium modifies placental structure via changes in Igf2P0, Dlk1, Mapk1 and Mapk14 expression. Additionally, B. breve UCC2003, acting through Slc2a1 and Fatp1-4 transporters, was shown to restore fetal glycaemia and fetal growth in association with changes in the fetal hepatic transcriptome. Our work emphasizes the importance of the maternal gut microbiota on feto-placental development and sets a foundation for future research towards the use of probiotics during pregnancy.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Zoe Schofield
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Raymond Kiu
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Matthew J Dalby
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Gwénaëlle Le Gall
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Lindsay J Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK.
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
12
|
Metabolic regulation of follicular helper T cell differentiation in a mouse model of lupus. Immunol Lett 2022; 247:13-21. [PMID: 35568323 DOI: 10.1016/j.imlet.2022.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/08/2022] [Accepted: 03/28/2022] [Indexed: 01/24/2023]
Abstract
Follicular helper T (TFH) cells are expanded in systemic lupus erythematosus (SLE), where they are required for production of high affinity autoantibodies. A better understanding of the mechanisms that regulate the differentiation of TFH cells is critical. Naïve T cells from lupus-prone B6.NZM2410.Sle1.Sle2.Sle3 (TC) mice showed an intrinsic higher capacity to differentiate into TFH cells. Metabolic reprogramming is a vital regulatory mechanism for T cell differentiation, but how metabolic pathways contribute to TFH cell expansion in SLE remains elusive. Here we show that glycolysis, mTOR signaling, FAO, and the activity of complex V of the electron transport chain support TFH lineage commitment. Blocking complex I uniquely decreased the expansion of TFH cells from lupus-prone mice, and inhibition of some pathways had a greater effect in lupus-prone than control TFH cells. However, blocking glutaminolysis, complex III and ADP/ATP translocase did not affect TFH cell expansion. Together, our results identified novel intrinsic metabolic requirements for TFH cell differentiation, and further defined the differential metabolic pathways that support the expansion of TFH cells in lupus-prone mice. Together, our data indicates the crucial but distinct roles for metabolic pathways in TFH cell differentiation and provide a comprehensive experimental basis for fully understanding the precise roles of distant metabolic signaling in regulating the TFH cell differentiation.
Collapse
|
13
|
Zhu P, Hamlish NX, Thakkar AV, Steffeck AWT, Rendleman EJ, Khan NH, Waldeck NJ, DeVilbiss AW, Martin-Sandoval MS, Mathews TP, Chandel NS, Peek CB. BMAL1 drives muscle repair through control of hypoxic NAD + regeneration in satellite cells. Genes Dev 2022; 36:149-166. [PMID: 35115380 PMCID: PMC8887128 DOI: 10.1101/gad.349066.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/05/2022] [Indexed: 01/07/2023]
Abstract
The process of tissue regeneration occurs in a developmentally timed manner, yet the role of circadian timing is not understood. Here, we identify a role for the adult muscle stem cell (MuSC)-autonomous clock in the control of muscle regeneration following acute ischemic injury. We observed greater muscle repair capacity following injury during the active/wake period as compared with the inactive/rest period in mice, and loss of Bmal1 within MuSCs leads to impaired muscle regeneration. We demonstrate that Bmal1 loss in MuSCs leads to reduced activated MuSC number at day 3 postinjury, indicating a failure to properly expand the myogenic precursor pool. In cultured primary myoblasts, we observed that loss of Bmal1 impairs cell proliferation in hypoxia (a condition that occurs in the first 1-3 d following tissue injury in vivo), as well as subsequent myofiber differentiation. Loss of Bmal1 in both cultured myoblasts and in vivo activated MuSCs leads to reduced glycolysis and premature activation of prodifferentiation gene transcription and epigenetic remodeling. Finally, hypoxic cell proliferation and myofiber formation in Bmal1-deficient myoblasts are restored by increasing cytosolic NAD+ Together, we identify the MuSC clock as a pivotal regulator of oxygen-dependent myoblast cell fate and muscle repair through the control of the NAD+-driven response to injury.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Noah X Hamlish
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Adam W T Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Emily J Rendleman
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Nabiha H Khan
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Nathan J Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Andrew W DeVilbiss
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Misty S Martin-Sandoval
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Thomas P Mathews
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Navdeep S Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
14
|
Nan J, Lee JS, Lee SA, Lee DS, Park KS, Chung SS. An Essential Role of the N-Terminal Region of ACSL1 in Linking Free Fatty Acids to Mitochondrial β-Oxidation in C2C12 Myotubes. Mol Cells 2021; 44:637-646. [PMID: 34511469 PMCID: PMC8490201 DOI: 10.14348/molcells.2021.0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
Free fatty acids are converted to acyl-CoA by long-chain acyl-CoA synthetases (ACSLs) before entering into metabolic pathways for lipid biosynthesis or degradation. ACSL family members have highly conserved amino acid sequences except for their N-terminal regions. Several reports have shown that ACSL1, among the ACSLs, is located in mitochondria and mainly leads fatty acids to the β-oxidation pathway in various cell types. In this study, we investigated how ACSL1 was localized in mitochondria and whether ACSL1 overexpression affected fatty acid oxidation (FAO) rates in C2C12 myotubes. We generated an ACSL1 mutant in which the N-terminal 100 amino acids were deleted and compared its localization and function with those of the ACSL1 wild type. We found that ACSL1 adjoined the outer membrane of mitochondria through interaction of its N-terminal region with carnitine palmitoyltransferase-1b (CPT1b) in C2C12 myotubes. In addition, overexpressed ACSL1, but not the ACSL1 mutant, increased FAO, and ameliorated palmitate-induced insulin resistance in C2C12 myotubes. These results suggested that targeting of ACSL1 to mitochondria is essential in increasing FAO in myotubes, which can reduce insulin resistance in obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Jinyan Nan
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ji Seon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung-Ah Lee
- Genomic Medicine Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Soo Chung
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
15
|
Fruit of Gardenia jasminoides Induces Mitochondrial Activation and Non-Shivering Thermogenesis through Regulation of PPARγ. Antioxidants (Basel) 2021; 10:antiox10091418. [PMID: 34573050 PMCID: PMC8466082 DOI: 10.3390/antiox10091418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
The extract of the Gardenia jasminoides fruit (GJFE) can been consumed as an herbal tea or used as a yellow dye. Recently, studies report that GFJE exerts inhibitory effects on lipid accumulation and adipogenesis in white adipocytes. We evaluated the thermogenic actions of GJFE by focusing on mitochondrial activation and studying the underlying mechanisms. To investigate the role of GJFE on thermogenesis in mice, we used an acute cold exposure model. After 2 weeks of feeding, the cold tolerance of GJFE-fed mice was notably increased compared to PBS-fed mice. This was due to an increase in thermogenic proteins in the inguinal white adipose tissue of the cold-exposed mice. Moreover, GJFE significantly increased thermogenic factors such as peroxisome proliferator-activated receptor gamma (PPARγ), uncoupling protein 1 (UCP1), and PPARγ coactivator 1 alpha (PGC1α) in vitro as well. Factors related to mitochondrial abundance and functions were also induced by GJFE in white and beige adipocytes. However, the treatment of PPARγ inhibitor abolished the GJFE-induced changes, indicating that activation of PPARγ is critical for the thermogenic effect of GJFE. In conclusion, GJFE induces thermogenic action by activating mitochondrial function via PPARγ activation. Through these findings, we suggest GJFE as a potential anti-obesity agent with a novel mechanism involving thermogenic action in white adipocytes.
Collapse
|
16
|
Ye J, Haskey N, Dadlani H, Zubaidi H, Barnett JA, Ghosh S, Gibson DL. Deletion of mucin 2 induces colitis with concomitant metabolic abnormalities in mice. Am J Physiol Gastrointest Liver Physiol 2021; 320:G791-G803. [PMID: 33728986 DOI: 10.1152/ajpgi.00277.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Patients with inflammatory bowel disease (IBD) are at increased risk of under-recognized metabolic comorbidities. Chronic intestinal inflammation in IBD along with changes to the gut microbiome leads to broader systemic effects. Despite the existence of multiple animal models to study colitis, limited studies have examined the metabolic abnormalities associated with these models. In this study, a spontaneous model of colitis (mucin 2 knock-out mouse, Muc2-/-) was used to investigate the impact of intestinal disease on metabolic dysfunction. Before the onset of severe colitis, such as rectal prolapse, Muc2-/- mice exhibited impaired glucose clearance. Defects were noted in the insulin signaling pathway corresponding with upregulated genes in lipid utilization pathways, increased mitochondrial number, and peroxisome proliferator-activated coactivator 1α (PGC-1α), a transcription factor central to energy metabolism regulation. Parallel to these metabolic alterations, Muc2-/- mice exhibited systemic inflammation and bacteremia. We further characterized the dysbiotic microbiome's predicted functional categories given its contributing role to the colitic phenotype in the Muc2-/- mice. In addition to less butyrate levels, we show an increased predisposition to lipid metabolism and lipid biosynthesis pathways in the microbiome associated with the host's altered metabolic state. This study establishes the Muc2-/- mouse model that develops spontaneous colitis, as an ideal model for studying early comorbid metabolic dysfunction. Clarification of the underlying etiology of two phenotypes in this model could unravel important clues regarding the treatment of metabolic comorbidities during colitis.NEW & NOTEWORTHY This study discloses the impaired systemic energy metabolism in a classic colitis murine model (Muc2-/- knock-out model). Investigating the interaction between colitis and metabolic disorders helps to extend our knowledge on deciphering inflammatory bowel disease-associated comorbidities and provides new insight into clinical treatment.
Collapse
Affiliation(s)
- Jiayu Ye
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Natasha Haskey
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Hansika Dadlani
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Hatem Zubaidi
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Jacqueline A Barnett
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Sanjoy Ghosh
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| | - Deanna L Gibson
- Department of Biology, University of British Columbia Okanagan , Kelowna, British Columbia, Canada.,Department of Medicine, University of British Columbia Okanagan , Kelowna, British Columbia, Canada
| |
Collapse
|
17
|
Zhong Y, Yan Z, Song B, Zheng C, Duan Y, Kong X, Deng J, Li F. Dietary supplementation with betaine or glycine improves the carcass trait, meat quality and lipid metabolism of finishing mini-pigs. ACTA ACUST UNITED AC 2021; 7:376-383. [PMID: 34258425 PMCID: PMC8245815 DOI: 10.1016/j.aninu.2020.08.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/19/2020] [Accepted: 08/14/2020] [Indexed: 10/25/2022]
Abstract
The objective of the study is to evaluate and compare the effects of betaine or glycine on carcass trait, meat quality and lipid metabolism of finishing Huan Jiang mini-pigs. Betaine called trimethylglycine is a methyl derivative of glycine, but few researches were conducted to compare the impact of dietary betaine and glycine on pigs. One hundred and forty-four Huan Jiang mini-pigs (body weight = 10.55 ± 0.15 kg; 70 d) were randomly divided to 3 treatment groups (basal diet, glycine or betaine). Results indicated that dietary betaine increased the average daily gain (ADG) and final weight (P < 0.05). Dietary glycine or betaine markedly reduced average backfat thickness (P < 0.05) and heightened lean percentage (P < 0.01) compared to the control group. Moreover, in comparison with the control group, betaine significantly improved the redness (a∗) and tenderness (shear force) of the longissimus dorsi (LD) muscle (P < 0.05), whereas glycine only raised the value of a∗ of the LD muscle (P < 0.05). These results showed that diet supplemented with 0.25% betaine and equimolar amounts of glycine could regulate cascass trait and meat quality of finishing Huan Jiang mini-pigs, and the effect of betaine was superior to that of glycine.
Collapse
Affiliation(s)
- Yinzhao Zhong
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China.,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - Zhaoming Yan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Bo Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China
| | - Xiangfeng Kong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China
| | - JinPing Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China
| |
Collapse
|
18
|
The role of FATP1 in lipid accumulation: a review. Mol Cell Biochem 2021; 476:1897-1903. [PMID: 33486652 DOI: 10.1007/s11010-021-04057-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
Lipid accumulation in mammals has been widely studied for decades due to its significant association with obesity in humans and meat quality in livestock animals. Fatty acid transport 1 (FATP1) is an evolutionarily conserved protein that localizes to the plasma membrane to enhance the transportation of fatty acids (FAs). In line with this function, FATP1 is involved in the metabolism of FAs, including their esterification and oxidation. In addition, the expression of FATP1 can be regulated by several energy-related factors, such as insulin and PPAR activators and transcription factors. These events connect FATP1 with cellular lipid accumulation. Recently, several studies have suggested that FATP1 acts as a facilitator in cellular lipid accumulation, whereas others hold a contrary view. Here, we will review these data and probe the possibility that FATP1 acts as a regulator in lipid accumulation, which will provide effective information for studies on the relationship between FATP1 and obesity in humans and meat quality in livestock animals.
Collapse
|
19
|
SOCS2 Inhibits Mitochondrial Fatty Acid Oxidation via Suppressing LepR/JAK2/AMPK Signaling Pathway in Mouse Adipocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3742542. [PMID: 32733634 PMCID: PMC7376435 DOI: 10.1155/2020/3742542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022]
Abstract
Suppressor of cytokine signaling 2 (SOCS2) plays an important role in fat deposition, skeletal muscle, central nervous system development, and mitochondria biogenesis. Nevertheless, the regulatory mechanisms of SOCS2 on mitochondrial fatty acid oxidation (FAO) remain unclear. Leptin could inhibit food intake and increase thermogenesis through leptin receptor (LepR), which was present in the hypothalamus and certain peripheral organs, including adipose tissue. With strong interest, we focused on the connection between leptin and SOCS2 and their effect on FAO in adipocytes. In our study, we found that the mRNA level of SOCS2 and the protein levels of PGC-1α, CPT-1b, FAT, and p-ACC were elevated by leptin in the inguinal adipose tissue of mice. On the contrary, the protein levels of FABP4, FATP1, and FAS were declined. The genes related to fatty acid oxidation such as PGC-1α, NRF-1, TFAM, CPT-1b, AOX1, COX2, and UCP2 were attenuated by SOCS2, but elevated by leptin. Moreover, fatty acid oxidation enzyme MCAD, LCAD, and Cyt C levels were reduced in response to SOCS2. These reductions correspond well with the reduced release of free fatty acid and the reduction of mitochondrial complexes I and III by SOCS2. Furthermore, JAK2/AMPK pathway-specific inhibitors could block the mitochondrial FAO; hence, this pathway was implied to have a potential impact on FAO. Together, these studies suggested that SOCS2 had a negative effect on mitochondrial fatty acid oxidation, and the LepR/JAK2/AMPK pathway played a crucial role in this process.
Collapse
|
20
|
The Effect of a 2-Week Red Ginseng Supplementation on Food Efficiency and Energy Metabolism in Mice. Nutrients 2020; 12:nu12061726. [PMID: 32526977 PMCID: PMC7352690 DOI: 10.3390/nu12061726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/28/2020] [Accepted: 06/05/2020] [Indexed: 12/31/2022] Open
Abstract
Red ginseng (RG) ingestion reportedly affects body weight, food intake, and fat accumulation reduction. It also induces changes in energy metabolism regulation and glycemic control. Previously, 2-week RG ingestion with endurance training was found to enhance fat oxidation during exercise. However, such effects on energy metabolism and the expression of mRNAs related to energy substrate utilization in resting mice (untrained mice) are still unclear. Here, we determined the effect of RG on energy metabolism and substrate utilization in untrained male mice. Twenty-four mice were separated into an RG group that received a daily dosage of 1 g/kg RG for 2 weeks, and a control (CON). Energy expenditure, blood and tissue glycogen levels, and expression of mRNAs related to energy substrate utilization in muscles were measured before and 2 weeks after treatment. Total food intake was significantly lower in the RG than in the CON group (p < 0.05), but final body weights did not differ. Carbohydrate and fat oxidation over 24 h did not change in either group. There were no significant differences in gastrocnemius GLUT4, MCT1, MCT4, FAT/CD36, and CPT1b mRNA levels between groups. Thus, the effects of RG ingested during rest differ from the effects of RG ingestion in combination with endurance exercise; administering RG to untrained mice for 2 weeks did not change body weight and energy metabolism. Therefore, future studies should consider examining the RG ingestion period and dosage for body weight control and improving energy metabolism.
Collapse
|
21
|
Zhang W, Bai Y, Chen Z, Li X, Fu S, Huang L, Lin S, Du H. Comprehensive analysis of long non-coding RNAs and mRNAs in skeletal muscle of diabetic Goto-Kakizaki rats during the early stage of type 2 diabetes. PeerJ 2020; 8:e8548. [PMID: 32095365 PMCID: PMC7023842 DOI: 10.7717/peerj.8548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/12/2020] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle long non-coding RNAs (lncRNAs) were reported to be involved in the development of type 2 diabetes (T2D). However, little is known about the mechanism of skeletal muscle lncRNAs on hyperglycemia of diabetic Goto-Kakizaki (GK) rats at the age of 3 and 4 weeks. To elucidate this, we used RNA-sequencing to profile the skeletal muscle transcriptomes including lncRNAs and mRNAs, in diabetic GK and control Wistar rats at the age of 3 and 4 weeks. In total, there were 438 differentially expressed mRNAs (DEGs) and 401 differentially expressed lncRNAs (DELs) in skeletal muscle of 3-week-old GK rats compared with age-matched Wistar rats, and 1000 DEGs and 726 DELs between GK rats and Wistar rats at 4 weeks of age. The protein–protein interaction analysis of overlapping DEGs between 3 and 4 weeks, the correlation analysis of DELs and DEGs, as well as the prediction of target DEGs of DELs showed that these DEGs (Pdk4, Stc2, Il15, Fbxw7 and Ucp3) might play key roles in hyperglycemia, glucose intolerance, and increased fatty acid oxidation. Considering the corresponding co-expressed DELs with high correlation coefficients or targeted DELs of these DEGs, our study indicated that these dysregulated lncRNA-mRNA pairs (NONRATG017315.2-Pdk4, NONRATG003318.2-Stc2, NONRATG011882.2-Il15, NONRATG013497.2-Fbxw7, MSTRG.1662-Ucp3) might be related to above biological processes in GK rats at the age of 3 and 4 weeks. Our study could provide more comprehensive knowledge of mRNAs and lncRNAs in skeletal muscle of GK rats at 3 and 4 weeks of age. And our study may provide deeper understanding of the underlying mechanism in T2D of GK rats at the age of 3 and 4 weeks.
Collapse
Affiliation(s)
- Wenlu Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yunmeng Bai
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xingsong Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shudai Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
22
|
Cui H, Xie N, Banerjee S, Ge J, Guo S, Liu G. Impairment of Fatty Acid Oxidation in Alveolar Epithelial Cells Mediates Acute Lung Injury. Am J Respir Cell Mol Biol 2019; 60:167-178. [PMID: 30183330 DOI: 10.1165/rcmb.2018-0152oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Profound impairment in cellular oxygen consumption, referred to as cytopathic dysoxia, is one of the pathological hallmarks in the lungs of patients with pathogen-induced acute lung injury (ALI). However, the underlying mechanism for this functional defect remains largely unexplored. In this study, we found that primary mouse alveolar epithelial cells (AECs) conducted robust fatty acid oxidation (FAO). More importantly, FAO was strikingly impaired in AECs of mice with LPS-induced ALI. The metabolic deficiency in these cells was likely due to decreased expression of key mediators involved in FAO and mitochondrial bioenergenesis, such as peroxisome proliferator-activated receptor γ coactivator (PGC)-1α, carnitine palmitoyltransferase 1A, and medium-chain acyl-CoA dehydrogenase (CAD). We found that treatment of alveolar epithelial line MLE-12 cells with BAL fluids from mice with ALI decreased FAO, and this effect was largely replicated in MLE-12 cells treated with the proinflammatory cytokine TNF-α, which was consistent with downregulations of PGC-1α, carnitine palmitoyltransferase 1A, long-chain CAD, and medium-chain CAD in the same treated cells. Furthermore, we found that the BAL fluids from ALI mice and TNF-α inhibited MLE-12 bioenergenesis and promoted cell apoptosis. In delineation of the role of FAO in ALI in vivo, we found that conditional ablation of AEC PGC-1α aggravated LPS-induced ALI. In contrast, fenofibrate, an activator of the PPAR-α/PGC-1α cascade, protected mice from this pathology. In summary, these data suggest that FAO is essential to AEC bioenergenesis and functional homeostasis. This study also indicates that FAO impairment-induced AEC dysfunction is an important contributing factor to the pathogenesis of ALI.
Collapse
Affiliation(s)
- Huachun Cui
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Na Xie
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sami Banerjee
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jing Ge
- 2 Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Sijia Guo
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,3 Department of Pulmonary, Allergy, and Critical Care Medicine, the Second Affiliated Hospital, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Gang Liu
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
23
|
Nyunt T, Britton M, Wanichthanarak K, Budamagunta M, Voss JC, Wilson DW, Rutledge JC, Aung HH. Mitochondrial oxidative stress-induced transcript variants of ATF3 mediate lipotoxic brain microvascular injury. Free Radic Biol Med 2019; 143:25-46. [PMID: 31356870 PMCID: PMC6848793 DOI: 10.1016/j.freeradbiomed.2019.07.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/30/2019] [Accepted: 07/23/2019] [Indexed: 12/06/2022]
Abstract
Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.
Collapse
Affiliation(s)
- Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Monica Britton
- Genome Center and Bioinformatics Core Facility, University of California, Davis, CA, 95616, USA
| | - Kwanjeera Wanichthanarak
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA, USA; Department of Biochemistry and Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Hnin H Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
24
|
|
25
|
Intake of fruit and leaves of sweet cherry beneficially affects lipid metabolism, oxidative stress and inflammation in Wistar rats fed with high fat-cholesterol diet. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.03.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
26
|
Dobrinskikh E, Al-Juboori SI, Shabeka U, Reisz JA, Zheng C, Marwan AI. Heterogeneous Pulmonary Response After Tracheal Occlusion: Clues to Fetal Lung Growth. J Surg Res 2019; 239:242-252. [PMID: 30856517 DOI: 10.1016/j.jss.2019.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/15/2019] [Accepted: 02/06/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Understanding inconsistent clinical outcomes in infants with severe congenital diaphragmatic hernia (CDH) after tracheal occlusion (TO) is a crucial step for advancing neonatal care. The objective of this study is to explore the heterogeneous airspace morphometry and the metabolic landscape changes in fetal lungs after TO. METHODS Fetal lungs on days 1 and 4 after TO were examined using mass spectrometry-based metabolomics, fluorescence lifetime imaging microscopy (FLIM), the number of airspaces, and tissue-to-airspace ratio (TAR). RESULTS Two morphometric areas were identified in TO lungs compared with controls (more small airspaces at day 1 and a higher number of enlarged airspaces at day 4). Global metabolomics analysis revealed a significant upregulation of glycolysis and a suppression of the tricarboxylic acid cycle in day 4 TO lungs compared with day 1 TO lungs. In addition, there was a significant increase in polyamines involved in cell growth and proliferation. Locally, FLIM analysis on day 1 TO lungs demonstrated two types of heterogeneous zones-similar to control and with increased oxidative phosphorylation. FLIM on day 4 TO lungs demonstrated appearance of zones with enlarged airspaces and a metabolic shift toward glycolysis, accompanied by a decrease in the FLIM "lipid-surfactant" signal. CONCLUSIONS In normal fetal lungs, we report a novel temporal pattern of varied morphometric and metabolic changes. Initially, there is formation of zones with small airspaces, followed by airspace enlargement over time. Metabolically day 1 TO lungs have zones with increased oxidative phosphorylation, whereas day 4 TO lungs have a shift toward glycolysis in the enlarged airspaces. Based on our observations, we speculate that the "best responders" to tracheal occlusion should have bigger lungs with small airspaces and normal surfactant production.
Collapse
Affiliation(s)
- Evgenia Dobrinskikh
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Saif I Al-Juboori
- Division of Pediatric Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Uladzimir Shabeka
- Division of Pediatric Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Connie Zheng
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Ahmed I Marwan
- Division of Pediatric Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
27
|
Zhong Y, Song B, Zheng C, Li F, Kong X, Duan Y, Deng J. α-Ketoisocaproate and β-hydroxy-β-methyl butyrate regulate fatty acid composition and lipid metabolism in skeletal muscle of growing pigs. J Anim Physiol Anim Nutr (Berl) 2019; 103:846-857. [PMID: 30775808 DOI: 10.1111/jpn.13077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/05/2019] [Accepted: 01/25/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES This study aims to investigate the effects and roles of excess leucine (Leu) versus its metabolites α-ketoisocaproate (KIC) and β-hydroxy-β-methyl butyrate (HMB) on fatty acid composition and lipid metabolism in skeletal muscle of growing pigs. METHODS AND RESULTS Thirty-two pigs with a similar initial weight (9.55 ± 0.19 kg) were fed one of the four diets (basal diet, L-Leu, KIC-Ca and HMB-Ca) for 45 days. Results indicated that dietary treatments did not affect the intramuscular fat (IMF) content (p > 0.05), but differently influenced the fatty acid composition of longissimus dorsi muscle (LM) and soleus muscle (SM). In particular, the proportion of N3 PUFA specifically in LM was significantly decreased in the Leu group and increased in both KIC and HMB group relative to the basal diet group (p < 0.05). Furthermore, pigs fed KIC-supplemented diets exhibited decreased expression of FATP-1, ACC, ATGL, C/EBPα, PPARγ and SREBP-1c in LM and increased expression of FATP-1, FAT/CD36, ATGL and M-CPT-1 in SM relative to the basal diet control (p < 0.05). CONCLUSIONS These findings indicated that doubling dietary Leu content decreased the percentage of N3 PUFA mainly in glycolytic skeletal muscle, whereas KIC and HMB improved muscular fatty acid composition and altered lipid metabolism in skeletal muscle of growing pigs. The mechanism of action of KIC might be related to the TFs, and the mechanism of action of HMB might be associated with the AMPK-mTOR signalling pathway.
Collapse
Affiliation(s)
- Yinzhao Zhong
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Bo Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Fengna Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China.,Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, China
| | - Xiangfeng Kong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yehui Duan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
28
|
Singh BK, Sinha RA, Tripathi M, Mendoza A, Ohba K, Sy JAC, Xie SY, Zhou J, Ho JP, Chang CY, Wu Y, Giguère V, Bay BH, Vanacker JM, Ghosh S, Gauthier K, Hollenberg AN, McDonnell DP, Yen PM. Thyroid hormone receptor and ERRα coordinately regulate mitochondrial fission, mitophagy, biogenesis, and function. Sci Signal 2018; 11:eaam5855. [PMID: 29945885 DOI: 10.1126/scisignal.aam5855] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Thyroid hormone receptor β1 (THRB1) and estrogen-related receptor α (ESRRA; also known as ERRα) both play important roles in mitochondrial activity. To understand their potential interactions, we performed transcriptome and ChIP-seq analyses and found that many genes that were co-regulated by both THRB1 and ESRRA were involved in mitochondrial metabolic pathways. These included oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, and β-oxidation of fatty acids. TH increased ESRRA expression and activity in a THRB1-dependent manner through the induction of the transcriptional coactivator PPARGC1A (also known as PGC1α). Moreover, TH induced mitochondrial biogenesis, fission, and mitophagy in an ESRRA-dependent manner. TH also induced the expression of the autophagy-regulating kinase ULK1 through ESRRA, which then promoted DRP1-mediated mitochondrial fission. In addition, ULK1 activated the docking receptor protein FUNDC1 and its interaction with the autophagosomal protein MAP1LC3B-II to induce mitophagy. siRNA knockdown of ESRRA, ULK1, DRP1, or FUNDC1 inhibited TH-induced autophagic clearance of mitochondria through mitophagy and decreased OXPHOS. These findings show that many of the mitochondrial actions of TH are mediated through stimulation of ESRRA expression and activity, and co-regulation of mitochondrial turnover through the PPARGC1A-ESRRA-ULK1 pathway is mediated by their regulation of mitochondrial fission and mitophagy. Hormonal or pharmacologic induction of ESRRA expression or activity could improve mitochondrial quality in metabolic disorders.
Collapse
Affiliation(s)
- Brijesh K Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| | - Rohit A Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Arturo Mendoza
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Kenji Ohba
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Jann A C Sy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sherwin Y Xie
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jia Pei Ho
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Sujoy Ghosh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Donald P McDonnell
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
29
|
Li S, Wang H, Wang X, Wang Y, Feng J. Betaine affects muscle lipid metabolism via regulating the fatty acid uptake and oxidation in finishing pig. J Anim Sci Biotechnol 2017; 8:72. [PMID: 28883917 PMCID: PMC5580292 DOI: 10.1186/s40104-017-0200-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Betaine affects fat metabolism in animals, but the specific mechanism is still not clear. The purpose of this study was to investigate possible mechanisms of betaine in altering lipid metabolism in muscle tissue in finishing pigs. METHODS A total of 120 crossbred gilts (Landrace × Yorkshire × Duroc) with an average initial body weight of 70.1 kg were randomly allotted to three dietary treatments. The treatments included a corn-soybean meal basal diet supplemented with 0, 1250 or 2500 mg/kg betaine. The feeding experiment lasted 42 d. RESULTS Betaine addition to the diet significantly increased the concentration of free fatty acids (FFA) in muscle (P < 0.05). Furthermore, the levels of serum cholesterol and high-density lipoprotein cholesterol were decreased (P < 0.05) and total cholesterol content was increased in muscle (P < 0.05) of betaine fed pigs. Experiments on genes involved in fatty acid transport showed that betaine increased expression of lipoprotein lipase(LPL), fatty acid translocase/cluster of differentiation (FAT/CD36), fatty acid binding protein (FABP3) and fatty acid transport protein (FATP1) (P < 0.05). The abundance of fatty acid transport protein and fatty acid binding protein were also increased by betaine (P < 0.05). As for the key factors involved in fatty acid oxidation, although betaine supplementation didn't affect the level of carnitine and malonyl-CoA, betaine increased mRNA and protein abundance of carnitine palmitransferase-1(CPT1) and phosphorylated-AMPK (P < 0.05). CONCLUSIONS The results suggested that betaine may promoted muscle fatty acid uptake via up-regulating the genes related to fatty acid transporter including FAT/CD36, FATP1 and FABP3. On the other hand, betaine activated AMPK and up-regulated genes related to fatty acid oxidation including PPARα and CPT1. The underlying mechanism regulating fatty acid metabolism in pigs supplemented with betaine is associated with the up-regulation of genes involved in fatty acid transport and fatty acid oxidation.
Collapse
Affiliation(s)
- Sisi Li
- Key Laboratory of Animal Nutrition & Feed, Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Haichao Wang
- Key Laboratory of Animal Nutrition & Feed, Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Xinxia Wang
- Key Laboratory of Animal Nutrition & Feed, Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition & Feed, Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Jie Feng
- Key Laboratory of Animal Nutrition & Feed, Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
30
|
Calabuig-Navarro V, Haghiac M, Minium J, Glazebrook P, Ranasinghe GC, Hoppel C, Hauguel de-Mouzon S, Catalano P, O’Tierney-Ginn P. Effect of Maternal Obesity on Placental Lipid Metabolism. Endocrinology 2017; 158:2543-2555. [PMID: 28541534 PMCID: PMC5551552 DOI: 10.1210/en.2017-00152] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/17/2017] [Indexed: 02/06/2023]
Abstract
Obese women, on average, give birth to babies with high fat mass. Placental lipid metabolism alters fetal lipid delivery, potentially moderating neonatal adiposity, yet how it is affected by maternal obesity is poorly understood. We hypothesized that fatty acid (FA) accumulation (esterification) is higher and FA β-oxidation (FAO) is lower in placentas from obese, compared with lean women. We assessed acylcarnitine profiles (lipid oxidation intermediates) in mother-baby-placenta triads, in addition to lipid content, and messenger RNA (mRNA)/protein expression of key regulators of FA metabolism pathways in placentas of lean and obese women with normal glucose tolerance recruited at scheduled term Cesarean delivery. In isolated trophoblasts, we measured [3H]-palmitate metabolism. Placentas of obese women had 17.5% (95% confidence interval: 6.1, 28.7%) more lipid than placentas of lean women, and higher mRNA and protein expression of FA esterification regulators (e.g., peroxisome proliferator-activated receptor γ, acetyl-CoA carboxylase, steroyl-CoA desaturase 1, and diacylglycerol O-acyltransferase-1). [3H]-palmitate esterification rates were increased in trophoblasts from obese compared with lean women. Placentas of obese women had fewer mitochondria and a lower concentration of acylcarnitines, suggesting a decrease in mitochondrial FAO capacity. Conversely, peroxisomal FAO was greater in placentas of obese women. Altogether, these changes in placental lipid metabolism may serve to limit the amount of maternal lipid transferred to the fetus, restraining excess fetal adiposity in this population of glucose-tolerant women.
Collapse
Affiliation(s)
- Virtu Calabuig-Navarro
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
- Center for Reproductive Biology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Maricela Haghiac
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
| | - Judi Minium
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
| | - Patricia Glazebrook
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
| | | | - Charles Hoppel
- Center for Mitochondrial Diseases, Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | | | - Patrick Catalano
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
- Center for Reproductive Biology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Perrie O’Tierney-Ginn
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, Ohio 44109
- Center for Reproductive Biology, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
31
|
Improvement of mitochondrial function by celastrol in palmitate-treated C2C12 myotubes via activation of PI3K-Akt signaling pathway. Biomed Pharmacother 2017; 93:903-912. [PMID: 28715871 DOI: 10.1016/j.biopha.2017.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022] Open
Abstract
Compelling evidences posited that high level of saturated fatty acid gives rise to mitochondrial dysfunction and inflammation in the development of insulin resistance in skeletal muscle. Celastrol is a pentacyclic triterpenoid derived from the root extracts of Tripterygium wilfordii that possesses potent anti-inflammatory properties in a number of animal models with metabolic diseases. However, the cellular mechanistic action of celastrol in alleviating obesity-induced insulin resistance in skeletal muscle remains largely unknown. Therefore, the present investigation evaluated the attributive properties of celastrol at different concentrations (10, 20, 30 and 40nM) on insulin resistance in C2C12 myotubes evoked by palmitate. We demonstrated that celastrol improved mitochondrial functions through significant enhancement of intracellular ATP content, mitochondrial membrane potential, citrate synthase activity and decrease of mitochondrial superoxide productions. Meanwhile, augmented mitochondrial DNA (mtDNA) content with suppressed DNA oxidative damage were observed following celastrol treatment. Celastrol significantly enhanced fatty acid oxidation rate and increased the level of tricarboxylic acid (TCA) cycle intermediates in palmitate-treated cells. Further analysis revealed that the improvement of glucose uptake activity in palmitate-loaded myotubes was partly mediated by celastrol via activation of PI3K-Akt insulin signaling pathway. Collectively, these findings provided evidence for the first time that the protection from palmitate-mediated insulin resistance in C2C12 myotubes by celastrol is likely associated with the improvement of mitochondrial functions-related metabolic activities.
Collapse
|
32
|
Qiu F, Xie L, Ma JE, Luo W, Zhang L, Chao Z, Chen S, Nie Q, Lin Z, Zhang X. Lower Expression of SLC27A1 Enhances Intramuscular Fat Deposition in Chicken via Down-Regulated Fatty Acid Oxidation Mediated by CPT1A. Front Physiol 2017; 8:449. [PMID: 28706492 PMCID: PMC5489693 DOI: 10.3389/fphys.2017.00449] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/14/2017] [Indexed: 01/11/2023] Open
Abstract
Intramuscular fat (IMF) is recognized as the predominant factor affecting meat quality due to its positive correlation with tenderness, juiciness, and flavor. Chicken IMF deposition depends on the balance among lipid synthesis, transport, uptake, and subsequent metabolism, involving a lot of genes and pathways, however, its precise molecular mechanisms remain poorly understood. In the present study, the breast muscle tissue of female Wenchang chickens (WC) (higher IMF content, 1.24 in D120 and 1.62 in D180) and female White Recessive Rock chickens (WRR; lower IMF content, 0.53 in D120 and 0.90 in D180) were subjected to RNA-sequencing (RNA-seq) analysis. Results showed that many genes related to lipid catabolism, such as SLC27A1, LPL, ABCA1, and CPT1A were down-regulated in WC chickens, and these genes were involved in the PPAR signaling pathway and formed an IPA® network related to lipid metabolism. Furthermore, SLC27A1 was more down-regulated in WRR.D180.B than in WRR.D120.B. Decreased cellular triglyceride (TG) and up-regulated CPT1A were observed in the SLC27A1 overexpression QM-7 cells, and increased cellular triglyceride (TG) and down-regulated CPT1A were observed in the SLC27A1 knockdown QM-7 cells. These results suggest that lower lipid catabolism exists in WC chickens but not in WRR chickens, and lower expression of SLC27A1 facilitate IMF deposition in chicken via down-regulated fatty acid oxidation mediated by CPT1A. These findings indicate that reduced lipid catabolism, rather than increased lipid anabolism, contributes to chicken IMF deposition.
Collapse
Affiliation(s)
- Fengfang Qiu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China.,School of Chemistry, Biology and Material Science, East China University of TechnologyNanchang, China
| | - Liang Xie
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Jing-E Ma
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Wen Luo
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Li Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Zhe Chao
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Shaohao Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Qinghua Nie
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Zhemin Lin
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Xiquan Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| |
Collapse
|
33
|
Zhao YD, Yin L, Archer S, Lu C, Zhao G, Yao Y, Wu L, Hsin M, Waddell TK, Keshavjee S, Granton J, de Perrot M. Metabolic heterogeneity of idiopathic pulmonary fibrosis: a metabolomic study. BMJ Open Respir Res 2017; 4:e000183. [PMID: 28883924 PMCID: PMC5531310 DOI: 10.1136/bmjresp-2017-000183] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 11/28/2022] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal disease of unknown cause characterised by progressive fibrotic formation in lung tissue. We hypothesise that disrupted metabolic pathways in IPF contribute to disease pathogenesis. Methods Metabolomics of human IPF was performed using mass spectroscopy (IPF lung=8; donor lung=8). Gene expression of key metabolic enzymes was measured using microarrays. Of the 108 metabolites whose levels were found altered, 48 were significantly increased, whereas 60 were significantly decreased in IPF samples compared with normal controls. Results Specific metabolic pathways mediating the IPF remodelling were found with a downregulated sphingolipid metabolic pathway but an upregulated arginine pathway in IPF. In addition, disrupted glycolysis, mitochondrial beta-oxidation and tricarboxylic acid cycle, altered bile acid, haem and glutamate/aspartate metabolism were found in IPF samples compared with control. Conclusions Our results show alterations in metabolic pathways for energy consumption during lung structural remodelling, which may contribute to IPF pathogenesis. We believe that this is the first report of simultaneously and systemically measuring changes of metabolites involving nine metabolic pathways in human severe IPF lungs. The measurement of the metabolites may serve in the future diagnosis and prognosis of IPF.
Collapse
Affiliation(s)
- Yidan D Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Li Yin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Stephen Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Catherine Lu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - George Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Yan Yao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Licun Wu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Michael Hsin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Thomas K Waddell
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - John Granton
- Division of Respirology, University Health Network, Toronto, Ontario, Canada
| | - Marc de Perrot
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Ochiai Y, Uchida Y, Ohtsuki S, Tachikawa M, Aizawa S, Terasaki T. The blood-brain barrier fatty acid transport protein 1 (FATP1/SLC27A1) supplies docosahexaenoic acid to the brain, and insulin facilitates transport. J Neurochem 2017; 141:400-412. [PMID: 28035674 DOI: 10.1111/jnc.13943] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 12/14/2016] [Accepted: 12/14/2016] [Indexed: 11/28/2022]
Abstract
We purposed to clarify the contribution of fatty acid transport protein 1 (FATP1/SLC 27A1) to the supply of docosahexaenoic acid (DHA) to the brain across the blood-brain barrier in this study. Transport experiments showed that the uptake rate of [14 C]-DHA in human FATP1-expressing HEK293 cells was significantly greater than that in empty vector-transfected (mock) HEK293 cells. The steady-state intracellular DHA concentration was nearly 2-fold smaller in FATP1-expressing than in mock cells, suggesting that FATP1 works as not only an influx, but also an efflux transporter for DHA. [14 C]-DHA uptake by a human cerebral microvascular endothelial cell line (hCMEC/D3) increased in a time-dependent manner, and was inhibited by unlabeled DHA and a known FATP1 substrate, oleic acid. Knock-down of FATP1 in hCMEC/D3 cells with specific siRNA showed that FATP1-mediated uptake accounts for 59.2-73.0% of total [14 C]-DHA uptake by the cells. Insulin treatment for 30 min induced translocation of FATP1 protein to the plasma membrane in hCMEC/D3 cells and enhanced [14 C]-DHA uptake. Immunohistochemical analysis of mouse brain sections showed that FATP1 protein is preferentially localized at the basal membrane of brain microvessel endothelial cells. We found that two neuroprotective substances, taurine and biotin, in addition to DHA, undergo FATP1-mediated efflux. Overall, our results suggest that FATP1 localized at the basal membrane of brain microvessels contributes to the transport of DHA, taurine and biotin into the brain, and insulin rapidly increases DHA supply to the brain by promoting translocation of FATP1 to the membrane. Read the Editorial Comment for this article on page 324.
Collapse
Affiliation(s)
- Yusuke Ochiai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Japan Agency for Medical Research and Development (AMED) CREST, Tokyo, Japan
| | - Masanori Tachikawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Sanshiro Aizawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| |
Collapse
|
35
|
α-MSH and Foxc2 promote fatty acid oxidation through C/EBPβ negative transcription in mice adipose tissue. Sci Rep 2016; 6:36661. [PMID: 27819350 PMCID: PMC5098202 DOI: 10.1038/srep36661] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/18/2016] [Indexed: 01/18/2023] Open
Abstract
Alpha melanocyte stimulating hormone (α-MSH) and Forkhead box C2 protein (Foxc2) enhance lipolysis in multiple tissues. However, their relationship in adipose fatty acid oxidation (FAO) remains unclear. Here, we demonstrated that α-MSH and Foxc2 increased palmitate oxidation to CO2 in white (WAT) and brown adipose tissue (BAT). C/EBPβ expression was reduced by α-MSH and Foxc2. FFA level was elevated by α-MSH and pc-Foxc2 treatment along with increased FAO in white and brown adipocytes. The expression of FAO key enzymes, medium-chain acyl-CoA dehydrogenase (MCAD) and long-chain acyl-CoA dehydrogenase (LCAD) were increased in α-MSH and pc-Foxc2 group. Combination of α-MSH and Foxc2 treatment synergistically promoted FAO through increasing the activity of CPT-1 and phosphorylation of ACC. We found C/EBPβ bind to MC5R and Foxc2 promoter regions and inhibited FAO. cAMP level was increased by α-MSH and Foxc2 individually treated or combined treatment. Furthermore, cAMP/PKA pathway-specific inhibitor (H89) blocked the FAO, despite in α-MSH and Foxc2 both added group. While forskolin, the cAMP agonist, promoted FAO and enhanced the effect of α-MSH and Foxc2. Collectively, α-MSH and Foxc2 mutual promote FAO in WAT and BAT via cAMP/PKA signal pathway. And C/EBPβ as a transcription suppressor inhibits α-MSH and Foxc2 expression and FAO.
Collapse
|
36
|
Temprano A, Sembongi H, Han GS, Sebastián D, Capellades J, Moreno C, Guardiola J, Wabitsch M, Richart C, Yanes O, Zorzano A, Carman GM, Siniossoglou S, Miranda M. Redundant roles of the phosphatidate phosphatase family in triacylglycerol synthesis in human adipocytes. Diabetologia 2016; 59:1985-94. [PMID: 27344312 PMCID: PMC4969345 DOI: 10.1007/s00125-016-4018-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS In mammals, the evolutionary conserved family of Mg(2+)-dependent phosphatidate phosphatases (PAP1), involved in phospholipid and triacylglycerol synthesis, consists of lipin-1, lipin-2 and lipin-3. While mutations in the murine Lpin1 gene cause lipodystrophy and its knockdown in mouse 3T3-L1 cells impairs adipogenesis, deleterious mutations of human LPIN1 do not affect adipose tissue distribution. However, reduced LPIN1 and PAP1 activity has been described in participants with type 2 diabetes. We aimed to characterise the roles of all lipin family members in human adipose tissue and adipogenesis. METHODS The expression of the lipin family was analysed in adipose tissue in a cross-sectional study. Moreover, the effects of lipin small interfering RNA (siRNA)-mediated depletion on in vitro human adipogenesis were assessed. RESULTS Adipose tissue gene expression of the lipin family is altered in type 2 diabetes. Depletion of every lipin family member in a human Simpson-Golabi-Behmel syndrome (SGBS) pre-adipocyte cell line, alters expression levels of adipogenic transcription factors and lipid biosynthesis genes in early stages of differentiation. Lipin-1 knockdown alone causes a 95% depletion of PAP1 activity. Despite the reduced PAP1 activity and alterations in early adipogenesis, lipin-silenced cells differentiate and accumulate neutral lipids. Even combinatorial knockdown of lipins shows mild effects on triacylglycerol accumulation in mature adipocytes. CONCLUSIONS/INTERPRETATION Overall, our data support the hypothesis of alternative pathways for triacylglycerol synthesis in human adipocytes under conditions of repressed lipin expression. We propose that induction of alternative lipid phosphate phosphatases, along with the inhibition of lipid hydrolysis, contributes to the maintenance of triacylglycerol content to near normal levels.
Collapse
Affiliation(s)
- Ana Temprano
- Joan XXIII University Hospital, Pere Virgili Health Research Institut (IISPV), Modular Building, C/ Mallafre Guasch, Tarragona, 43005, Spain
- Department of Biochemistry and Molecular Biology, Rovira i Virgili University, Tarragona, Spain
| | - Hiroshi Sembongi
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge, CB2 0XY, UK
- , Chesterford Research Park, Little Chesterford, Saffron Walden, UK
| | - Gil-Soo Han
- Department of Food Science and the Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - David Sebastián
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Capellades
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Centre for Omic Sciences, Rovira i Virgili University, Reus, Spain
| | - Cristóbal Moreno
- Joan XXIII University Hospital, Pere Virgili Health Research Institut (IISPV), Modular Building, C/ Mallafre Guasch, Tarragona, 43005, Spain
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Guardiola
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Louisville, Louisville, KY, USA
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Interdisciplinary Obesity Clinic, University Clinic for Child and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Cristóbal Richart
- Joan XXIII University Hospital, Pere Virgili Health Research Institut (IISPV), Modular Building, C/ Mallafre Guasch, Tarragona, 43005, Spain
- GEMMAIR Research Group - Applied Medicine, Department of Medicine and Surgery, Rovira i Virgili University (URV), Tarragona, Spain
| | - Oscar Yanes
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Centre for Omic Sciences, Rovira i Virgili University, Reus, Spain
- Department of Electronic Engineering, Rovira i Virgili University, Tarragona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - George M Carman
- Department of Food Science and the Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Symeon Siniossoglou
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge, CB2 0XY, UK.
| | - Merce Miranda
- Joan XXIII University Hospital, Pere Virgili Health Research Institut (IISPV), Modular Building, C/ Mallafre Guasch, Tarragona, 43005, Spain.
- Biomedical Research Networking Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain, .
| |
Collapse
|
37
|
Ratman D, Mylka V, Bougarne N, Pawlak M, Caron S, Hennuyer N, Paumelle R, De Cauwer L, Thommis J, Rider MH, Libert C, Lievens S, Tavernier J, Staels B, De Bosscher K. Chromatin recruitment of activated AMPK drives fasting response genes co-controlled by GR and PPARα. Nucleic Acids Res 2016; 44:10539-10553. [PMID: 27576532 PMCID: PMC5159533 DOI: 10.1093/nar/gkw742] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 12/21/2022] Open
Abstract
Adaptation to fasting involves both Glucocorticoid Receptor (GRα) and Peroxisome Proliferator-Activated Receptor α (PPARα) activation. Given both receptors can physically interact we investigated the possibility of a genome-wide cross-talk between activated GR and PPARα, using ChIP- and RNA-seq in primary hepatocytes. Our data reveal extensive chromatin co-localization of both factors with cooperative induction of genes controlling lipid/glucose metabolism. Key GR/PPAR co-controlled genes switched from transcriptional antagonism to cooperativity when moving from short to prolonged hepatocyte fasting, a phenomenon coinciding with gene promoter recruitment of phosphorylated AMP-activated protein kinase (AMPK) and blocked by its pharmacological inhibition. In vitro interaction studies support trimeric complex formation between GR, PPARα and phospho-AMPK. Long-term fasting in mice showed enhanced phosphorylation of liver AMPK and GRα Ser211. Phospho-AMPK chromatin recruitment at liver target genes, observed upon prolonged fasting in mice, is dampened by refeeding. Taken together, our results identify phospho-AMPK as a molecular switch able to cooperate with nuclear receptors at the chromatin level and reveal a novel adaptation mechanism to prolonged fasting.
Collapse
Affiliation(s)
- Dariusz Ratman
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Viacheslav Mylka
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Nadia Bougarne
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Michal Pawlak
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Sandrine Caron
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Nathalie Hennuyer
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Réjane Paumelle
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Lode De Cauwer
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Jonathan Thommis
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Mark H Rider
- de Duve Institute and Université catholique de Louvain, 1200 Brussels, Belgium
| | - Claude Libert
- Inflammation Research Center, VIB, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Sam Lievens
- Department of Biochemistry, Ghent University, 9000 Ghent, Belgium.,Receptor Research Laboratories, Cytokine Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium
| | - Jan Tavernier
- Department of Biochemistry, Ghent University, 9000 Ghent, Belgium.,Receptor Research Laboratories, Cytokine Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium
| | - Bart Staels
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France.,CHU Lille, Department of Biology, 59000 Lille, France
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium .,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
38
|
Duan Y, Duan Y, Li F, Li Y, Guo Q, Ji Y, Tan B, Li T, Yin Y. Effects of supplementation with branched-chain amino acids to low-protein diets on expression of genes related to lipid metabolism in skeletal muscle of growing pigs. Amino Acids 2016; 48:2131-44. [PMID: 27156063 DOI: 10.1007/s00726-016-2223-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/24/2016] [Indexed: 11/29/2022]
Abstract
Branched-chain amino acids (BCAA), including leucine (Leu), isoleucine (Ile), and valine (Val), play critical roles in energy homeostasis and lipid metabolism in addition to their other functions, such as in protein metabolism. This study investigated the effects of different dietary BCAA ratios on the intramuscular fat (IMF) content and fatty acid composition in different location of skeletal muscles, including the longissimus dorsi (LD), biceps femoris (BF), and psoas major (PM) muscles of growing pigs, and also examined the mRNA expression levels of genes involved in lipid metabolism in these muscle tissues. The experiment was performed on 40 growing pigs (Large White × Landrace) with a similar initial weight (9.85 ± 0.35 kg). The pigs were randomly assigned to one of five diets: diet A was a positive control and contained 20 % crude protein (CP) with a Leu:Ile:Val ratio of 1:0.51:0.63 according to the recommendation of the National Research Council (NRC); for diets B to E, the CP level was reduced to 17 %, and the Leu:Ile:Val ratios were 1:1:1, 1:0.75:0.75, 1:0.51:0.63, and 1:0.25:0.25, respectively. No significant difference was observed in the average feed intake and feed efficiency of the pigs fed the low protein diet (17 % CP) with BCAA treatments relative to the positive control. However, there was a tendency for increased feed efficiency of the 1:0.75:0.75 group compared with the 1:1:1 group (P = 0.09). The BCAA ratio of 1:0.75:0.75 (17 % CP) increased the IMF content of BF muscle (P < 0.01). Moreover, varied dietary BCAA supplementation with a reduced protein level had different effects on the fatty acid composition of the LD, BF, and PM muscles. The BCAA ratio of 1:0.51:0.63-1:0.75:0.75 (17 % CP) significantly lowered the ratio of n-6 to n-3 polyunsaturated fatty acid in these muscles compared with the positive control group (20 % CP). This effect was associated with an increase in mRNA expression levels of acetyl-CoA carboxylase, lipoprotein lipase, fatty acid transport protein, and fatty acid binding protein 4 in the muscles (P < 0.05). The results indicated that the reduced protein diet (17 % CP) with the BCAA ratio within 1:0.25:0.25-1:0.75:0.75 could increase the IMF content in BF muscle and significantly improve the fatty acid composition in different skeletal muscles accompanied by changes in the expression of genes involved in lipid metabolism, compared with those in the pigs that received adequate dietary protein (20 %), which might result in improved eating quality and nutritional value of the meat.
Collapse
Affiliation(s)
- Yehui Duan
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yangmiao Duan
- University of Chinese Academy of Sciences, Beijing, 100039, China.,Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fengna Li
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China. .,Hunan Co-Innovation Center of Animal Production Safety (CICAPS), Changsha, 410125, China.
| | - Yinghui Li
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Qiuping Guo
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yujiao Ji
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China
| | - Bie Tan
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety (CICAPS), Changsha, 410125, China.,Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, 410128, China
| | - Tiejun Li
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China
| | - Yulong Yin
- Key Laboratory of Agroecology in Subtropical Region, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, Research Center for Healthy Breeding Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Institute of Subtropical Agriculture, Ministry of Agriculture, Chinese Academy of Science, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China. .,Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, 410128, China. .,School of Biology, Hunan Normal University, Changsha, 410018, China.
| |
Collapse
|
39
|
Freitag Luglio H. Genetic Variation of Fatty Acid Oxidation and Obesity, A Literature Review. INTERNATIONAL JOURNAL OF BIOMEDICAL SCIENCE : IJBS 2016; 12:1-8. [PMID: 27127449 PMCID: PMC4841982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Modulation of fat metabolism is an important component of the etiology of obesity as well as individual response to weight loss program. The influence of lipolysis process had receives many attentions in recent decades. Compared to that, fatty acid oxidation which occurred after lipolysis seems to be less exposed. There are limited publications on how fatty acid oxidation influences predisposition to obesity, especially the importance of genetic variations of fatty acid oxidation proteins on development of obesity. The aim of this review is to provide recent knowledge on how polymorphism of genes related fatty acid oxidation is obtained. Studies in human as well as animal model showed that disturbance of genes related fatty acid oxidation process gave impact on body weight and risks to obesity. Several polymorphisms on CD36, CPT, ACS and FABP had been shown to be related to obesity either by regulating enzymatic activity or directly influence fatty acid oxidation process.
Collapse
|
40
|
Sanghez V, Cubuk C, Sebastián-Leon P, Carobbio S, Dopazo J, Vidal-Puig A, Bartolomucci A. Chronic subordination stress selectively downregulates the insulin signaling pathway in liver and skeletal muscle but not in adipose tissue of male mice. Stress 2016; 19:214-24. [PMID: 26946982 PMCID: PMC4841025 DOI: 10.3109/10253890.2016.1151491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Chronic stress has been associated with obesity, glucose intolerance, and insulin resistance. We developed a model of chronic psychosocial stress (CPS) in which subordinate mice are vulnerable to obesity and the metabolic-like syndrome while dominant mice exhibit a healthy metabolic phenotype. Here we tested the hypothesis that the metabolic difference between subordinate and dominant mice is associated with changes in functional pathways relevant for insulin sensitivity, glucose and lipid homeostasis. Male mice were exposed to CPS for four weeks and fed either a standard diet or a high-fat diet (HFD). We first measured, by real-time PCR candidate genes, in the liver, skeletal muscle, and the perigonadal white adipose tissue (pWAT). Subsequently, we used a probabilistic analysis approach to analyze different ways in which signals can be transmitted across the pathways in each tissue. Results showed that subordinate mice displayed a drastic downregulation of the insulin pathway in liver and muscle, indicative of insulin resistance, already on standard diet. Conversely, pWAT showed molecular changes suggestive of facilitated fat deposition in an otherwise insulin-sensitive tissue. The molecular changes in subordinate mice fed a standard diet were greater compared to HFD-fed controls. Finally, dominant mice maintained a substantially normal metabolic and molecular phenotype even when fed a HFD. Overall, our data demonstrate that subordination stress is a potent stimulus for the downregulation of the insulin signaling pathway in liver and muscle and a major risk factor for the development of obesity, insulin resistance, and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Valentina Sanghez
- Department of Integrative Biology and Physiology, University of Minnesota,
Minneapolis,
MN,
USA
- Department of Neuroscience, University of Parma, Parma,
Italy
- Correspondence: Alessandro Bartolomucci,
Department of Integrative Biology and Physiology, University of Minnesota,
Minneapolis,
MN,
USA. Tel: +1-612-626-7006. Fax: +1-612-625-5149. E-mail:
| | - Cankut Cubuk
- Department of Computational Genomics, Centro de Investigación Principe Felipe, Valencia,
Spain
| | - Patricia Sebastián-Leon
- Department of Computational Genomics, Centro de Investigación Principe Felipe, Valencia,
Spain
| | - Stefania Carobbio
- Wellcome Trust MRC Metabolic Disease Unit, Institute Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge,
UK
| | - Joaquin Dopazo
- Department of Computational Genomics, Centro de Investigación Principe Felipe, Valencia,
Spain
| | - Antonio Vidal-Puig
- Wellcome Trust MRC Metabolic Disease Unit, Institute Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge,
UK
- Wellcome Trust Sanger Institute, Hinxton,
UK
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota,
Minneapolis,
MN,
USA
- Correspondence: Alessandro Bartolomucci,
Department of Integrative Biology and Physiology, University of Minnesota,
Minneapolis,
MN,
USA. Tel: +1-612-626-7006. Fax: +1-612-625-5149. E-mail:
| |
Collapse
|
41
|
Gan L, Liu Z, Cao W, Zhang Z, Sun C. FABP4 reversed the regulation of leptin on mitochondrial fatty acid oxidation in mice adipocytes. Sci Rep 2015; 5:13588. [PMID: 26310911 PMCID: PMC4550852 DOI: 10.1038/srep13588] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 07/31/2015] [Indexed: 11/09/2022] Open
Abstract
Fatty acid binding protein 4 (FABP4), plays key role in fatty acid transportation and oxidation, and increases with leptin synergistically during adipose inflammation process. However, the regulation mechanism between FABP4 and leptin on mitochondrial fatty acid oxidation remains unclear. In this study, we found that FABP4 reduced the expression of leptin, CPT-1 and AOX1 in mice adipocytes. Conversely, FABP4 was down-regulated in a time-dependent manner by leptin treatment. Additionally, forced expression of FABP4 attenuated the expression of PGC1-α, UCP2, CPT-1, AOX1 and COX2 compared with leptin incubation. Moreover, mitochondrial membrane potential, fatty acid oxidation enzyme medium-chain acyl-CoA dehydrogenase (MCAD), long-chain acyl-CoA dehydrogenase (LCAD) and Cyt C levels were reduced in response to the overexpression of FABP4. These reductions correspond well with the reduced release of free fatty acid and the inactivation of mitochondrial complexes I and III by FABP4 overexpression. Furthermore, addition of the Akt/mTOR pathway-specific inhibitor (MK2206) blocked the mitochondrial fatty acid oxidation and respiration factors, whereas interference of FABP4 overcame these effects. Taken together, FABP4 could reverse the activation of the leptin-induced mitochondrial fatty acid oxidation, and the inhibition of Akt/mTOR signal pathway played a key role in this process.
Collapse
Affiliation(s)
- Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Weina Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenzhen Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
42
|
Georgiou DK, Dagnino-Acosta A, Lee CS, Griffin DM, Wang H, Lagor WR, Pautler RG, Dirksen RT, Hamilton SL. Ca2+ Binding/Permeation via Calcium Channel, CaV1.1, Regulates the Intracellular Distribution of the Fatty Acid Transport Protein, CD36, and Fatty Acid Metabolism. J Biol Chem 2015; 290:23751-65. [PMID: 26245899 DOI: 10.1074/jbc.m115.643544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Indexed: 01/08/2023] Open
Abstract
Ca(2+) permeation and/or binding to the skeletal muscle L-type Ca(2+) channel (CaV1.1) facilitates activation of Ca(2+)/calmodulin kinase type II (CaMKII) and Ca(2+) store refilling to reduce muscle fatigue and atrophy (Lee, C. S., Dagnino-Acosta, A., Yarotskyy, V., Hanna, A., Lyfenko, A., Knoblauch, M., Georgiou, D. K., Poché, R. A., Swank, M. W., Long, C., Ismailov, I. I., Lanner, J., Tran, T., Dong, K., Rodney, G. G., Dickinson, M. E., Beeton, C., Zhang, P., Dirksen, R. T., and Hamilton, S. L. (2015) Skelet. Muscle 5, 4). Mice with a mutation (E1014K) in the Cacna1s (α1 subunit of CaV1.1) gene that abolishes Ca(2+) binding within the CaV1.1 pore gain more body weight and fat on a chow diet than control mice, without changes in food intake or activity, suggesting that CaV1.1-mediated CaMKII activation impacts muscle energy expenditure. We delineate a pathway (Cav1.1→ CaMKII→ NOS) in normal skeletal muscle that regulates the intracellular distribution of the fatty acid transport protein, CD36, altering fatty acid metabolism. The consequences of blocking this pathway are decreased mitochondrial β-oxidation and decreased energy expenditure. This study delineates a previously uncharacterized CaV1.1-mediated pathway that regulates energy utilization in skeletal muscle.
Collapse
Affiliation(s)
- Dimitra K Georgiou
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Adan Dagnino-Acosta
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Chang Seok Lee
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Deric M Griffin
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Hui Wang
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - William R Lagor
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Robia G Pautler
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| | - Robert T Dirksen
- the Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York 14642
| | - Susan L Hamilton
- From the Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030 and
| |
Collapse
|
43
|
Schrader M, Costello J, Godinho LF, Islinger M. Peroxisome-mitochondria interplay and disease. J Inherit Metab Dis 2015; 38:681-702. [PMID: 25687155 DOI: 10.1007/s10545-015-9819-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 12/16/2022]
Abstract
Peroxisomes and mitochondria are ubiquitous, highly dynamic organelles with an oxidative type of metabolism in eukaryotic cells. Over the years, substantial evidence has been provided that peroxisomes and mitochondria exhibit a close functional interplay which impacts on human health and development. The so-called "peroxisome-mitochondria connection" includes metabolic cooperation in the degradation of fatty acids, a redox-sensitive relationship, an overlap in key components of the membrane fission machineries and cooperation in anti-viral signalling and defence. Furthermore, combined peroxisome-mitochondria disorders with defects in organelle division have been revealed. In this review, we present the latest progress in the emerging field of peroxisomal and mitochondrial interplay in mammals with a particular emphasis on cooperative fatty acid β-oxidation, redox interplay, organelle dynamics, cooperation in anti-viral signalling and the resulting implications for disease.
Collapse
Affiliation(s)
- Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK,
| | | | | | | |
Collapse
|
44
|
Inflammatory and Metabolic Alterations of Kager's Fat Pad in Chronic Achilles Tendinopathy. PLoS One 2015; 10:e0127811. [PMID: 25996876 PMCID: PMC4440827 DOI: 10.1371/journal.pone.0127811] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/18/2015] [Indexed: 12/14/2022] Open
Abstract
Background Achilles tendinopathy is a painful inflammatory condition characterized by swelling, stiffness and reduced function of the Achilles tendon. Kager’s fat pad is an adipose tissue located in the area anterior to the Achilles tendon. Observations reveal a close physical interplay between Kager’s fat pad and its surrounding structures during movement of the ankle, suggesting that Kager’s fat pad may stabilize and protect the mechanical function of the ankle joint. Aim The aim of this study was to characterize whether Achilles tendinopathy was accompanied by changes in expression of inflammatory markers and metabolic enzymes in Kager’s fat pad. Methods A biopsy was taken from Kager’s fat pad from 31 patients with chronic Achilles tendinopathy and from 13 healthy individuals. Gene expression was measured by reverse transcription-quantitative PCR. Focus was on genes related to inflammation and lipid metabolism. Results Expression of the majority of analyzed inflammatory marker genes was increased in patients with Achilles tendinopathy compared to that in healthy controls. Expression patterns of the patient group were consistent with reduced lipolysis and increased fatty acid β-oxidation. In the fat pad, the pain-signaling neuropeptide substance P was found to be present in one third of the subjects in the Achilles tendinopathy group but in none of the healthy controls. Conclusion Gene expression changes in Achilles tendinopathy patient samples were consistent with Kager’s fat pad being more inflamed than in the healthy control group. Additionally, the results indicate an altered lipid metabolism in Kager’s fat pad of Achilles tendinopathy patients.
Collapse
|
45
|
Monaco C, Whitfield J, Jain SS, Spriet LL, Bonen A, Holloway GP. Activation of AMPKα2 Is Not Required for Mitochondrial FAT/CD36 Accumulation during Exercise. PLoS One 2015; 10:e0126122. [PMID: 25965390 PMCID: PMC4429092 DOI: 10.1371/journal.pone.0126122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/30/2015] [Indexed: 12/30/2022] Open
Abstract
Exercise has been shown to induce the translocation of fatty acid translocase (FAT/CD36), a fatty acid transport protein, to both plasma and mitochondrial membranes. While previous studies have examined signals involved in the induction of FAT/CD36 translocation to sarcolemmal membranes, to date the signaling events responsible for FAT/CD36 accumulation on mitochondrial membranes have not been investigated. In the current study muscle contraction rapidly increased FAT/CD36 on plasma membranes (7.5 minutes), while in contrast, FAT/CD36 only increased on mitochondrial membranes after 22.5 minutes of muscle contraction, a response that was exercise-intensity dependent. Considering that previous research has shown that AMP activated protein kinase (AMPK) α2 is not required for FAT/CD36 translocation to the plasma membrane, we investigated whether AMPK α2 signaling is necessary for mitochondrial FAT/CD36 accumulation. Administration of 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) induced AMPK phosphorylation, and resulted in FAT/CD36 accumulation on SS mitochondria, suggesting AMPK signaling may mediate this response. However, SS mitochondrial FAT/CD36 increased following acute treadmill running in both wild-type (WT) and AMPKα 2 kinase dead (KD) mice. These data suggest that AMPK signaling is not required for SS mitochondrial FAT/CD36 accumulation. The current data also implicates alternative signaling pathways that are exercise-intensity dependent, as IMF mitochondrial FAT/CD36 content only occurred at a higher power output. Taken altogether the current data suggests that activation of AMPK signaling is sufficient but not required for exercise-induced accumulation in mitochondrial FAT/CD36.
Collapse
Affiliation(s)
- Cynthia Monaco
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Jamie Whitfield
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Swati S. Jain
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Lawrence L. Spriet
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Arend Bonen
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Graham P. Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
- * E-mail:
| |
Collapse
|
46
|
Effects of dietary n-6:n-3 PUFA ratio on fatty acid composition, free amino acid profile and gene expression of transporters in finishing pigs. Br J Nutr 2015; 113:739-48. [PMID: 25704496 DOI: 10.1017/s0007114514004346] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Revealing the expression patterns of fatty acid and amino acid transporters as affected by dietary n-6:n-3 PUFA ratio would be useful for further clarifying the importance of the balance between n-6 and n-3 PUFA. A total of ninety-six finishing pigs were fed one of four diets with the ratio of 1:1, 2·5:1, 5:1 and 10:1. Pigs fed the dietary n-6:n-3 PUFA ratio of 5:1 had the highest (P< 0·05) daily weight gain, and those fed the dietary n-6:n-3 PUFA ratio of 1:1 had the largest loin muscle area (P< 0·01). The concentration of n-3 PUFA was raised as the ratio declined (P< 0·05) in the longissimus dorsi and subcutaneous adipose tissue. The contents of tryptophan, tasty amino acids and branched-chain amino acids in the longissimus dorsi were enhanced in pigs fed the dietary n-6:n-3 PUFA ratios of 1:1-5:1. The mRNA expression level of the fatty acid transporter fatty acid transport protein-1 (FATP-1) was declined (P< 0·05) in the longissimus dorsi of pigs fed the dietary n-6:n-3 PUFA ratios of 1:1-5:1, and increased (P< 0·05) in the subcutaneous adipose tissue of pigs fed the dietary n-6:n-3 PUFA ratios of 5:1 and 10:1. The expression profile of FATP-4 was similar to those of FATP-1 in the adipose tissue. The mRNA expression level of the amino acid transceptors LAT1 and SNAT2 was up-regulated (P< 0·05) in the longissimus dorsi of pigs fed the dietary n-6:n-3 PUFA ratios of 1:1 and 2·5:1. In conclusion, maintaining the dietary n-6:n-3 PUFA ratios of 1:1-5:1 would facilitate the absorption and utilisation of fatty acids and free amino acids, and result in improved muscle and adipose composition.
Collapse
|
47
|
Liepinsh E, Makrecka-Kuka M, Kuka J, Vilskersts R, Makarova E, Cirule H, Loza E, Lola D, Grinberga S, Pugovics O, Kalvins I, Dambrova M. Inhibition of L-carnitine biosynthesis and transport by methyl-γ-butyrobetaine decreases fatty acid oxidation and protects against myocardial infarction. Br J Pharmacol 2015; 172:1319-32. [PMID: 25363063 DOI: 10.1111/bph.13004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 10/21/2014] [Accepted: 10/28/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE The important pathological consequences of ischaemic heart disease arise from the detrimental effects of the accumulation of long-chain acylcarnitines in the case of acute ischaemia-reperfusion. The aim of this study is to test whether decreasing the L-carnitine content represents an effective strategy to decrease accumulation of long-chain acylcarnitines and to reduce fatty acid oxidation in order to protect the heart against acute ischaemia-reperfusion injury. KEY RESULTS In this study, we used a novel compound, 4-[ethyl(dimethyl)ammonio]butanoate (Methyl-GBB), which inhibits γ-butyrobetaine dioxygenase (IC₅₀ 3 μM) and organic cation transporter 2 (OCTN2, IC₅₀ 3 μM), and, in turn, decreases levels of L-carnitine and acylcarnitines in heart tissue. Methyl-GBB reduced both mitochondrial and peroxisomal palmitate oxidation rates by 44 and 53% respectively. In isolated hearts treated with Methyl-GBB, uptake and oxidation rates of labelled palmitate were decreased by 40%, while glucose oxidation was increased twofold. Methyl-GBB (5 or 20 mg·kg(-1)) decreased the infarct size by 45-48%. In vivo pretreatment with Methyl-GBB (20 mg·kg(-1)) attenuated the infarct size by 45% and improved 24 h survival of rats by 20-30%. CONCLUSIONS AND IMPLICATIONS Reduction of L-carnitine and long-chain acylcarnitine content by the inhibition of OCTN2 represents an effective strategy to protect the heart against ischaemia-reperfusion-induced damage. Methyl-GBB treatment exerted cardioprotective effects and increased survival by limiting long-chain fatty acid oxidation and facilitating glucose metabolism.
Collapse
Affiliation(s)
- E Liepinsh
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liepinsh E, Makrecka M, Kuka J, Cirule H, Makarova E, Sevostjanovs E, Grinberga S, Vilskersts R, Lola D, Loza E, Stonans I, Pugovics O, Dambrova M. Selective inhibition of OCTN2 is more effective than inhibition of gamma-butyrobetaine dioxygenase to decrease the availability of l-carnitine and to reduce myocardial infarct size. Pharmacol Res 2014; 85:33-8. [DOI: 10.1016/j.phrs.2014.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/09/2014] [Accepted: 05/05/2014] [Indexed: 12/24/2022]
|
49
|
Guitart M, Osorio-Conles Ó, Pentinat T, Cebrià J, García-Villoria J, Sala D, Sebastián D, Zorzano A, Ribes A, Jiménez-Chillarón JC, García-Martínez C, Gómez-Foix AM. Fatty acid transport protein 1 (FATP1) localizes in mitochondria in mouse skeletal muscle and regulates lipid and ketone body disposal. PLoS One 2014; 9:e98109. [PMID: 24858472 PMCID: PMC4032244 DOI: 10.1371/journal.pone.0098109] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 04/29/2014] [Indexed: 12/20/2022] Open
Abstract
FATP1 mediates skeletal muscle cell fatty acid import, yet its intracellular localization and metabolic control role are not completely defined. Here, we examine FATP1 localization and metabolic effects of its overexpression in mouse skeletal muscle. The FATP1 protein was detected in mitochondrial and plasma membrane fractions, obtained by differential centrifugation, of mouse gastrocnemius muscle. FATP1 was most abundant in purified mitochondria, and in the outer membrane and soluble intermembrane, but not in the inner membrane plus matrix, enriched subfractions of purified mitochondria. Immunogold electron microscopy localized FATP1-GFP in mitochondria of transfected C2C12 myotubes. FATP1 was overexpressed in gastrocnemius mouse muscle, by adenovirus-mediated delivery of the gene into hindlimb muscles of newborn mice, fed after weaning a chow or high-fat diet. Compared to GFP delivery, FATP1 did not alter body weight, serum fed glucose, insulin and triglyceride levels, and whole-body glucose tolerance, in either diet. However, fatty acid levels were lower and β-hydroxybutyrate levels were higher in FATP1- than GFP-mice, irrespective of diet. Moreover, intramuscular triglyceride content was lower in FATP1- versus GFP-mice regardless of diet, and β-hydroxybutyrate content was unchanged in high-fat-fed mice. Electroporation-mediated FATP1 overexpression enhanced palmitate oxidation to CO2, but not to acid-soluble intermediate metabolites, while CO2 production from β-hydroxybutyrate was inhibited and that from glucose unchanged, in isolated mouse gastrocnemius strips. In summary, FATP1 was localized in mitochondria, in the outer membrane and intermembrane parts, of mouse skeletal muscle, what may be crucial for its metabolic effects. Overexpressed FATP1 enhanced disposal of both systemic fatty acids and intramuscular triglycerides. Consistently, it did not contribute to the high-fat diet-induced metabolic dysregulation. However, FATP1 lead to hyperketonemia, likely secondary to the sparing of ketone body oxidation by the enhanced oxidation of fatty acids.
Collapse
Affiliation(s)
- Maria Guitart
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- * E-mail:
| | - Óscar Osorio-Conles
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Thais Pentinat
- Hospital Sant Joan de Déu, Endocrinology, Esplugues, Barcelona, Spain
| | - Judith Cebrià
- Hospital Sant Joan de Déu, Endocrinology, Esplugues, Barcelona, Spain
| | - Judit García-Villoria
- Sección de Errores Congénitos del Metabolismo (IBC), Servicio de Bioquímica y Genética Molecular, Hospital Clínico, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain
| | - David Sala
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - David Sebastián
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Antonio Zorzano
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Antonia Ribes
- Sección de Errores Congénitos del Metabolismo (IBC), Servicio de Bioquímica y Genética Molecular, Hospital Clínico, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain
| | | | - Celia García-Martínez
- Departament de Patologia i Terapèutica Experimental, UB, Hospitalet de Llobregat, Barcelona, Spain
| | - Anna M. Gómez-Foix
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| |
Collapse
|
50
|
Abstract
Long-chain fatty acyl-coenzyme As (CoAs) are critical regulatory molecules and metabolic intermediates. The initial step in their synthesis is the activation of fatty acids by one of 13 long-chain acyl-CoA synthetase isoforms. These isoforms are regulated independently and have different tissue expression patterns and subcellular locations. Their acyl-CoA products regulate metabolic enzymes and signaling pathways, become oxidized to provide cellular energy, and are incorporated into acylated proteins and complex lipids such as triacylglycerol, phospholipids, and cholesterol esters. Their differing metabolic fates are determined by a network of proteins that channel the acyl-CoAs toward or away from specific metabolic pathways and serve as the basis for partitioning. This review evaluates the evidence for acyl-CoA partitioning by reviewing experimental data on proteins that are believed to contribute to acyl-CoA channeling, the metabolic consequences of loss of these proteins, and the potential role of maladaptive acyl-CoA partitioning in the pathogenesis of metabolic disease and carcinogenesis.
Collapse
|