1
|
Barbhuiya PA, Sen S, Pathak MP. Ameliorative role of bioactive phytoconstituents targeting obesity associated NAFLD by modulation of inflammation and lipogenesis pathways: a comprehensive review. PHYTOCHEMISTRY REVIEWS 2024; 23:969-996. [DOI: 10.1007/s11101-023-09912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2025]
|
2
|
Wang Y, Zeng D, Wei L, Chen J, Li H, Wen L, Huang G, Dai Z, Luo J, Sun J, Xi Q, Zhang Y, Chen T. Effects of emulsifiers on lipid metabolism and performance of yellow-feathered broilers. BMC Vet Res 2024; 20:246. [PMID: 38849831 PMCID: PMC11157903 DOI: 10.1186/s12917-024-04095-8] [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: 02/21/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Reducing production costs while producing high-quality livestock and poultry products is an ongoing concern in the livestock industry. The addition of oil to livestock and poultry diets can enhance feed palatability and improve growth performance. Emulsifiers can be used as potential feed supplements to improve dietary energy utilization and maintain the efficient productivity of broilers. Therefore, further investigation is warranted to evaluate whether dietary emulsifier supplementation can improve the efficiency of fat utilization in the diet of yellow-feathered broilers. In the present study, the effects of adding emulsifier to the diet on lipid metabolism and the performance of yellow-feathered broilers were tested. A total of 240 yellow-feasted broilers (21-day-old) were randomly divided into 4 groups (6 replicates per group, 10 broilers per replicate, half male and half female within each replicate). The groups were as follows: the control group (fed with basal diet), the group fed with basal diet supplemented with 500 mg/kg emulsifier, the group fed with a reduced oil diet (reduced by 1%) supplemented with 500 mg/kg emulsifier, and the group fed with a reduced oil diet supplemented with 500 mg/kg emulsifier. The trial lasted for 42 days, during which the average daily feed intake, average daily gain, and feed-to-gain ratio were measured. Additionally, the expression levels of lipid metabolism-related genes in the liver, abdominal fat and each intestinal segment were assessed. RESULTS The results showed that compared with the basal diet group, (1) The average daily gain of the basal diet + 500 mg/kg emulsifier group significantly increased (P < 0.05), and the half-even-chamber rate was significantly increased (P < 0.05); (2) The mRNA expression levels of Cd36, Dgat2, Apob, Fatp4, Fabp2, and Mttp in the small intestine were significantly increased (P < 0.05). (3) Furthermore, liver TG content significantly decreased (P < 0.05), and the mRNA expression level of Fasn in liver was significantly decreased (P < 0.05), while the expression of Apob, Lpl, Cpt-1, and Pparα significantly increased (P < 0.05). (4) The mRNA expression levels of Lpl and Fatp4 in adipose tissue were significantly increased (P < 0.05), while the expression of Atgl was significantly decreased (P < 0.05). (5) Compared with the reduced oil diet group, the half-evading rate and abdominal fat rate of broilers in the reduced oil diet + 500 mg/kg emulsifier group were significantly increased (P < 0.05), and the serum level of LDL-C increased significantly (P < 0.05)0.6) The mRNA expression levels of Cd36, Fatp4, Dgat2, Apob, and Mttp in the small intestine were significantly increased (P < 0.05). 7) The mRNA expression levels of Fasn and Acc were significantly decreased in the liver (P < 0.05), while the mRNA expression levels of Lpin1, Dgat2, Apob, Lpl, Cpt-1, and Pparα were significantly increased (P < 0.05). CONCLUSIONS These results suggest that dietary emulsifier can enhance the fat utilization efficiency of broilers by increasing the small intestinal fatty acid uptake capacity, inhibiting hepatic fatty acid synthesis and promoting hepatic TG synthesis and transport capacity. This study provides valuable insights for the potential use of emulsifier supplementation to improve the performance of broiler chickens.
Collapse
Affiliation(s)
- Yuxuan Wang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Dewei Zeng
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Limin Wei
- Hainan Key Laboratory of Tropical Animal Breeding and Epidemic Research, Institute of Animal Husbandry and Veterinary Research, Hainan Academy of Agricultural Sciences, Haikou, Hainan, 571100, China
| | - Jingshen Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Hongyi Li
- Yingdong College of Biology and Agriculture, Shaoguan University, Shaoguan, Guangdong, 512005, China
| | - Lijun Wen
- Guangdong Hainachuan Biotechnology Co., LTD, Guangzhou, Guangdong, 528515, China
| | - Guangming Huang
- Guangdong Hainachuan Biotechnology Co., LTD, Guangzhou, Guangdong, 528515, China
| | - Zhenqing Dai
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Junyi Luo
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jiajie Sun
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qianyun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yongliang Zhang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Ting Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
3
|
Zhang Z, Xu S, Song M, Huang W, Yan M, Li X. Association between blood lipid levels and the risk of liver cancer: a systematic review and meta-analysis. Cancer Causes Control 2024; 35:943-953. [PMID: 38376693 PMCID: PMC11129988 DOI: 10.1007/s10552-024-01853-9] [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: 08/08/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE The association between blood lipid levels and the risk of developing liver cancer remains a subject of ongoing debate. To elucidate this association, we conducted a meta-analysis by systematically incorporating data from all relevant prospective cohort studies. METHODS We conducted a systematic search of the PubMed, Embase, Web of Science, and Cochrane Library databases covering studies published from database inception through July 2023. This study included prospective cohort studies related to lipid profiles (e.g., total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) levels) that reported hazard ratios (HRs) or relative risks (RRs) with corresponding 95% confidence intervals (95% CIs) to investigate their association with the risk of liver cancer. During the analysis process, we used fixed-effects or random-effects models based on the level of heterogeneity among the studies and obtained pooled risk ratios using these models. To ensure the robustness and reliability of the study findings, we also conducted sensitivity analyses and publication bias analyses. RESULTS After conducting a systematic search, 12 studies were identified from a total of 11,904 articles and were included in the meta-analysis. These studies included a combined population of 10,765,221 participants, among whom 31,055 cases of liver cancer were reported. The analysis revealed that the pooled HR for the serum TC concentration (highest versus lowest) was 0.45 (95% CI = 0.35-0.58, I2 = 78%). For TGs, the HR was 0.67 (95% CI = 0.46-0.96, I2 = 86%), while for HDL-C, the HR was 0.72 (95% CI = 0.58-0.90, I2 = 65%). The HR for LDL-C was 0.51 (95% CI = 0.23-1.13, I2 = 93%). CONCLUSION The findings of this study indicate that serum TC, TG, and HDL-C levels are negatively associated with liver cancer risk, suggesting that higher concentrations of these lipids are associated with a reduced risk of liver cancer. However, no significant association has been found between LDL-C levels and liver cancer risk.
Collapse
Affiliation(s)
- Zhihui Zhang
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shicong Xu
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Meixuan Song
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
| | - Weirong Huang
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Manlin Yan
- School of Nursing, Southwest Medical University, Luzhou, 646000, China
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xianrong Li
- Department of Gastrointestinal surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
4
|
Yang Y, Yang C, Zhuang Z, Mao J, Chen A, Zhou T, Bai H, Jiang Y, Chang G, Wang Z. RNA-Seq Analysis Revealed circRNAs and Genes Associated with Abdominal Fat Deposition in Ducks. Animals (Basel) 2024; 14:260. [PMID: 38254429 PMCID: PMC10812634 DOI: 10.3390/ani14020260] [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: 11/21/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Fat deposition is an important factor affecting meat quality and feed conversion efficiency in meat ducks. This study aims to identify key circRNAs and genes affecting abdominal fat deposition. The correlations between abdominal fat and other growth performances were analyzed in 304 F2 generation of Cherry Valley duck Runzhou Crested White ducks, and an RNA-seq analysis of abdominal fat tissues from ducks with high and low rates of abdominal fat was performed. Growth performance results showed that Abdominal fat ratio and Intramuscular fat were significantly higher in the high rates of abdominal fat (HF)group than in the low rates of abdominal fat (LF) group for ducks. RNA-seq analysis of abdominal fat tissue unveiled 85 upregulated and 72 downregulated circRNAs among the differentially expressed ones. Notably, 74 circRNAs displayed more than four-fold differential expression, constituting 47.13% of the differentially expressed genes. Functional enrichment analysis of the differentially expressed circRNA source and target genes indicated that 17 circRNAs might partake in regulating duck abdominal fat production by influencing pathways like PPAR signaling, lipid droplets, and triglyceride metabolism. Lastly, multiple circRNA-microRNA-messenger RNA interaction networks were constructed. The results of this study establish the groundwork for understanding the molecular mechanisms that regulate abdominal fat deposition in ducks, offering a theoretical reference for the selective breeding of high-quality meat-producing ducks.
Collapse
Affiliation(s)
- Yunfeng Yang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Chunyan Yang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Jiaming Mao
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Anqi Chen
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Tingting Zhou
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China;
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China;
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Y.); (C.Y.); (Z.Z.); (J.M.); (A.C.); (T.Z.); (Y.J.); (G.C.)
| |
Collapse
|
5
|
Dehghan H, Ghasempour A, Sabeti Akbar-Abad M, Khademi Z, Sedighi M, Jamialahmadi T, Sahebkar A. An update on the therapeutic role of RNAi in NAFLD/NASH. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 204:45-67. [PMID: 38458743 DOI: 10.1016/bs.pmbts.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Unhealthy lifestyles have given rise to a growing epidemic of metabolic liver diseases, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). NAFLD often occurs as a consequence of obesity, and currently, there is no FDA-approved drug for its treatment. However, therapeutic oligonucleotides, such as RNA interference (RNAi), represent a promising class of pharmacotherapy that can target previously untreatable conditions. The potential significance of RNAi in maintaining physiological homeostasis, understanding pathogenesis, and improving metabolic liver diseases, including NAFLD, is discussed in this article. We explore why NAFLD/NASH is an ideal target for therapeutic oligonucleotides and provide insights into the delivery platforms of RNAi and its therapeutic role in addressing NAFLD/NASH.
Collapse
Affiliation(s)
- Hamideh Dehghan
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Ghasempour
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Zahra Khademi
- Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahsa Sedighi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Irshad Z, Lund J, Sillars A, Løvsletten NG, Gharanei S, Salt IP, Freeman DJ, Gill JMR, Thoresen GH, Rustan AC, Zammit VA. The roles of DGAT1 and DGAT2 in human myotubes are dependent on donor patho-physiological background. FASEB J 2023; 37:e23209. [PMID: 37779421 PMCID: PMC10947296 DOI: 10.1096/fj.202300960rr] [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: 05/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023]
Abstract
The roles of DGAT1 and DGAT2 in lipid metabolism and insulin responsiveness of human skeletal muscle were studied using cryosections and myotubes prepared from muscle biopsies from control, athlete, and impaired glucose regulation (IGR) cohorts of men. The previously observed increases in intramuscular triacylglycerol (IMTG) in athletes and IGR were shown to be related to an increase in lipid droplet (LD) area in type I fibers in athletes but, conversely, in type II fibers in IGR subjects. Specific inhibition of both diacylglycerol acyltransferase (DGAT) 1 and 2 decreased fatty acid (FA) uptake by myotubes, whereas only DGAT2 inhibition also decreased fatty acid oxidation. Fatty acid uptake in myotubes was negatively correlated with the lactate thresholds of the respective donors. DGAT2 inhibition lowered acetate uptake and oxidation in myotubes from all cohorts whereas DGAT1 inhibition had no effect. A positive correlation between acetate oxidation in myotubes and resting metabolic rate (RMR) from fatty acid oxidation in vivo was observed. Myotubes from athletes and IGR had higher rates of de novo lipogenesis from acetate that were normalized by DGAT2 inhibition. Moreover, DGAT2 inhibition in myotubes also resulted in increased insulin-induced Akt phosphorylation. The differential effects of DGAT1 and DGAT2 inhibition suggest that the specialized role of DGAT2 in esterifying nascent diacylglycerols and de novo synthesized FA is associated with synthesis of a pool of triacylglycerol, which upon hydrolysis results in effectors that promote mitochondrial fatty acid oxidation but decrease insulin signaling in skeletal muscle cells.
Collapse
Affiliation(s)
- Zehra Irshad
- Translational and Experimental Medicine, Warwick Medical SchoolUniversity of WarwickCoventryUK
| | - Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
| | - Anne Sillars
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Nils Gunnar Løvsletten
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
| | - Seley Gharanei
- Translational and Experimental Medicine, Warwick Medical SchoolUniversity of WarwickCoventryUK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM)University Hospitals Coventry and Warwickshire NHS TrustCoventryUK
| | - Ian P. Salt
- School of Molecular Biosciences, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Dilys J. Freeman
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Jason M. R. Gill
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - G. Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
- Department of Pharmacology, Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Arild C. Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
| | - Victor A. Zammit
- Translational and Experimental Medicine, Warwick Medical SchoolUniversity of WarwickCoventryUK
| |
Collapse
|
7
|
Elkin RG, Harvatine KJ. A review of recent studies on the enrichment of eggs and poultry meat with omega-3 polyunsaturated fatty acids: novel findings and unanswered questions. Poult Sci 2023; 102:102938. [PMID: 37572619 PMCID: PMC10428063 DOI: 10.1016/j.psj.2023.102938] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/10/2023] [Accepted: 07/15/2023] [Indexed: 08/14/2023] Open
Abstract
Studies from our laboratory over the past decade have yielded new information with regard to the dietary enrichment of eggs and poultry meat with omega-3 (n-3) polyunsaturated fatty acids (PUFA) but have also generated a number of unanswered questions. In this review, we summarize the novel findings from this work, identify knowledge gaps, and offer possible explanations for some perplexing observations. Specifically discussed are: 1) Why feeding laying hens and broilers an oil rich in stearidonic acid (SDA; 18:4 n-3), which theoretically bypasses the putative rate-limiting step in the hepatic n-3 PUFA biosynthetic pathway, does not enrich egg yolks and tissues with very long-chain (VLC; ≥20 C) n-3 PUFA to the same degree as obtained by feeding birds oils rich in preformed VLC n-3 PUFA; 2) Why in hens fed an SDA-rich oil, SDA fails to accumulate in egg yolk but is readily incorporated into adipose tissue; 3) How oils rich in oleic acid (OA; 18:1 n-9), when co-fed with various sources of n-3 PUFA, attenuates egg and tissue n-3 PUFA contents or rescues egg production when co-fed with a level of docosahexaenoic acid (DHA; 22:6 n-3) that causes severe hypotriglyceridemia; and 4) Why the efficiency of VLC n-3 PUFA deposition into eggs and poultry meat is inversely related to the dietary content of α-linolenic acid (ALA; 18:3 n-3), SDA, or DHA.
Collapse
Affiliation(s)
- Robert G Elkin
- Department of Animal Science, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Kevin J Harvatine
- Department of Animal Science, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
8
|
Kwon EB, Moon DO, Oh ES, Song YN, Park JY, Ryu HW, Kim DY, Chin YW, Lee HS, Lee SU, Kim MO. Garcinia mangostana Suppresses Triacylglycerol Synthesis in Hepatocytes and Enterocytes. J Med Food 2023. [PMID: 37566462 DOI: 10.1089/jmf.2023.k.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023] Open
Abstract
Regulation of diacylglycerol acyltransferase (DGAT) and pancreatic lipase (PL) activities is important in the treatment of triacylglycerol (TG)-related metabolic diseases. Garcinia mangostana, also known as mangosteen, is a traditional medicine ingredient used in the treatment of inflammation in Southeast Asia. In this study, The ethanolic extract of G. mangostana peel inhibited human recombinant DGAT1 and DGAT2, and PL enzyme activities in vitro. The inhibitory activity of DGAT1 and DGAT2 enzymes of four representative bioactive substances in mangosteen was confirmed. In addition, G. mangostana was confirmed to suppress the serum TG levels in C57 mice by inhibiting the absorption and synthesis of TG in the gastrointestinal tract. Through this study, it was revealed that G. mangostana extract could be useful for the prevention and amelioration of TG-related metabolic diseases such as obesity and fatty liver.
Collapse
Affiliation(s)
- Eun-Bin Kwon
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Dong-Oh Moon
- Department of Biology Education, Daegu University, Gyeongsan, Korea
| | - Eun Sol Oh
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
- Departments of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Yu Na Song
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
- Departments of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Ji-Yoon Park
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
- Department of Anatomy & Cell Biology, Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Hyung Won Ryu
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Doo-Young Kim
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Korea
| | - Hyun-Sun Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Su Ui Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Mun-Ock Kim
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| |
Collapse
|
9
|
Yenilmez B, Harney S, DiMarzio C, Kelly M, Min K, Echeverria D, Bramato BM, Jackson SO, Reddig K, Kim JK, Khvorova A, Czech MP. Dual targeting of hepatocyte DGAT2 and stellate cell FASN alleviates nonalcoholic steatohepatitis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547848. [PMID: 37461560 PMCID: PMC10350091 DOI: 10.1101/2023.07.05.547848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a malady of multiple cell types associated with hepatocyte triglyceride (TG) accumulation, macrophage inflammation, and stellate cell-induced fibrosis, with no approved therapeutics yet available. Here, we report that stellate cell fatty acid synthase (FASN) in de novo lipogenesis drives the autophagic flux that is required for stellate cell activation and fibrotic collagen production. Further, we employ a dual targeting approach to NASH that selectively depletes collagen through selective stellate cell knockout of FASN (using AAV9-LRAT Cre in FASNfl/fl mice), while lowering hepatocyte triglyceride by depleting DGAT2 with a GalNac-conjugated, fully chemically modified siRNA. DGAT2 silencing in hepatocytes alone or in combination with stellate cell FASNKO reduced liver TG accumulation in a choline-deficient NASH mouse model, while FASNKO in hepatocytes alone (using AAV8-TBG Cre in FASNfl/fl mice) did not. Neither hepatocyte DGAT2 silencing alone nor FASNKO in stellate cells alone decreased fibrosis (total collagen), while loss of both DGAT2 plus FASN caused a highly significant attenuation of NASH. These data establish proof of concept that dual targeting of DGAT2 plus FASN alleviates NASH progression in mice far greater than targeting either gene product alone.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shauna Harney
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dimas Echeverria
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Brianna M. Bramato
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Samuel O. Jackson
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Keith Reddig
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
10
|
Laupsa-Borge J, Grytten E, Bohov P, Bjørndal B, Strand E, Skorve J, Nordrehaug JE, Berge RK, Rostrup E, Mellgren G, Dankel SN, Nygård OK. Sex-specific responses in glucose-insulin homeostasis and lipoprotein-lipid components after high-dose supplementation with marine n-3 PUFAs in abdominal obesity: a randomized double-blind crossover study. Front Nutr 2023; 10:1020678. [PMID: 37404855 PMCID: PMC10315503 DOI: 10.3389/fnut.2023.1020678] [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: 08/16/2022] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Background Clinical studies on effects of marine-derived omega-3 (n-3) polyunsaturated fatty acids (PUFAs), mainly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and the plant-derived omega-6 (n-6) PUFA linoleic acid (LA) on lipoprotein-lipid components and glucose-insulin homeostasis have shown conflicting results, which may partly be explained by differential responses in females and males. However, we have lacked data on sexual dimorphism in the response of cardiometabolic risk markers following increased consumption of n-3 or n-6 PUFAs. Objective To explore sex-specific responses after n-3 (EPA + DHA) or n-6 (LA) PUFA supplementation on circulating lipoprotein subfractions, standard lipids, apolipoproteins, fatty acids in red blood cell membranes, and markers of glycemic control/insulin sensitivity among people with abdominal obesity. Methods This was a randomized double-blind crossover study with two 7-week intervention periods separated by a 9-week washout phase. Females (n = 16) were supplemented with 3 g/d of EPA + DHA (fish oil) or 15 g/d of LA (safflower oil), while males (n = 23) received a dose of 4 g/d of EPA + DHA or 20 g/d of LA. In fasting blood samples, we measured lipoprotein particle subclasses, standard lipids, apolipoproteins, fatty acid profiles, and markers of glycemic control/insulin sensitivity. Results The between-sex difference in relative change scores was significant after n-3 for total high-density lipoproteins (females/males: -11%*/-3.3%, p = 0.036; *: significant within-sex change), high-density lipoprotein particle size (+2.1%*/-0.1%, p = 0.045), and arachidonic acid (-8.3%*/-12%*, p = 0.012), and after n-6 for total (+37%*/+2.1%, p = 0.041) and small very-low-density lipoproteins (+97%*/+14%, p = 0.021), and lipoprotein (a) (-16%*/+0.1%, p = 0.028). Circulating markers of glucose-insulin homeostasis differed significantly after n-3 for glucose (females/males: -2.1%/+3.9%*, p = 0.029), insulin (-31%*/+16%, p < 0.001), insulin C-peptide (-12%*/+13%*, p = 0.001), homeostasis model assessment of insulin resistance index 2 (-12%*/+14%*, p = 0.001) and insulin sensitivity index 2 (+14%*/-12%*, p = 0.001), and quantitative insulin sensitivity check index (+4.9%*/-3.4%*, p < 0.001). Conclusion We found sex-specific responses after high-dose n-3 (but not n-6) supplementation in circulating markers of glycemic control/insulin sensitivity, which improved in females but worsened in males. This may partly be related to the sex differences we observed in several components of the lipoprotein-lipid profile following the n-3 intervention. Clinical trial registration https://clinicaltrials.gov/, identifier [NCT02647333].
Collapse
Affiliation(s)
- Johnny Laupsa-Borge
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elise Grytten
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jon Skorve
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Rolf K. Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Espen Rostrup
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon N. Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ottar K. Nygård
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
11
|
Begemann K, Heyde I, Witt P, Inderhees J, Leinweber B, Koch CE, Jöhren O, Oelkrug R, Liskiewicz A, Müller TD, Oster H. Rest phase snacking increases energy resorption and weight gain in male mice. Mol Metab 2023; 69:101691. [PMID: 36746332 PMCID: PMC9950950 DOI: 10.1016/j.molmet.2023.101691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Snacking, i.e., the intake of small amounts of palatable food items, is a common behavior in modern societies, promoting overeating and obesity. Shifting food intake into the daily rest phase disrupts circadian rhythms and is also known to stimulate weight gain. We therefore hypothesized that chronic snacking in the inactive phase may promote body weight gain and that this effect is based on disruption of circadian clocks. METHODS Male mice were fed a daily chocolate snack either during their rest or their active phase and body weight development and metabolic parameters were investigated. Snacking experiments were repeated in constant darkness and in clock-deficient mutant mice to examine the role of external and internal time cues in mediating the metabolic effects of snacking. RESULTS Chronic snacking in the rest phase increased body weight gain and disrupted metabolic circadian rhythms in energy expenditure, body temperature, and locomotor activity. Additionally, these rest phase snacking mice assimilated more energy during the inactive phase. Body weight remained increased in rest phase snacking wildtype mice in constant darkness as well as in clock-deficient mutant mice under a regular light-dark cycle compared to mice snacking in the active phase. Weight gain effects were abolished in clock-deficient mice in constant darkness. CONCLUSIONS Our data suggest that mistimed snacking increases energy resorption and promotes body weight gain. This effect requires a functional circadian clock at least under constant darkness conditions.
Collapse
Affiliation(s)
- Kimberly Begemann
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| | - Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Pia Witt
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Julica Inderhees
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Bioanalytic Core Facility, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Brinja Leinweber
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Christiane E. Koch
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Olaf Jöhren
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Bioanalytic Core Facility, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Rebecca Oelkrug
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany,Institute for Endocrinology and Diabetes, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany,Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice 40-752, Poland
| | - Timo D. Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; Center of Brain, Behavior, and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
12
|
Mak KM, Wu C, Cheng CP. Lipid droplets, the Holy Grail of hepatic stellate cells: In health and hepatic fibrosis. Anat Rec (Hoboken) 2022; 306:983-1010. [PMID: 36516055 DOI: 10.1002/ar.25138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
Lipid droplets (LDs) are distinct morphological markers of hepatic stellate cells (HSCs). They are composed of a core of predominantly retinyl esters and triacylglycerols surrounded by a phospholipid layer; the latter harbors perilipins 2, 3, and 5, which help control LD lipolysis. Electron microscopy distinguishes between Types I and II LDs. Type I LDs are surrounded by acid phosphatase-positive lysosomes, which likely digest LDs. LD count and retinoid concentration are modulated by vitamin A intake. Alcohol consumption depletes hepatic retinoids and HSC LDs, with concomitant transformation of HSCs to fibrogenic myofibroblast-like cells. LD loss and accompanying HSC activation occur in HSC cell culture models. Loss of LDs is a consequence of and not a prerequisite for HSC activation. LDs are endowed with enzymes for synthesizing retinyl esters and triacylglycerols as well as neutral lipases and lysosomal acid lipase for breaking down LDs. HSCs have two distinct metabolic LD pools: an "original" pool in quiescent HSCs and a "new" pool emerging in HSC activation; this two-pool model provides a platform for analyzing LD dynamics in HSC activation. Besides lipolysis, LDs are degraded by lipophagy; however, the coordination between and relative contributions of these two pathways to LD removal are unclear. While induction of autophagy accelerates LD loss in quiescent HSCs and promotes HSC activation, blocking autophagy impairs LD degradation and inhibits HSC activation and fibrosis. This article is a critique of five decades of investigations into the morphology, molecular structure, synthesis, and degradation of LDs associated with HSC activation and fibrosis.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Catherine Wu
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christopher P Cheng
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
13
|
Elkin RG, El-Zenary AS, Bomberger R, Haile AB, Weaver EA, Ramachandran R, Harvatine KJ. Feeding laying hens docosa hexaenoic acid-rich microalgae oil at 40 g/kg diet causes hypotriglyceridemia, depresses egg production, and attenuates expression of key genes affecting hepatic triglyceride synthesis and secretion, but is rescued by dietary co-supplementation of high-oleic sunflower oil. Poult Sci 2022; 102:102318. [PMID: 36525748 PMCID: PMC9758496 DOI: 10.1016/j.psj.2022.102318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
The primary goal of this study was to investigate the effect of feeding White Leghorn hens graded levels of a docosahexaenoic acid (DHA)-rich microalgae oil (MAO) on productive performance and enrichment of eggs with very long-chain (VLC) omega-3 (n-3) polyunsaturated fatty acids (PUFA). Forty-nine-week-old hens (8 per diet) were fed the following diets for 28 d: 1) A corn-soybean meal-based diet with no supplemental oil (CON); 2) CON + 10 g/kg MAO; 3) CON + 20 g/kg MAO; 4) CON + 30 g/kg MAO; 5) CON + 40 g/kg MAO; 6) CON + 40 g/kg MAO + 20 g/kg high-oleic sunflower oil (HOSO); and 7) CON + 40 g/kg MAO + 40 g/kg HOSO. Diets 6 and 7 were included because we previously reported that co-feeding high-oleic acid oils with n-3 PUFA-containing oils attenuated egg yolk n-3 PUFA contents vs. feeding hens the n-3 oils alone. All data were collected on an individual hen basis. Egg VLC n-3 PUFA enrichment plateaued, in terms of statistical significance, at the 30 g/kg MAO level (266 mg/yolk). Hens fed 40 g/kg MAO had greatly attenuated measures of hen performance, marked liver enlargement, an altered ovarian follicle hierarchy, greatly lowered circulating triglyceride levels, and depressed hepatic expression of key genes involved in triglyceride synthesis and secretion. As compared to hens fed 40 g/kg MAO alone, feeding hens 40 g/kg MAO co-supplemented with HOSO (Diets 6 and 7) restored egg production, ovarian morphology, and all other measures of hen productive performance to CON levels, elevated plasma triglyceride levels, prevented liver enlargement, and increased the hepatic expression of key genes involved in triglyceride synthesis and secretion. In conclusion, MAO can greatly enrich hens' eggs with VLC n-3 PUFA, but its recommended dietary inclusion should not exceed 20 g/kg. This would allow for near-maximal yolk VLC n-3 PUFA enrichment without impairing hen productive performance, altering the ovarian follicle hierarchy or, based on the work of others, presumably imparting off-flavors in the egg.
Collapse
|
14
|
Yenilmez B, Kelly M, Zhang GF, Wetoska N, Ilkayeva OR, Min K, Rowland L, DiMarzio C, He W, Raymond N, Lifshitz L, Pan M, Han X, Xie J, Friedline RH, Kim JK, Gao G, Herman MA, Newgard CB, Czech MP. Paradoxical activation of transcription factor SREBP1c and de novo lipogenesis by hepatocyte-selective ATP-citrate lyase depletion in obese mice. J Biol Chem 2022; 298:102401. [PMID: 35988648 PMCID: PMC9490592 DOI: 10.1016/j.jbc.2022.102401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 01/26/2023] Open
Abstract
Hepatic steatosis associated with high-fat diet, obesity, and type 2 diabetes is thought to be the major driver of severe liver inflammation, fibrosis, and cirrhosis. Cytosolic acetyl CoA (AcCoA), a central metabolite and substrate for de novo lipogenesis (DNL), is produced from citrate by ATP-citrate lyase (ACLY) and from acetate through AcCoA synthase short chain family member 2 (ACSS2). However, the relative contributions of these two enzymes to hepatic AcCoA pools and DNL rates in response to high-fat feeding are unknown. We report here that hepatocyte-selective depletion of either ACSS2 or ACLY caused similar 50% decreases in liver AcCoA levels in obese mice, showing that both pathways contribute to the generation of this DNL substrate. Unexpectedly however, the hepatocyte ACLY depletion in obese mice paradoxically increased total DNL flux measured by D2O incorporation into palmitate, whereas in contrast, ACSS2 depletion had no effect. The increase in liver DNL upon ACLY depletion was associated with increased expression of nuclear sterol regulatory element-binding protein 1c and of its target DNL enzymes. This upregulated DNL enzyme expression explains the increased rate of palmitate synthesis in ACLY-depleted livers. Furthermore, this increased flux through DNL may also contribute to the observed depletion of AcCoA levels because of its increased conversion to malonyl CoA and palmitate. Together, these data indicate that in fat diet-fed obese mice, hepatic DNL is not limited by its immediate substrates AcCoA or malonyl CoA but rather by activities of DNL enzymes.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Leslie Rowland
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Wentao He
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Naideline Raymond
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Lawrence Lifshitz
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Meixia Pan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jun Xie
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mark A Herman
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA.
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
15
|
Winichayakul S, Curran A, Moraga R, Cookson R, Xue H, Crowther T, Roldan M, Bryan G, Roberts N. An alternative angiosperm DGAT1 topology and potential motifs in the N-terminus. FRONTIERS IN PLANT SCIENCE 2022; 13:951389. [PMID: 36186081 PMCID: PMC9523541 DOI: 10.3389/fpls.2022.951389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/05/2022] [Indexed: 06/16/2023]
Abstract
The highly variable cytoplasmic N-terminus of the plant diacylglycerol acyltransferase 1 (DGAT1) has been shown to have roles in oligomerization as well as allostery; however, the biological significance of the variation within this region is not understood. Comparing the coding sequences over the variable N-termini revealed the Poaceae DGAT1s contain relatively high GC compositional gradients as well as numerous direct and inverted repeats in this region. Using a variety of reciprocal chimeric DGAT1s from angiosperms we show that related N-termini had similar effects (positive or negative) on the accumulation of the recombinant protein in Saccharomyces cerevisiae. When expressed in Camelina sativa seeds the recombinant proteins of specific chimeras elevated total lipid content of the seeds as well as increased seed size. In addition, we combine N- and C-terminal as well as internal tags with high pH membrane reformation, protease protection and differential permeabilization. This led us to conclude the C-terminus is in the ER lumen; this contradicts earlier reports of the cytoplasmic location of plant DGAT1 C-termini.
Collapse
Affiliation(s)
- Somrutai Winichayakul
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Amy Curran
- ZeaKal Inc., San Diego, CA, United States
| | - Roger Moraga
- Bioinformatics and Statistics, AgResearch Ltd., Palmerston North, New Zealand
| | - Ruth Cookson
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Hong Xue
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Tracey Crowther
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Marissa Roldan
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Greg Bryan
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
- ZeaKal Inc., San Diego, CA, United States
| | - Nick Roberts
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
- ZeaKal Inc., San Diego, CA, United States
| |
Collapse
|
16
|
Chen X, Luo J, Yang L, Guo Y, Fan Y, Liu J, Sun J, Zhang Y, Jiang Q, Chen T, Xi Q. miR-143-Mediated Responses to Betaine Supplement Repress Lipogenesis and Hepatic Gluconeogenesis by Targeting MAT1a and MAPK11. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7981-7992. [PMID: 35734958 DOI: 10.1021/acs.jafc.2c02940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liver as the central organ is responsible for lipogenesis, gluconeogenesis and one-carbon metabolism. Methyl donors (e.g., betaine) modulate metabolic homeostasis and gene regulation through one-carbon metabolism. MiR-143 regulates DNA methylation by targeting DNMT3A, thereby suggesting that this miRNA participates in one-carbon metabolic pathways. However, the effect and mechanism that regulate glucose and lipid metabolism via the methyl group metabolism pathway remain elusive. In this study, we found that a betaine supplement and miR-143 KO significantly promoted lipolysis and glucose utilization and repressed lipogenesis and gluconeogenesis through enhancing energy consumption and thermogenesis, repressing GPNMB and targeting MAPK11, respectively. We further explored the relationship between miR-143 and a methyl donor (betaine) and the miR-143-mediated responses to the betaine supplement regulating the mechanism of the glucose and lipid metabolism. The results showed that betaine significantly down-regulated the expression of miR-143 that subsequently increased SAM levels in the liver by targeting MAT1a. In brief, the regulations of glucose and lipid metabolism are related to the miR-143-regulation of one-carbon units, and the relationship between betaine and miR-143 in the methionine cycle is a typical yin-yang type of regulation. Thus, betaine and miR-143 function together as key regulators and biomarkers for preventing and diagnosing metabolic diseases such as fatty liver disease, obesity, and diabetes.
Collapse
Affiliation(s)
- Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Lekai Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yue Guo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yaotian Fan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Jie Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| |
Collapse
|
17
|
Liesa M. The goodies of chelated fat: iron-regulated lipid droplet biogenesis precedes and preserves mitophagy. EMBO J 2022; 41:e111238. [PMID: 35451092 PMCID: PMC9108591 DOI: 10.15252/embj.2022111238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
The role of iron-regulated mitophagy in lipid metabolism is unclear. Recent work by Long, Sanchez-Martinez et al (2022) shows that iron chelation induces a primary change in lipid metabolism that promotes fat accumulation and precedes mitophagy.
Collapse
Affiliation(s)
- Marc Liesa
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain.,Department of Medicine, Endocrinology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
18
|
Velázquez AM, Bentanachs R, Sala‐Vila A, Lázaro I, Rodríguez‐Morató J, Sánchez RM, Alegret M, Roglans N, Laguna JC. ChREBP-driven DNL and PNPLA3 Expression Induced by Liquid Fructose are Essential in the Production of Fatty Liver and Hypertriglyceridemia in a High-Fat Diet-Fed Rat Model. Mol Nutr Food Res 2022; 66:e2101115. [PMID: 35124887 PMCID: PMC9286604 DOI: 10.1002/mnfr.202101115] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Indexed: 11/25/2022]
Abstract
SCOPE The aim of this study is to delineate the contribution of dietary saturated fatty acids (FA) versus liquid fructose to fatty liver and hypertriglyceridemia. METHODS AND RESULTS Three groups of female rats are maintained for 3 months in standard chow (CT); High-fat diet (46.9% of fat-derived calories, rich in palmitic and stearic FA, HFD); and HFD with 10% w/v fructose in drinking water (HFHFr). Zoometric parameters, plasma biochemistry, and liver Oil-Red O (ORO) staining, lipidomics, and expression of proteins involved in FA metabolism are analyzed. Both diets increase ingested calories without modifying body weight. Only the HFHFr diet increases liver triglycerides (x11.0), with hypertriglyceridemia (x1.7) and reduces FA β-oxidation (x0.7), and increases liver FA markers of DNL (de novo lipogenesis). Whereas HFD livers show a high content of ceramides, HFHFr samples show unchanged ceramides, and an increase in diacylglycerols. Only the HFHFr diet leads to a marked increase in the expression of enzymes involved in DNL and triglyceride metabolism, such as carbohydrate response element binding protein β (ChREBPβ, x3.2), a transcription factor that regulates DNL, and patatin-like phospholipase domain-containing 3 (PNPLA3, x2.6), a lipase that mobilizes stored triglycerides for VLDL secretion. CONCLUSION The addition of liquid-fructose to dietary FA is determinant in liver steatosis and hypertriglyceridemia production, through increased DNL and PNPLA3 expression, and reduced FA catabolism.
Collapse
Affiliation(s)
- Ana Magdalena Velázquez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
| | - Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
| | - Aleix Sala‐Vila
- IMIM‐Hospital del Mar Medical Research InstituteBarcelona08003Spain
- Barcelonaβeta Brain Research CenterPasqual Maragall FoundationBarcelona08005Spain
| | - Iolanda Lázaro
- IMIM‐Hospital del Mar Medical Research InstituteBarcelona08003Spain
| | - Jose Rodríguez‐Morató
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- IMIM‐Hospital del Mar Medical Research InstituteBarcelona08003Spain
- Department of Experimental and Health SciencesUniversitat Pompeu Fabra (CEXS‐UPF)Barcelona08003Spain
| | - Rosa M. Sánchez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
- Institute of BiomedicineUniversity of BarcelonaBarcelona08028Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
- Institute of BiomedicineUniversity of BarcelonaBarcelona08028Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
- Institute of BiomedicineUniversity of BarcelonaBarcelona08028Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food ScienceUniversity of BarcelonaAvda Joan XXIII 27–31Barcelona08028Spain
- Institute of BiomedicineUniversity of BarcelonaBarcelona08028Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| |
Collapse
|
19
|
Amin NB, Darekar A, Anstee QM, Wong VWS, Tacke F, Vourvahis M, Lee DS, Charlton M, Alkhouri N, Nakajima A, Yunis C. Efficacy and safety of an orally administered DGAT2 inhibitor alone or coadministered with a liver-targeted ACC inhibitor in adults with non-alcoholic steatohepatitis (NASH): rationale and design of the phase II, dose-ranging, dose-finding, randomised, placebo-controlled MIRNA (Metabolic Interventions to Resolve NASH with fibrosis) study. BMJ Open 2022; 12:e056159. [PMID: 35354614 PMCID: PMC8968568 DOI: 10.1136/bmjopen-2021-056159] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Small molecule inhibitors of the terminal step in intrahepatic triglyceride synthesis (diacylglycerol acyltransferase 2 inhibitor (DGAT2i, PF-06865571, ervogastat)) and upstream blockade of de novo lipogenesis via acetyl-coenzyme A carboxylase inhibitor (ACCi, PF-05221304, clesacostat) showed promise in reducing hepatic steatosis in early clinical trials. This study assesses efficacy and safety of these metabolic interventions to resolve non-alcoholic steatohepatitis (NASH) with fibrosis. METHODS AND ANALYSIS This phase II, randomised, dose-ranging, dose-finding study evaluates DGAT2i 25-300 mg two times per day (BID) or 150-300 mg once a day, DGAT2i 150-300 mg BID+ACCi 5-10 mg BID coadministration or matching placebo in a planned 450 adults with biopsy-confirmed NASH and liver fibrosis stages 2-3 from approximately 220 sites in 11 countries across North America, Europe and Asia. A triage approach including double-confirmation via non-invasive markers is included prior to screening/baseline liver biopsy. On confirmation of histological diagnosis, participants enter a ≥6-week run-in period, then a 48-week double-blind, double-dummy dosing period. The primary endpoint is the proportion of participants achieving histological NASH resolution without worsening fibrosis, ≥1 stage improvement in fibrosis without worsening NASH, or both, assessed by central pathologists. Other endpoints include assessment of hepatic steatosis (imaging substudy), overall safety and tolerability, and evaluation of blood-based biomarkers and quantitative ultrasound parameters over time. ETHICS AND DISSEMINATION Metabolic Interventions to Resolve NASH with fibrosis (MIRNA) is conducted in accordance with the Declaration of Helsinki and Council for International Organisations of Medical Sciences (CIOMS) International Ethical Guidelines, International Council on Harmonisation Good Clinical Practice guidelines, applicable laws and regulations, including privacy laws. Local independent review board/ethics committees (IRB/ECs) review/approve the protocol, any amendments, informed consent and other forms. Participants provide written informed consent. Details of all IRB/ECs, as well as results, will be published in a peer-reviewed journal and publicly disclosed through ClinicalTrials.gov, EudraCT, and/or www.pfizer.com and other public registries as per applicable local laws/regulations. TRIAL REGISTRATION NUMBER NCT04321031.
Collapse
Affiliation(s)
- Neeta B Amin
- Pfizer Global Product Development, Cambridge, Massachusetts, USA
| | | | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Frank Tacke
- Charité-Universitätsmedizin Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | | | - Douglas S Lee
- Pfizer Global Product Development, Groton, Connecticut, USA
| | - Michael Charlton
- Transplant Institute, Center for Liver Diseases, University of Chicago Biological Sciences, Chicago, Illinois, USA
| | | | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Carla Yunis
- Pfizer Global Product Development, New York, New York, USA
| |
Collapse
|
20
|
Yenilmez B, Wetoska N, Kelly M, Echeverria D, Min K, Lifshitz L, Alterman JF, Hassler MR, Hildebrand S, DiMarzio C, McHugh N, Vangjeli L, Sousa J, Pan M, Han X, Brehm MA, Khvorova A, Czech MP. An RNAi therapeutic targeting hepatic DGAT2 in a genetically obese mouse model of nonalcoholic steatohepatitis. Mol Ther 2022; 30:1329-1342. [PMID: 34774753 PMCID: PMC8899521 DOI: 10.1016/j.ymthe.2021.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/31/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe liver disorder characterized by triglyceride accumulation, severe inflammation, and fibrosis. With the recent increase in prevalence, NASH is now the leading cause of liver transplant, with no approved therapeutics available. Although the exact molecular mechanism of NASH progression is not well understood, a widely held hypothesis is that fat accumulation is the primary driver of the disease. Therefore, diacylglycerol O-acyltransferase 2 (DGAT2), a key enzyme in triglyceride synthesis, has been explored as a NASH target. RNAi-based therapeutics is revolutionizing the treatment of liver diseases, with recent chemical advances supporting long-term gene silencing with single subcutaneous administration. Here, we identified a hyper-functional, fully chemically stabilized GalNAc-conjugated small interfering RNA (siRNA) targeting DGAT2 (Dgat2-1473) that, upon injection, elicits up to 3 months of DGAT2 silencing (>80%-90%, p < 0.0001) in wild-type and NSG-PiZ "humanized" mice. Using an obesity-driven mouse model of NASH (ob/ob-GAN), Dgat2-1473 administration prevents and reverses triglyceride accumulation (>85%, p < 0.0001) without increased accumulation of diglycerides, resulting in significant improvement of the fatty liver phenotype. However, surprisingly, the reduction in liver fat did not translate into a similar impact on inflammation and fibrosis. Thus, while Dgat2-1473 is a practical, long-lasting silencing agent for potential therapeutic attenuation of liver steatosis, combinatorial targeting of a second pathway may be necessary for therapeutic efficacy against NASH.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Dimas Echeverria
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Lawrence Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Julia F Alterman
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Matthew R Hassler
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Samuel Hildebrand
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Nicholas McHugh
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Lorenc Vangjeli
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Jacquelyn Sousa
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Meixia Pan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xianlin Han
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA.
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA.
| |
Collapse
|
21
|
Cisbani G, Koppel A, Metherel AH, Smith ME, Aji KN, Andreazza AC, Mizrahi R, Bazinet RP. Serum lipid analysis and isotopic enrichment is suggestive of greater lipogenesis in young long-term cannabis users: A secondary analysis of a case-control study. Lipids 2022; 57:125-140. [PMID: 35075659 PMCID: PMC8923992 DOI: 10.1002/lipd.12336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023]
Abstract
Cannabis is now legal in many countries and while numerous studies have reported on its impact on cognition and appetite regulation, none have examined fatty acid metabolism in young cannabis users. We conducted an exploratory analysis to evaluate cannabis impact on fatty acid metabolism in cannabis users (n = 21) and non-cannabis users (n = 16). Serum levels of some saturated and monounsaturated fatty acids, including palmitic, palmitoleic, and oleic acids were higher in cannabis users compared to nonusers. As palmitic acid can be derived from diet or lipogenesis from sugars, we evaluated lipogenesis using a de novo lipogenesis index (palmitate/linoleic acid) and carbon-specific isotope analysis, which allows for the determination of fatty acid 13 C signature. The significantly higher de novo lipogenesis index in the cannabis users group along with a more enriched 13 C signature of palmitic acid suggested an increase in lipogenesis. In addition, while serum glucose concentration did not differ between groups, pyruvate and lactate were lower in the cannabis user group, with pyruvate negatively correlating with palmitic acid. Furthermore, the endocannabinoid 2-arachidonoylglycerol was elevated in cannabis users and could contribute to lipogenesis by activating the cannabinoid receptor 1. Because palmitic acid has been suggested to increase inflammation, we measured peripheral cytokines and observed no changes in inflammatory cytokines. Finally, an anti-inflammatory metabolite of palmitic acid, palmitoylethanolamide was elevated in cannabis users. Our results suggest that lipogenic activity is increased in cannabis users; however, future studies, including prospective studies that control dietary intake are required.
Collapse
Affiliation(s)
- Giulia Cisbani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Alex Koppel
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Adam H. Metherel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Mackenzie E. Smith
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Kankana N. Aji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Ana C. Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Romina Mizrahi
- Department of Psychiatry, McGill University, Montreal, Canada,Douglas Research Center, Montreal, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| | - Richard P. Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| |
Collapse
|
22
|
Nagarajan SR, Cross E, Sanna F, Hodson L. Dysregulation of hepatic metabolism with obesity: factors influencing glucose and lipid metabolism. Proc Nutr Soc 2022; 81:1-11. [PMID: 34726148 DOI: 10.1017/s0029665121003761] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The liver is a key metabolic organ that undertakes a multitude of physiological processes over the course of a day, including intrahepatic lipid and glucose metabolism which plays a key role in the regulation of systemic lipid and glucose concentrations. It serves as an intermediary organ between exogenous (dietary) and endogenous energy supply to extrahepatic organs. Thus, perturbations in hepatic metabolism can impact widely on metabolic disease risk. For example, the accumulation of intra-hepatocellular TAG (IHTG), for which adiposity is almost invariably a causative factor may result in dysregulation of metabolic pathways. Accumulation of IHTG is likely due to an imbalance between fatty acid delivery, synthesis and removal (via oxidation or export as TAG) from the liver; insulin plays a key role in all of these processes.
Collapse
Affiliation(s)
- S R Nagarajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - E Cross
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - F Sanna
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - L Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Green CJ, Marjot T, Walsby-Tickle J, Charlton C, Cornfield T, Westcott F, Pinnick KE, Moolla A, Hazlehurst JM, McCullagh J, Tomlinson JW, Hodson L. Metformin maintains intrahepatic triglyceride content through increased hepatic de novo lipogenesis. Eur J Endocrinol 2022; 186:367-377. [PMID: 35038311 PMCID: PMC8859923 DOI: 10.1530/eje-21-0850] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/17/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVE Metformin is a first-line pharmacotherapy in the treatment of type 2 diabetes, a condition closely associated with non-alcoholic fatty liver disease (NAFLD). Although metformin promotes weight loss and improves insulin sensitivity, its effect on intrahepatic triglyceride (IHTG) remains unclear. We investigated the effect of metformin on IHTG, hepatic de novo lipogenesis (DNL), and fatty acid (FA) oxidation in vivo in humans. DESIGN AND METHODS Metabolic investigations, using stable-isotope tracers, were performed in ten insulin-resistant, overweight/obese human participants with NAFLD who were treatment naïve before and after 12 weeks of metformin treatment. The effect of metformin on markers of s.c. adipose tissue FA metabolism and function, along with the plasma metabolome, was investigated. RESULTS Twelve weeks of treatment with metformin resulted in a significant reduction in body weight and improved insulin sensitivity, but IHTG content and FA oxidation remained unchanged. Metformin treatment was associated with a significant decrease in VLDL-triglyceride (TG) concentrations and a significant increase in the relative contribution of DNL-derived FAs to VLDL-TG. There were subtle and relatively few changes in s.c. adipose tissue FA metabolism and the plasma metabolome with metformin treatment. CONCLUSIONS We demonstrate the mechanisms of action of metformin whereby it improves insulin sensitivity and promotes weight loss, without improvement in IHTG; these observations are partly explained through increased hepatic DNL and a lack of change in FA oxidation.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | - Catriona Charlton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Thomas Cornfield
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Felix Westcott
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Ahmad Moolla
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Jonathan M Hazlehurst
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, UK
| | - James McCullagh
- Chemistry Research Laboratory, University of Oxford, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, UK
- Correspondence should be addressed to L Hodson;
| |
Collapse
|
24
|
Lambie M, Bonomini M, Davies SJ, Accili D, Arduini A, Zammit V. Insulin resistance in cardiovascular disease, uremia, and peritoneal dialysis. Trends Endocrinol Metab 2021; 32:721-730. [PMID: 34266706 PMCID: PMC8893168 DOI: 10.1016/j.tem.2021.06.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/03/2021] [Accepted: 06/15/2021] [Indexed: 02/09/2023]
Abstract
Diabetic nephropathy is highly correlated with the occurrence of other complications of type 1 diabetes (T1D) and type 2 diabetes (T2D) mellitus; for example, hypertension with cardiovascular disease (CVD) being the most frequent cause of death in patients with end-stage renal disease and undergoing renal dialysis. Hyperglycemia and insulin resistance (IR) are responsible for the micro- and macrovascular complications of diabetes through different mechanisms. In particular, IR plays a key role in the etiology of atherosclerosis in both diabetic and non-diabetic patients. IR - exacerbated by organ-level selectivity - is more important than glycemic control per se in determining cardiovascular outcomes. This may be exacerbated by the fact that IR is organ and pathway specific due to the only selective loss of sensitivity to insulin action of specific pathways/processes. Therefore, it is counterintuitive that the use of peritoneal dialysis (PD) in (frequently) diabetic renal disease patients should involve their exposure to high daily doses of glucose peritoneally. In view of the controversy about the causal association between glucose load and CVD in PD patients, we discuss the role that selective IR may play in the progression of CVD in diabetic renal end-stage patients. In discussing these associations, we propose that reducing glucose exposure in PD solutions may be beneficial especially if coupled with strategies that address IR directly, and the avoidance of excessive use of insulin treatment in T2D.
Collapse
Affiliation(s)
- Mark Lambie
- Faculty of Medicine and Health Sciences, Keele University, Keele ST5 5BG, UK
| | - Mario Bonomini
- Department of Medicine, G. d'Annunzio University, Chieti 66100, Italy
| | - Simon J Davies
- Faculty of Medicine and Health Sciences, Keele University, Keele ST5 5BG, UK
| | - Domenico Accili
- Columbia University College of Physicians and Surgeons, Department of Medicine, New York, NY 10032, USA
| | | | - Victor Zammit
- Translational & Experimental Medicine, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
25
|
Akerele OA, Manning SJ, Dixon SE, Lacey AE, Cheema SK. Maternal omega-3 fatty acids maintained positive maternal lipids and cytokines profile, and improved pregnancy outcomes of C57BL/6 mice. J Nutr Biochem 2021; 98:108813. [PMID: 34242722 DOI: 10.1016/j.jnutbio.2021.108813] [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: 09/02/2020] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Abstract
Omega (n)-3 polyunsaturated fatty acids (PUFA) are known to regulate lipid metabolism and inflammation; however, the regulation of maternal lipid metabolism and cytokines profile by n-3 PUFA during different gestation stages, and its impact on fetal sustainability is not known. We investigated the effects of maternal diet varying in n-3 PUFA prior to, and during gestation, on maternal metabolic profile, placental inflammatory cytokines, and fetal outcomes. Female C57BL/6 mice were fed either a high, low or very low (9, 3 or 1% w/w n-3 PUFA) diet, containing n-6:n-3 PUFA of 5:1, 20:1 and 40:1, respectively for two weeks before mating, and throughout pregnancy. Animals were sacrificed prior to mating (NP), and during pregnancy at gestation days 6.5, 12.5 and 18.5. Maternal metabolic profile, placental cytokines and fetal outcomes were determined. Our results show for the first time that a maternal diet high in n-3 PUFA prevented dyslipidemia in NP mice, and maintained the expected lipid profile during pregnancy. However, females fed the very low n-3 PUFA diet became hyperlipidemic prior to pregnancy, and carried this profile into pregnancy. Maternal diet high in n-3 PUFA maintained maternal plasma progesterone and placental pro-inflammatory cytokines profile, and sustained fetal numbers throughout pregnancy, while females fed the low and very-low n-3 PUFA diet had fewer fetuses. Our findings demonstrate the importance of maternal diet before, and during pregnancy, to maintain maternal metabolic profile and fetus sustainability. These findings are important when designing dietary strategies to optimize maternal metabolism during pregnancy for successful pregnancy outcome.
Collapse
Affiliation(s)
- Olatunji Anthony Akerele
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sarah Jane Manning
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sarah Emily Dixon
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Amelia Estelle Lacey
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sukhinder Kaur Cheema
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada.
| |
Collapse
|
26
|
Eraky SM, Ramadan NM. Effects of omega-3 fatty acids and metformin combination on diabetic cardiomyopathy in rats through autophagic pathway. J Nutr Biochem 2021; 97:108798. [PMID: 34102283 DOI: 10.1016/j.jnutbio.2021.108798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/12/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022]
Abstract
Diabetic cardiomyopathy is a primary cause of increased morbidity and mortality in diabetics. Evidence has suggested a pivotal role for interrupted mitochondrial dynamics and quality control machinery in the onset and development of diabetic cardiomyopathy. Sequestosome 1 (SQSTM1) is a major reporter of selective autophagic activity. Other than controlling the expression of genes involved in mitochondrial biogenesis, recently peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) was reported to directly affect SQSTM1 gene expression. Calcineurin, a pivotal mediator of cardiac hypertrophy, has been also linked to enhanced expression of SQSTM1. This study aimed to test the cardioprotective effects of adding ω-3 polyunsaturated fatty acids (PUFAs) to metformin in a rat model of type 2 diabetes mellitus and to evaluate the molecular mechanisms underlying their effects on mitochondrial quality. Diabetes was induced in male Sprague Dawley rats by a high-fat diet for 6 weeks, followed by a low-dose streptozotocin (35 mg/kg). Diabetic rats were either treated with metformin (150 mg/kg/d), ω-3 PUFAs (300 mg/kg/d), or their combination in the same doses for further 8 weeks. Along with metabolic and pathological derangements, we report that correlating with electron microscopic evidence of mitochondrial degeneration, gene expression of the autophagic indicators SQSTM1, PGC-1α, and calcineurin were decreased in the hearts of diabetic rats. Independent of its anti-hyperglycemic effects, metformin successfully preserved mitochondrial integrity and upregulated myocardial PGC-1α, calcineurin, and SQSTM1 gene expression. ω-3 PUFAs possess synergistic cardioprotection when added to metformin, suggested by improvements in myocardial ultrastructure, autophagic activity, and SQSTM1 gene expression.
Collapse
Affiliation(s)
- Salma M Eraky
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Nehal M Ramadan
- Clinical Pharmacology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
27
|
Wang Q, Chen S, Li T, Yang Q, Liu J, Tao Y, Meng Y, Chen J, Feng X, Han Z, Shi M, Huang H, Han M, Jiang E. Critical Role of Lkb1 in the Maintenance of Alveolar Macrophage Self-Renewal and Immune Homeostasis. Front Immunol 2021; 12:629281. [PMID: 33968022 PMCID: PMC8100336 DOI: 10.3389/fimmu.2021.629281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/01/2021] [Indexed: 01/27/2023] Open
Abstract
Alveolar macrophages (AMs) are pivotal for maintaining lung immune homeostasis. We demonstrated that deletion of liver kinase b1 (Lkb1) in CD11c+ cells led to greatly reduced AM abundance in the lung due to the impaired self-renewal of AMs but not the impeded pre-AM differentiation. Mice with Lkb1-deficient AMs exhibited deteriorated diseases during airway Staphylococcus aureus (S. aureus) infection and allergic inflammation, with excessive accumulation of neutrophils and more severe lung pathology. Drug-mediated AM depletion experiments in wild type mice indicated a cause for AM reduction in aggravated diseases in Lkb1 conditional knockout mice. Transcriptomic sequencing also revealed that Lkb1 inhibited proinflammatory pathways, including IL-17 signaling and neutrophil migration, which might also contribute to the protective function of Lkb1 in AMs. We thus identified Lkb1 as a pivotal regulator that maintains the self-renewal and immune function of AMs.
Collapse
MESH Headings
- AMP-Activated Protein Kinases
- Animals
- Asthma/enzymology
- Asthma/genetics
- Asthma/immunology
- CD11 Antigens/genetics
- CD11 Antigens/metabolism
- Cell Self Renewal
- Disease Models, Animal
- Homeostasis
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Lung/enzymology
- Lung/immunology
- Lung/microbiology
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/microbiology
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Infiltration
- Pneumonia, Bacterial/enzymology
- Pneumonia, Bacterial/genetics
- Pneumonia, Bacterial/immunology
- Pneumonia, Bacterial/microbiology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction
- Staphylococcal Infections/enzymology
- Staphylococcal Infections/genetics
- Staphylococcal Infections/immunology
- Staphylococcal Infections/microbiology
- Transcriptome
- Mice
Collapse
Affiliation(s)
- Qianqian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Song Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tengda Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qiongmei Yang
- Department of Hematology, The First Affiliated Hospital of Kunming Medical University, Hematology Research Center of Yunnan Province, Kunming, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan Tao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuan Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jiadi Chen
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingxia Shi
- Department of Hematology, The First Affiliated Hospital of Kunming Medical University, Hematology Research Center of Yunnan Province, Kunming, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
28
|
Playing Jekyll and Hyde-The Dual Role of Lipids in Fatty Liver Disease. Cells 2020; 9:cells9102244. [PMID: 33036257 PMCID: PMC7601321 DOI: 10.3390/cells9102244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Lipids play Jekyll and Hyde in the liver. On the one hand, the lipid-laden status of hepatic stellate cells is a hallmark of healthy liver. On the other hand, the opposite is true for lipid-laden hepatocytes—they obstruct liver function. Neglected lipid accumulation in hepatocytes can progress into hepatic fibrosis, a condition induced by the activation of stellate cells. In their resting state, these cells store substantial quantities of fat-soluble vitamin A (retinyl esters) in large lipid droplets. During activation, these lipid organelles are gradually degraded. Hence, treatment of fatty liver disease is treading a tightrope—unsophisticated targeting of hepatic lipid accumulation might trigger problematic side effects on stellate cells. Therefore, it is of great importance to gain more insight into the highly dynamic lipid metabolism of hepatocytes and stellate cells in both quiescent and activated states. In this review, part of the special issue entitled “Cellular and Molecular Mechanisms underlying the Pathogenesis of Hepatic Fibrosis 2020”, we discuss current and highly versatile aspects of neutral lipid metabolism in the pathogenesis of non-alcoholic fatty liver disease (NAFLD).
Collapse
|
29
|
NASH, Fibrosis and Hepatocellular Carcinoma: Lipid Synthesis and Glutamine/Acetate Signaling. Int J Mol Sci 2020; 21:ijms21186799. [PMID: 32947972 PMCID: PMC7555727 DOI: 10.3390/ijms21186799] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
Primary liver cancer is predicted to be the sixth most common cancer and the fourth leading cause of cancer mortality worldwide. Recent studies identified nonalcoholic fatty liver disease (NAFLD) as the underlying cause in 13-38.2% of patients with hepatocellular carcinoma unrelated to viral hepatitis and alcohol abuse. NAFLD progresses to nonalcoholic steatohepatitis (NASH), which increases the risk for the development of liver fibrosis, cirrhosis, and hepatocellular carcinoma. NAFLD is characterized by dysregulation of lipid metabolism. In addition, lipid metabolism is effected not only in NAFLD, but also in a broad range of chronic liver diseases and tumor development. Cancer cells manipulate a variety of metabolic pathways, including lipid metabolism, in order to build up their own cellular components. Identifying tumor dependencies on lipid metabolism would provide options for novel targeting strategies. This review article summarizes the research evidence on metabolic reprogramming and focuses on lipid metabolism in NAFLD, NASH, fibrosis, and cancer. As alternative routes of acetyl-CoA production for fatty acid synthesis, topics on glutamine and acetate metabolism are included. Further, studies on small compound inhibitors targeting lipid metabolism are discussed. Understanding reprogramming strategies in liver diseases, as well as the visualization of the metabolism reprogramming networks, could uncover novel therapeutic options.
Collapse
|
30
|
Cañón-Beltrán K, Giraldo-Giraldo J, Cajas YN, Beltrán-Breña P, Hidalgo CO, Vásquez N, Leal CLV, Gutiérrez-Adán A, González EM, Rizos D. Inhibiting diacylglycerol acyltransferase-1 reduces lipid biosynthesis in bovine blastocysts produced in vitro. Theriogenology 2020; 158:267-276. [PMID: 33002770 DOI: 10.1016/j.theriogenology.2020.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 11/28/2022]
Abstract
Diacylglycerol acyltransferase-1 (DGAT1) is one of the DGAT enzymes that catalyzes the final step in the synthesis of triacylglycerol, which is a major component of the lipid droplets in embryos. Intracellular lipids accumulated in embryos produced in vitro have been associated with reduced cryotolerance and quality. The objective of the present study was to investigate the influence of DGAT1 inhibition on embryo development, quality, and post-vitrification survival, in addition to expression profiles of selected lipid metabolism-regulating and oxidative stress genes. Bovine cumulus-oocyte complexes were matured and fertilized in vitro and were cultured in synthetic oviduct fluid (SOF) supplemented with 5% fetal calf serum (FCS) alone (Control) or with 1, 5, 10 or 50 μM DGAT1 inhibitor (A922500®; D1, D5, D10, and D50, respectively) or 0.1% dimethyl sulfoxide (CDMSO: vehicle for DGAT1 inhibitor dilution) from 54 h post-insemination until Day 8 post insemination. No differences were found in blastocyst yield on days 7 and 8 in Control, CDMSO, D10, and D50 groups. Embryos cultured with 10 or 50 μM DGAT1 inhibitor had greater mitochondrial activity (P < 0.01), and increased number of cells (P < 0.05), while the cytoplasmic lipid content was reduced (P < 0.01), the latter associated with altered expression profiles of selected genes regulating lipid metabolism or genes related with oxidative stress (transcript abundance increased for SLC2A1 and SLC2A5 and decreased for DGAT1 and GPX1). Importantly, the survival rate of blastocysts produced with 10 μM DGAT1 was higher than that of Control, CDMSO and D50 groups at 72 h after vitrification and warming (73.8 vs 57.1, 55.9 and 56.1%, respectively, P < 0.001). In conclusion, inhibition of DGAT1 synthesis in bovine embryos produced in vitro abrogates the negative effect of FCS by decreasing their lipid content, increasing mitochondria activity and improving embryo cryotolerance, as well as favoring the expression of lipid metabolism regulating and oxidative stress-related transcripts.
Collapse
Affiliation(s)
- K Cañón-Beltrán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain; Departamento de Ciencias Biológicas, Universidad Técnica Particular de Loja, Loja, Ecuador
| | - J Giraldo-Giraldo
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain; Reproductive Biotechnology Laboratory, School of Biosciences, Science Faculty, National University of Colombia, Medellín, Colombia
| | - Y N Cajas
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - P Beltrán-Breña
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - C O Hidalgo
- Department of Animal Selection and Reproduction, The Regional Agri-Food Research and Development Service of Asturias (SERIDA), Gijon, Spain
| | - N Vásquez
- Reproductive Biotechnology Laboratory, School of Biosciences, Science Faculty, National University of Colombia, Medellín, Colombia
| | - C L V Leal
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain; Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - A Gutiérrez-Adán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - E M González
- Department of Anatomy and Embryology, Veterinary Faculty, Complutense University of Madrid (UCM), Madrid, Spain
| | - D Rizos
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain.
| |
Collapse
|
31
|
Amin NB, Carvajal-Gonzalez S, Purkal J, Zhu T, Crowley C, Perez S, Chidsey K, Kim AM, Goodwin B. Targeting diacylglycerol acyltransferase 2 for the treatment of nonalcoholic steatohepatitis. Sci Transl Med 2020; 11:11/520/eaav9701. [PMID: 31776293 DOI: 10.1126/scitranslmed.aav9701] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/25/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by the accumulation of hepatocyte triglycerides, the synthesis of which is catalyzed by diacylglycerol acyltransferases (DGATs). Here, we investigate DGAT2 as a potential therapeutic target using an orally administered, selective DGAT2 inhibitor, PF-06427878. Treatment with PF-06427878 resulted in the reduction of hepatic and circulating plasma triglyceride concentrations and decreased lipogenic gene expression in rats maintained on a Western-type diet. In a mouse model of NASH, histological improvements in steatosis, ballooning, and fibrosis were evident in the livers of animals receiving PF-06427878 compared with mice treated with vehicle alone. We extended these nonclinical studies to two phase 1 studies in humans [NCT02855177 (n = 24) and NCT02391623 (n = 39; n = 38 completed)] and observed that PF-06427878 was well tolerated and influenced markers of liver function (alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and total bilirubin) in healthy adults, with statistically significant reductions from baseline at day 14 in participants treated with PF-06427878 1500 milligrams per day (P < 0.05). Moreover, magnetic resonance imaging using proton density fat fraction showed that PF-06427878 1500 milligrams per day reduced hepatic steatosis in healthy adult participants. Our findings highlight DGAT2 inhibition by a small, potent, selective compound as a potential therapeutic approach for the treatment of NASH.
Collapse
Affiliation(s)
- Neeta B Amin
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA.
| | | | - Julie Purkal
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Tong Zhu
- Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Collin Crowley
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Sylvie Perez
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Kristin Chidsey
- Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Albert M Kim
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Bryan Goodwin
- Internal Medicine Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| |
Collapse
|
32
|
Luo X, Liu Z, Ge X, Huang S, Zhou Y, Li D, Li L, Chen X, Huang L, Hou Q, Cheng H, Xiao L, Liu C, Zou Y, Yang X. High manganese exposure decreased the risk of high triglycerides in workers: a cross-sectional study. BMC Public Health 2020; 20:874. [PMID: 32503499 PMCID: PMC7275562 DOI: 10.1186/s12889-020-09011-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/29/2020] [Indexed: 01/04/2023] Open
Abstract
Background Manganese (Mn) participates in lipid metabolism. However, the associations between Mn exposure and dyslipidaemia is unclear. Methods This was a cross-sectional study. Data were collected from the 2017 the Mn-exposed workers healthy cohort (MEWHC). Finally, 803 occupationally Mn-exposed workers included in the study. The workers were divided into two groups. The grouping of this study was based on Mn-Time Weighted Averages (Mn-TWA). The high-exposure group included participants with Mn-TWA greater than 0.15 mg/m3. The low-exposure group included participants with Mn-TWA less than or equal to 0.15 mg/m3. Mn-TWA levels and dyslipidaemia were assessed. Results After adjustment for seniority, sex, cigarette consumption, alcohol consumption, high-fat diet frequency, medicine intake in the past two weeks, egg intake frequency, drinking tea, WHR, and hypertension, Mn-TWA levels was negatively correlated with high triglycerides (TG) risk in workers overall (OR = 0.51; 95% CI: 0.36, 0.73; p < 0.01). The results of males and females were consistent (OR = 0.53; 95% CI: 0.34, 0.81; p < 0.01) and (OR = 0.47; 95% CI: 0.24, 0.94; p < 0.01), respectively. By performing interactions analyses of workers overall, we observed no significant interactions among confounders. Mn-TWA levels and pack-years on high TG risk (relative excess risk for the interactions (RERI = 2.29, 95% CI: − 2.07, 6.66), (RERI) = 2.98, 95% CI: − 2.30, 8.26). Similarly, smoking status, drinking status, high-fat diet frequency, and Waist-to-Hip Ratio (WHR) showed non-significant interactions with Mn-TWA levels on high TG risk. Conclusions This research indicates that high Mn exposure was negatively related to high TG risk in workers.
Collapse
Affiliation(s)
- Xiaoyu Luo
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhenfang Liu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoting Ge
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Sifang Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanting Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Defu Li
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Longman Li
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiang Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Lulu Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingzhi Hou
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Lili Xiao
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Chaoqun Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, Guangxi, China. .,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
33
|
Li Y, Li J, Cao P, Liu Y. Sinapine-enriched rapeseed oils reduced fatty liver formation in high-fat diet-fed C57BL/6J mice. RSC Adv 2020; 10:21248-21258. [PMID: 35518778 PMCID: PMC9054371 DOI: 10.1039/d0ra00215a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/02/2020] [Indexed: 12/22/2022] Open
Abstract
Oil enrichment with trace amounts of components has significant effects on animal nutrition and health. In this work, the potential impact of sinapine, a trace amount of polyphenol naturally present in rapeseeds, was investigated in high-fat diet (HF)-fed C57BL/6J mice. The mice were fed with different diets including chow diet (LF), HF diet, rapeseed oil-containing HF diet (RO), and rapeseed oils enriched with sinapine (500 mg kg-1 oil, high-fat diet, RP) for 12 weeks. Here, it was demonstrated that sinapine supplementation significantly reduced (P < 0.05) body weight increase, fat accumulation, and fatty liver formation in mice when compared with those fed with a high-fat diet. The TG, LDL-C, ALT and AST levels in the RP group were significantly reduced (P < 0.05) by 15.67%, 73.62%, 20.67%, and 31.58%, respectively, compared with that in the HF group. Besides, the addition of sinapine prevented the degeneration of mouse adipocytes and lipid accumulation in the liver. Moreover, this change was achieved by downregulating SREBP-1c and FAS and upregulating PPAR-α and ACOX1 gene expression levels. Our results indicate that sinapine can be used as a prebiotic to enhance the nutritional function of vegetable oils to prevent obesity-related chronic diseases such as NAFLD.
Collapse
Affiliation(s)
- Youdong Li
- School of Food Science and Technology, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China +86-510-85876799 +86-510-85329081 +86-510-85876799 +86-510-85329081
| | - Jinwei Li
- School of Food Science and Technology, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China +86-510-85876799 +86-510-85329081 +86-510-85876799 +86-510-85329081
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Centre for Functional Food, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China
| | - Peirang Cao
- School of Food Science and Technology, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China +86-510-85876799 +86-510-85329081 +86-510-85876799 +86-510-85329081
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Centre for Functional Food, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China
| | - Yuanfa Liu
- School of Food Science and Technology, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China +86-510-85876799 +86-510-85329081 +86-510-85876799 +86-510-85329081
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Centre for Functional Food, Jiangnan University 1800 Lihu Road Wuxi 214122 Jiangsu People's Republic of China
| |
Collapse
|
34
|
Parry SA, Rosqvist F, Mozes FE, Cornfield T, Hutchinson M, Piche ME, Hülsmeier AJ, Hornemann T, Dyson P, Hodson L. Intrahepatic Fat and Postprandial Glycemia Increase After Consumption of a Diet Enriched in Saturated Fat Compared With Free Sugars. Diabetes Care 2020; 43:1134-1141. [PMID: 32165444 PMCID: PMC7171936 DOI: 10.2337/dc19-2331] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/25/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Debate continues regarding the influence of dietary fats and sugars on the risk of developing metabolic diseases, including insulin resistance and nonalcoholic fatty liver disease (NAFLD). We investigated the effect of two eucaloric diets, one enriched with saturated fat (SFA) and the other enriched with free sugars (SUGAR), on intrahepatic triacylglycerol (IHTAG) content, hepatic de novo lipogenesis (DNL), and whole-body postprandial metabolism in overweight males. RESEARCH DESIGN AND METHODS Sixteen overweight males were randomized to consume the SFA or SUGAR diet for 4 weeks before consuming the alternate diet after a 7-week washout period. The metabolic effects of the respective diets on IHTAG content, hepatic DNL, and whole-body metabolism were investigated using imaging techniques and metabolic substrates labeled with stable-isotope tracers. RESULTS Consumption of the SFA diet significantly increased IHTAG by mean ± SEM 39.0 ± 10.0%, while after the SUGAR diet IHTAG was virtually unchanged. Consumption of the SFA diet induced an exaggerated postprandial glucose and insulin response to a standardized test meal compared with SUGAR. Although whole-body fat oxidation, lipolysis, and DNL were similar following the two diets, consumption of the SUGAR diet resulted in significant (P < 0.05) decreases in plasma total, HDL, and non-HDL cholesterol and fasting β-hydroxybutyrate plasma concentrations. CONCLUSIONS Consumption of an SFA diet had a potent effect, increasing IHTAG together with exaggerating postprandial glycemia. The SUGAR diet did not influence IHTAG and induced minor metabolic changes. Our findings indicate that a diet enriched in SFA is more harmful to metabolic health than a diet enriched in free sugars.
Collapse
Affiliation(s)
- Siôn A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Fredrik Rosqvist
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Ferenc E Mozes
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, John Radcliffe Hospital, Oxford, U.K
| | - Thomas Cornfield
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Matthew Hutchinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Marie-Eve Piche
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Quebec Heart and Lung Institute, Laval University, Quebec, Canada
| | - Andreas J Hülsmeier
- Institute for Clinical Chemistry, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Pamela Dyson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, U.K
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K.
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, U.K
| |
Collapse
|
35
|
Green CJ, Pramfalk C, Charlton CA, Gunn PJ, Cornfield T, Pavlides M, Karpe F, Hodson L. Hepatic de novo lipogenesis is suppressed and fat oxidation is increased by omega-3 fatty acids at the expense of glucose metabolism. BMJ Open Diabetes Res Care 2020; 8:8/1/e000871. [PMID: 32188593 PMCID: PMC7078804 DOI: 10.1136/bmjdrc-2019-000871] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 02/07/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Increased hepatic de novo lipogenesis (DNL) is suggested to be an underlying cause in the development of nonalcoholic fatty liver disease and/or insulin resistance. It is suggested that omega-3 fatty acids (FA) lower hepatic DNL. We investigated the effects of omega-3 FA supplementation on hepatic DNL and FA oxidation using a combination of human in vivo and in vitro studies. RESEARCH DESIGN AND METHODS Thirty-eight healthy men were randomized to take either an omega-3 supplement (4 g/day eicosapentaenoic acid (EPA)+docosahexaenoic acid (DHA) as ethyl esters) or placebo (4 g/day olive oil) and fasting measurements were made at baseline and 8 weeks. The metabolic effects of omega-3 FAs on intrahepatocellular triacylglycerol (IHTAG) content, hepatic DNL and FA oxidation were investigated using metabolic substrates labeled with stable-isotope tracers. In vitro studies, using a human liver cell-line was undertaken to gain insight into the intrahepatocellular effects of omega-3 FAs. RESULTS Fasting plasma TAG concentrations significantly decreased in the omega-3 group and remained unchanged in the placebo group. Eight weeks of omega-3 supplementation significantly decreased IHTAG, fasting and postprandial hepatic DNL while significantly increasing dietary FA oxidation and fasting and postprandial plasma glucose concentrations. In vitro studies supported the in vivo findings of omega-3 FAs (EPA+DHA) decreasing intracellular TAG through a shift in cellular metabolism away from FA esterification toward oxidation. CONCLUSIONS Omega-3 supplementation had a potent effect on decreasing hepatic DNL and increasing FA oxidation and plasma glucose concentrations. Attenuation of hepatic DNL may be considered advantageous; however, consideration is required as to what the potential excess of nonlipid substrates (eg, glucose) will have on intrahepatic and extrahepatic metabolic pathways. TRIAL REGISTRATION NUMBER NCT01936779.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Pavlides
- University of Oxford, Oxford, Oxfordshire, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, UK
| | - Fredrik Karpe
- University of Oxford, Oxford, Oxfordshire, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - Leanne Hodson
- University of Oxford, Oxford, Oxfordshire, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| |
Collapse
|
36
|
Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease. J Med Chem 2020; 63:5031-5073. [PMID: 31930920 DOI: 10.1021/acs.jmedchem.9b01701] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease (NAFLD) characterized by liver steatosis, inflammation, and hepatocellular damage. NASH is a serious condition that can progress to cirrhosis, liver failure, and hepatocellular carcinoma. The association of NASH with obesity, type 2 diabetes mellitus, and dyslipidemia has led to an emerging picture of NASH as the liver manifestation of metabolic syndrome. Although diet and exercise can dramatically improve NASH outcomes, significant lifestyle changes can be challenging to sustain. Pharmaceutical therapies could be an important addition to care, but currently none are approved for NASH. Here, we review the most promising targets for NASH treatment, along with the most advanced therapeutics in development. These include targets involved in metabolism (e.g., sugar, lipid, and cholesterol metabolism), inflammation, and fibrosis. Ultimately, combination therapies addressing multiple aspects of NASH pathogenesis are expected to provide benefit for patients.
Collapse
Affiliation(s)
- F Anthony Romero
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Christopher T Jones
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Yingzi Xu
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Martijn Fenaux
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Randall L Halcomb
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| |
Collapse
|
37
|
Løvsletten NG, Vu H, Skagen C, Lund J, Kase ET, Thoresen GH, Zammit VA, Rustan AC. Treatment of human skeletal muscle cells with inhibitors of diacylglycerol acyltransferases 1 and 2 to explore isozyme-specific roles on lipid metabolism. Sci Rep 2020; 10:238. [PMID: 31937853 PMCID: PMC6959318 DOI: 10.1038/s41598-019-57157-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022] Open
Abstract
Diacylglycerol acyltransferases (DGAT) 1 and 2 catalyse the final step in triacylglycerol (TAG) synthesis, the esterification of fatty acyl-CoA to diacylglycerol. Despite catalysing the same reaction and being present in the same cell types, they exhibit different functions on lipid metabolism in various tissues. Yet, their roles in skeletal muscle remain poorly defined. In this study, we investigated how selective inhibitors of DGAT1 and DGAT2 affected lipid metabolism in human primary skeletal muscle cells. The results showed that DGAT1 was dominant in human skeletal muscle cells utilizing fatty acids (FAs) derived from various sources, both exogenously supplied FA, de novo synthesised FA, or FA derived from lipolysis, to generate TAG, as well as being involved in de novo synthesis of TAG. On the other hand, DGAT2 seemed to be specialised for de novo synthesis of TAG from glycerol-3-posphate only. Interestingly, DGAT activities were also important for regulating FA oxidation, indicating a key role in balancing FAs between storage in TAG and efficient utilization through oxidation. Finally, we observed that inhibition of DGAT enzymes could potentially alter glucose-FA interactions in skeletal muscle. In summary, treatment with DGAT1 or DGAT2 specific inhibitors resulted in different responses on lipid metabolism in human myotubes, indicating that the two enzymes play distinct roles in TAG metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Nils G Løvsletten
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Helene Vu
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Christine Skagen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Eili T Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Victor A Zammit
- Division of Translational and Experimental medicine, Warwick Medical School, University of Warwick, Coventry, UK
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway.
| |
Collapse
|
38
|
Hodson L, Karpe F. Hyperinsulinaemia: does it tip the balance toward intrahepatic fat accumulation? Endocr Connect 2019; 8:R157-R168. [PMID: 31581129 PMCID: PMC6826170 DOI: 10.1530/ec-19-0350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Abstract
In health, the liver is metabolically flexible over the course of the day, as it undertakes a multitude of physiological processes including the regulation of intrahepatic and systemic glucose and lipid levels. The liver is the first organ to receive insulin and through a cascade of complex metabolic processes, insulin not only plays a key role in the intrahepatic regulation of glucose and lipid metabolism, but also in the regulation of systemic glucose and lipid concentrations. Thus, when intrahepatic insulin signalling becomes aberrant then this may lead to perturbations in intrahepatic metabolic processes that have the potential to impact on metabolic health. For example, obesity is associated with intrahepatic fat accumulation (known as nonalcoholic liver disease (NAFLD)) and hyperinsulinaemia, the latter as a result of insulin hypersecretion or impaired hepatic insulin extraction. Although insulin signalling directly alters intra- and extrahepatic metabolism, the regulation of hepatic glucose and fatty acid metabolism is also indirectly driven by substrate availability. Here we discuss the direct and indirect effects of insulin on intrahepatic processes such as the synthesis of fatty acids and peripherally regulating the flux of fatty acids to the liver; processes that may play a role in the development of insulin resistance and/or intrahepatocellular triacylglycerol (IHTAG) accumulation in humans.
Collapse
Affiliation(s)
- Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford University Hospital Trusts, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford University Hospital Trusts, Oxford, UK
| |
Collapse
|
39
|
The loss of ERE-dependent ERα signaling potentiates the effects of maternal high-fat diet on energy homeostasis in female offspring fed an obesogenic diet. J Dev Orig Health Dis 2019; 11:285-296. [PMID: 31543088 DOI: 10.1017/s2040174419000515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal high-fat diet (HFD) alters hypothalamic programming and disrupts offspring energy homeostasis in rodents. We previously reported that the loss of ERα signaling partially blocks the effects of maternal HFD in female offspring fed a standard chow diet. In a companion study, we determined if the effects of maternal HFD were magnified by an adult obesogenic diet in our transgenic mouse models. Heterozygous ERα knockout (wild-type (WT)/KO) dams were fed a control breeder chow diet (25% fat) or a semipurified HFD (45% fat) 4 weeks prior to mating with heterozygous males (WT/KO or WT/ knockin) to produce WT, ERα KO, or ERα knockin/knockout (KIKO) (no estrogen response element (ERE) binding) female offspring, which were fed HFD for 20 weeks. Maternal HFD potentiated the effects of adult HFD on KIKO and KO body weight due to increased adiposity and decreased activity. Maternal HFD also produced KIKO females that exhibit KO-like insulin intolerance and impaired glucose homeostasis. Maternal HFD increased plasma interleukin 6 and monocyte chemoattractant protein 1 levels and G6pc and Pepck liver expression only in WT mice. Insulin and tumor necrosis factor α levels were higher in KO offspring from HFD-fed dams. Arcuate and liver expression of Esr1 was altered in KIKO and WT, respectively. These data suggest that loss of ERE-dependent ERα signaling, and not total ERα signaling, sensitizes females to the deleterious influence of maternal HFD on offspring energy and glucose potentially through the control of peripheral inflammation and hypothalamic and liver gene expression. Future studies will interrogate the tissue-specific mechanisms of maternal HFD programming through ERα signaling.
Collapse
|
40
|
Xu S, Chen Y, Ma Y, Liu T, Zhao M, Wang Z, Zhao L. Lipidomic Profiling Reveals Disruption of Lipid Metabolism in Valproic Acid-Induced Hepatotoxicity. Front Pharmacol 2019; 10:819. [PMID: 31379584 PMCID: PMC6659130 DOI: 10.3389/fphar.2019.00819] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Abstract
Valproic acid (VPA) is one of the most widely prescribed antiepileptic drugs, as VPA-induced hepatotoxicity is one of the most severe adverse reaction that can lead to death. The objective of this study was to gain an understanding of dysregulated lipid metabolism in mechanism of hepatotoxicity. Nontargeted lipidomics analysis with liquid chromatography-quadrupole-time-of-flight mass spectrometry (LC-Q-TOF/MS) was performed to explore differential lipids from the patient serum and L02 cells. Lipidomics data interpretation was augmented by gene expression analyses for the key enzymes in lipid metabolism pathways. From patient serum lipidomics, pronouncedly changed lipid species between abnormal liver function (ALF) patients and normal liver function (NLF) patients were identified. Among these lipid species, LPCs, Cers, and SMs were markedly reduced in the ALF group and showed negative relationships with liver injury severity [alanine aminotransferase (ALT) levels], while significantly increased triacylglycerols (TAG) with higher summed carbon numbers demonstrated a positive relationship with ALT levels. Regarding lipidomics in hepatic L02 cells, TAG was markedly elevated after VPA exposure, especially in TAGs with more than 53 summed carbons. Besides, gene expression analysis revealed dysregulated lipid metabolism in VPA-treated L02 cells. Peroxime proliferators-activated receptor (PPARγ) pathway played an important role in VPA-induced lipid disruption through inducing long-chain fatty acid uptake and TAG synthesis, which was also regulated by Akt pathway. Our findings present that VPA-induced lipid metabolism disruption might lead to lipotoxicity in the liver. This approach is expected to be applicable for other drug-induced toxicity assessments.
Collapse
Affiliation(s)
- Shansen Xu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanan Chen
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yiyi Ma
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting Liu
- Shanghai AB Sciex Analytical Instrument Trading Co. Ltd., Shanghai, China
| | - Mingming Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhanyou Wang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, China
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
41
|
Gabbia D, Roverso M, Guido M, Sacchi D, Scaffidi M, Carrara M, Orso G, Russo FP, Floreani A, Bogialli S, De Martin S. Western Diet-Induced Metabolic Alterations Affect Circulating Markers of Liver Function before the Development of Steatosis. Nutrients 2019; 11:1602. [PMID: 31311123 PMCID: PMC6683046 DOI: 10.3390/nu11071602] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/06/2019] [Accepted: 07/12/2019] [Indexed: 12/15/2022] Open
Abstract
Since nutrition might have a significant impact on liver function, we analyzed the early effect of Western-type diet on hepatic tissue and lipid and drug metabolism in Wistar-Kyoto rats (n = 8); eight rats fed with a standard diet were used as controls. Histological analysis of liver tissue was performed, and plasma biochemical parameters were measured. Plasma concentration of six bile acids was determined by ultra-liquid chromatography-tandem mass spectrometry UHPLC-MS/MS. Hepatic gene expressions of enzymes involved in drug and lipid metabolism were assessed by means of real-time reverse transcription (qRT)-PCR. Liver of rats fed with a Western diet did not show macroscopic histological alterations, but number and diameter of lipid droplets increased, as well as DGAT1, GPAT4, SCD, FASN and SREBP2 expression. Furthermore, Western diet-fed animals showed an increase in the activation of hepatic stellate cells and macrophage number in liver tissue, as well as a significant increase in AST and bilirubin levels (p < 0.01), and in the LDL:HDL cholesterol ratio (p < 0.001). Plasma chenodeoxycholic acid concentration increased significantly, whereas cholic acid decreased (p < 0.05), and cytochrome P450 genes were generally downregulated. Significant changes in hepatic lipid and drug metabolism are early induced by the Western diet, prior to steatosis development. Such changes are associated with a peculiar alteration in circulating bile acids, which could represent an early marker of non-alcoholic fatty liver disease (NAFLD) development.
Collapse
Affiliation(s)
- Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Marco Roverso
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| | - Maria Guido
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Diana Sacchi
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Michela Scaffidi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Maria Carrara
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Annarosa Floreani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Sara Bogialli
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy.
| |
Collapse
|
42
|
XX sex chromosome complement promotes atherosclerosis in mice. Nat Commun 2019; 10:2631. [PMID: 31201301 PMCID: PMC6643208 DOI: 10.1038/s41467-019-10462-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/08/2019] [Indexed: 12/27/2022] Open
Abstract
Men and women differ in circulating lipids and coronary artery disease (CAD). While sex hormones such as estrogens decrease CAD risk, hormone replacement therapy increases risk. Biological sex is determined by sex hormones and chromosomes, but effects of sex chromosomes on circulating lipids and atherosclerosis are unknown. Here, we use mouse models to separate effects of sex chromosomes and hormones on atherosclerosis, circulating lipids and intestinal fat metabolism. We assess atherosclerosis in multiple models and experimental paradigms that distinguish effects of sex chromosomes, and male or female gonads. Pro-atherogenic lipids and atherosclerosis are greater in XX than XY mice, indicating a primary effect of sex chromosomes. Small intestine expression of enzymes involved in lipid absorption and chylomicron assembly are greater in XX male and female mice with higher intestinal lipids. Together, our results show that an XX sex chromosome complement promotes the bioavailability of dietary fat to accelerate atherosclerosis. Men and women differ in their risk of developing coronary artery disease, in part due to differences in their levels of sex hormones. Here, AlSiraj et al. show that the XX sex genotype regulates lipid metabolism and promotes atherosclerosis independently of sex hormones in mice.
Collapse
|
43
|
Cheng Z, Wen Y, Liang B, Chen S, Liu Y, Wang Z, Cheng J, Tang X, Xin H, Deng L. Gene expression profile-based drug screen identifies SAHA as a novel treatment for NAFLD. Mol Omics 2019; 15:50-58. [PMID: 30603757 DOI: 10.1039/c8mo00214b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. Being part of the metabolic syndrome, NAFLD is characterized by the deposition of triglycerides (TGs) as lipid droplets in the cytoplasm of hepatic cells. Recently, the rapid development of high-throughput genome analysis technologies provided opportunities to screen for new drugs for NAFLD. In this study, we screened for potential drugs based on the gene expression profiles of 73 compounds and identified histone deacetylase (HDAC) inhibitors as a novel treatment for the accumulation of lipids in hepatocytes. In the subsequent analysis and experiments, we discovered that SAHA inhibited the fatty acid and lipid metabolism pathways in hepatic cells and induced a significant deficiency of lipid accumulation in HepG2 and SMMC-7721 cells. Furthermore, SAHA inhibited lipid synthesis in hepatic cells by directly suppressing the expression of DGAT2. Hence, our study provides a novel method to screen for effective drugs for liver diseases and identifies SAHA as a potent treatment for NAFLD.
Collapse
Affiliation(s)
- Zhujun Cheng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi Province 330031, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Conte M, Armani A, Conte G, Serra A, Franceschi C, Mele M, Sandri M, Salvioli S. Muscle-specific Perilipin2 down-regulation affects lipid metabolism and induces myofiber hypertrophy. J Cachexia Sarcopenia Muscle 2019; 10:95-110. [PMID: 30288961 PMCID: PMC6438344 DOI: 10.1002/jcsm.12355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/06/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Perilipin2 (Plin2) belongs to a family of five highly conserved proteins, known for their role in lipid storage. Recent data indicate that Plin2 has an important function in cell metabolism and is involved in several human pathologies, including liver steatosis and Type II diabetes. An association between Plin2 and lower muscle mass and strength has been found in elderly and inactive people, but its function in skeletal muscle is still unclear. Here, we addressed the role of Plin2 in adult muscle by gain and loss of function experiments. METHODS By mean of in vivo Plin2 down-regulation (shPlin2) and overexpression (overPlin2) in murine tibialis anterior muscle, we analysed the effects of Plin2 genetic manipulations on myofiber size and lipid composition. An analysis of skeletal muscle lipid composition was also performed in vastus lateralis samples from young and old patients undergoing hip surgery. RESULTS We found that Plin2 down-regulation was sufficient to induce a 30% increase of myofiber cross-sectional area, independently of mTOR pathway. Alterations of lipid content and modulation of genes involved in lipid synthesis occurred in hypertrophic muscles. In particular, we showed a decrease of triglycerides, ceramides, and phosphatidylcoline:phosphatidylethanolamine ratio, a condition known to impact negatively on muscle function. Plin2 overexpression did not change fibre size; however, lipid composition was strongly affected in a way that is similar to that observed in human samples from old patients. CONCLUSIONS Altogether these data indicate that Plin2 is a critical mediator for the control of muscle mass, likely, but maybe not exclusively, through its critical role in the regulation of intracellular lipid content and composition.
Collapse
Affiliation(s)
- Maria Conte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Centre "L. Galvani" (CIG), University of Bologna, Bologna, Italy
| | - Andrea Armani
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Giuseppe Conte
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Andrea Serra
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy.,Research Center of Nutraceuticals and Food for Health, University of Pisa, Pisa, Italy
| | | | - Marcello Mele
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy.,Research Center of Nutraceuticals and Food for Health, University of Pisa, Pisa, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Science, University of Padova, Padova, Italy
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Centre "L. Galvani" (CIG), University of Bologna, Bologna, Italy
| |
Collapse
|
45
|
Irshad Z, Chmel N, Adya R, Zammit VA. Hepatic VLDL secretion: DGAT1 determines particle size but not particle number, which can be supported entirely by DGAT2. J Lipid Res 2019; 60:111-120. [PMID: 30397187 PMCID: PMC6314258 DOI: 10.1194/jlr.m089300] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
We investigated whether, in view of its activity being expressed on both aspects of the endoplasmic reticulum (ER; dual membrane topology), diacylglycerol acyltransferase 1 (DGAT1) plays a distinctive role in determining the triglyceride (TAG) content of VLDL particles secreted by the liver. Mice in which the DGAT1 gene was specifically ablated in hepatocytes (DGAT1-LKO mice) had the same number of VLDL particles (apoB concentration) in the plasma 1 h after Triton 1339 treatment, but these particles were approximately half the size of VLDL particles secreted by control mice and had a proportionately decreased content of TAG, with normal cholesterol and cholesteryl ester contents. Analyses of purified microsomal fractions prepared from 16 h fasted control and DAGT1-LKO mice showed that the TAG/protein ratio in the ER was significantly lower in the latter. Electron micrographs of these livers showed that those from DGAT1-LKO mice did not show the increased lipid content of the smooth ER shown by control livers. The effects of DGAT1- and DGAT2-specific inhibitors on apoB secretion by HepG2 cells showed that DGAT1 is not indispensable for apoB secretion and demonstrated redundancy in the ability of the two enzymes to support apoB secretion. Therefore, our findings show that DGAT1 is essential for the complete lipidation and maturation of VLDL particles within the lumen of the ER, consistent with its dual topology within the ER membrane. In the mouse, DGAT2 can support apoB secretion (particle number) even when TAG availability for full VLDL lipidation is restricted in the absence of DGAT1.
Collapse
Affiliation(s)
- Zehra Irshad
- Translational and Experimental Medicine, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Nikola Chmel
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Raghu Adya
- Translational and Experimental Medicine, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Victor A Zammit
- Translational and Experimental Medicine, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
46
|
Zhang Q, Huang Y, Li X, Liu H, He B, Wang B, Ma Y, Zhou X, Liu Y, Wu S. Tangduqing Granules Attenuate Insulin Resistance and Abnormal Lipid Metabolism through the Coordinated Regulation of PPAR γ and DGAT2 in Type 2 Diabetic Rats. J Diabetes Res 2019; 2019:7403978. [PMID: 31019978 PMCID: PMC6452558 DOI: 10.1155/2019/7403978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance (IR) is a vital hallmark of type 2 diabetes mellitus, which is characterized by an impaired ability of insulin to promote glucose uptake and utilization. Lipid deposition is closely associated with impaired insulin sensitivity. PPARγ plays an important role in glucose homeostasis, adipocyte differentiation, and insulin sensitivity. Likewise, DGAT2 also exerts a crucial role in integrating carbohydrate and lipid metabolism in the liver. The present study is aimed at evaluating a Chinese medicinal formula, Tangduqing granules (TDQ), with multifaceted actions against lipid and glucose metabolism disorder and IR of type 2 diabetes. An animal model of type 2 diabetes was developed by high-fat diet feeding plus low-dose streptozotocin injection. After oral administration of TDQ for 5 weeks, the effects on glucose and lipid metabolism and the underlying mechanism were evaluated by biochemical, histological, RT-PCR, and western blotting methods. The results showed that TDQ decreased fasting blood glucose, ameliorated glucose tolerance, and improved IR. Besides, TDQ regulated hyperlipidemia symptoms, decreased serum lipid levels and liver TG, and reduced hepatic steatosis in a type 2 diabetic rat model. Furthermore, TDQ reversed diabetes-induced decrease in the mRNA and protein expression of PPARγ and elevation in the mRNA and protein levels of DGAT2 in the liver. In addition, we showed that interference of TDQ ameliorated palmitate-induced glucose and lipid metabolic abnormalities in HepG2 cells. TDQ are, therefore, a potential Chinese medicinal formula that relieves IR and lipid metabolism disorder might be through promotion of PPARγ and decrease of DGAT2 expression.
Collapse
Affiliation(s)
- Qinghua Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingying Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaojin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongyi Liu
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Yunnan, China
| | | | - Bin Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuntao Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiang Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yaqin Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shentao Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
47
|
Döring S, Seeßle J, Gan-Schreier H, Javaheri B, Jiao L, Cheng Y, Tuma-Kellner S, Liebisch G, Herrmann T, Stremmel W, Chamulitrat W. Elevation of blood lipids in hepatocyte-specific fatty acid transport 4-deficient mice fed with high glucose diets. Mol Genet Metab 2019; 126:30-38. [PMID: 30497809 DOI: 10.1016/j.ymgme.2018.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/12/2018] [Accepted: 11/17/2018] [Indexed: 11/20/2022]
Abstract
Fatty acid transport protein4 (FATP4) is upregulated in acquired and central obesity and its polymorphisms are associated with blood lipids and insulin resistance. Patients with FATP4 mutations and mice with global FATP4 deletion exhibit skin abnormalities characterized as ischthyosis prematurity syndrome (IPS). Cumulating data have shown that an absence of FATP4 increases the levels of cellular triglycerides (TG). However, FATP4 role and consequent lipid and TG metabolism in the hepatocyte is still elusive. Here, hepatocyte-specific FATP4 deficient (Fatp4L-/-) mice were generated. When fed with chow, these mutant mice displayed no phenotypes regarding blood lipids. However when fed low-fat/high-sugar (HS) or high-fat/high-sugar (HFS) for 12 weeks, Fatp4L-/- mice showed a significant increase of plasma TG, free fatty acids and glycerol when compared with diet-fed control mice. Interestingly, Fatp4L-/- mice under HS diet had lower body and liver weights and they were not protected from HFS-induced body weight gain and hepatic steatosis. Male mutant mice were more sensitive to HFS diet than female mutant mice. Glucose intolerance was observed only in female Fatp4L-/- mice fed with HS diet. Lipidomics analyses revealed that hepatic phospholipids were not disturbed in mutant mice under both diets. Thus, hepatic FATP4 deletion rendered an increase of blood lipids including glycerol indicating a preferential fatty-acid channeling to TG pools that are specifically available for lipolysis. Our results imply a possible risk of hyperlipidemia as a result of abnormal metabolism in liver in IPS patients with FATP4 mutations who consume high-sugar diets.
Collapse
Affiliation(s)
- Stephan Döring
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jessica Seeßle
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Bahador Javaheri
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Li Jiao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, Yunnan 650118, China
| | - Yuting Cheng
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Thomas Herrmann
- Westkuesten Hospital, Esmarchstraße 50, 25746 Heide, Germany
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
48
|
Yang P, Zhang H, Wan J, Hu J, Liu J, Wang J, Zhang Y, Yu LL. Dietary sn-2 palmitic triacylglycerols reduced faecal lipids, calcium contents and altered lipid metabolism in Sprague-Dawley rats. Int J Food Sci Nutr 2018; 70:474-483. [PMID: 30569770 DOI: 10.1080/09637486.2018.1541968] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this study, the impact of dietary sn-2 palmitic triacylglycerol (sn-2 PTAG) on faecal lipids, calcium excretion and lipid metabolic alternation was investigated in Sprague-Dawley (SD) rats fed with high-fat diet containing either palm olein (PO, sn-2 palmitic acid (PA) of 14.8%), sn-2 PTAG50 (sn-2 PA of 56.4%) or sn-2 PTAG70 (sn-2 PA of 72.4%), respectively. After 4-week feeding period, SD rats fed with sn-2 PTAGs showed reduced faecal soap fatty acids, neutral lipid and calcium excretion compared to those of PO-fed rats, whereas a significant difference was only observed for the sn-2 PTAG70-fed rats (p < .05). Moreover, dietary sn-2 PTAG70 also showed a significant effect on decreasing serum triacylglycerol (TAG) level, reducing perirenal adipocyte size and regulating lipid metabolism in small intestine and perirenal adipose tissue of SD rats. Significantly increased mRNA levels of genes involved in intestinal lipid anabolism as well as lipid catabolism were both observed in the sn-2 PTAG70-fed rats (p < .05). Meanwhile, dietary sn-2 PTAG70 also significantly up-regulated lipolysis, mitochondrial fatty acid oxidation and thermogenesis-related gene and protein levels in perirenal adipose tissue, which might be correlated with the reduced perirenal adipocyte size. Taken together, our findings indicated that sn-2 PTAG70 may have some beneficial effects on intestinal lipid utilisation and lipid metabolic activity for energy supply in visceral adipose tissue.
Collapse
Affiliation(s)
- Puyu Yang
- a Institute of Food and Nutraceutical Science, School of Agriculture and Biology , Shanghai Jiao Tong University , Shanghai , China
| | - Hong Zhang
- b Wilmar (Shanghai) Biotechnology Research & Development Center Co. Ltd , Shanghai , China
| | - Jianchun Wan
- b Wilmar (Shanghai) Biotechnology Research & Development Center Co. Ltd , Shanghai , China
| | - Jinyu Hu
- a Institute of Food and Nutraceutical Science, School of Agriculture and Biology , Shanghai Jiao Tong University , Shanghai , China
| | - Junchen Liu
- a Institute of Food and Nutraceutical Science, School of Agriculture and Biology , Shanghai Jiao Tong University , Shanghai , China
| | - Jing Wang
- c Beijing Advanced Innovation Center for Food Nutrition and Human Health , Beijing Technology & Business University (BTBU) , Beijing , China
| | - Yaqiong Zhang
- a Institute of Food and Nutraceutical Science, School of Agriculture and Biology , Shanghai Jiao Tong University , Shanghai , China
| | - Liangli Lucy Yu
- d Department of Nutrition and Food Science , University of Maryland , College Park , ML , USA
| |
Collapse
|
49
|
Singh M, Bittner S, Li Y, Bittner A, Han L, Cortez Y, Inayathullah M, Arif Z, Parthasarathi R, Rajadas J, Shen WJ, Nicolls MR, Kraemer FB, Azhar S. Anti-hyperlipidaemic effects of synthetic analogues of nordihydroguaiaretic acid in dyslipidaemic rats. Br J Pharmacol 2018; 176:369-385. [PMID: 30374952 DOI: 10.1111/bph.14528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/07/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Previous studies have shown that Creosote bush-derived nordihydroguaiaretic acid (NDGA) exerts beneficial actions on the key components of metabolic syndrome including dyslipidaemia, insulin resistance and hypertension in several relevant rodent models. Here, we synthesized and screened a total of 6 anti-hyperlipidaemic analogues of NDGA and tested their efficacy against hepatic lipid metabolism in a high-fructose diet (HFrD) fed dyslipidaemic rat model. EXPERIMENTAL APPROACH HFrD fed Sprague-Dawley rats treated with NDGA or one of the six analogues were used. Serum samples were analysed for blood metabolites, whereas liver samples were quantified for changes in various mRNA levels by real-time RT-PCR. KEY RESULTS Oral gavage of HFrD-fed rats for 4 days with NDGA analogues 1 and 2 (100 mg·kg-1 ·day-1 ) suppressed the hepatic triglyceride content, whereas the NDGA analogues 2, 3 and 4, like NDGA, decreased the plasma triglyceride levels by 70-75%. qRT-PCR measurements demonstrated that among NDGA analogues 1, 2, 4 and 5, analogue 4 was the most effective at inhibiting the mRNA levels of some key enzymes and transcription factors involved in lipogenesis. All four analogues almost equally inhibited the key genes involved in triglyceride synthesis and fatty acid elongation. Unlike NDGA, none of the analogues affected the genes of hepatic fatty acid oxidation or transport. CONCLUSIONS AND IMPLICATIONS Our data suggest that NDGA analogues 1, 2, 4 and 5, particularly analogue 4, exert their anti-hyperlipidaemic actions by negatively targeting genes of key enzymes and transcription factors involved in lipogenesis, triglyceride synthesis and fatty acid elongation. These analogues have therapeutic potential.
Collapse
Affiliation(s)
- Madhurima Singh
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Yihang Li
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Alex Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Lu Han
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Yuan Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Zeeshan Arif
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | | | - Jayakumar Rajadas
- Division of Cardiovascular Pharmacology CVI, BioADD Laboratory, Stanford, CA, USA
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Mark R Nicolls
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.,Division of Endocrinology, Gerontology and Metabolism, Standford, CA, USA
| |
Collapse
|
50
|
Pabst B, Futatsugi K, Li Q, Ahn K. Mechanistic Characterization of Long Residence Time Inhibitors of Diacylglycerol Acyltransferase 2 (DGAT2). Biochemistry 2018; 57:6997-7010. [DOI: 10.1021/acs.biochem.8b01096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|