1
|
Corbalan J, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized Monoacylglycerol Acyltransferase 2 Mice Develop Metabolic Dysfunction-Associated Steatohepatitis. J Lipid Res 2024:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [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: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquintinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Hamilton, NJ 08691, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Hamilton, NJ 08691, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Hamilton, NJ 08691, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Hamilton, NJ 08691, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Hamilton, NJ 08691, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901, USA.
| |
Collapse
|
2
|
Powell DR, Doree DD, Shadoan MK, Platt KA, Brommage R, Vogel P, Revelli JP. Mice Lacking Mrs2 Magnesium Transporter are Hypophagic and Thin When Maintained on a High-Fat Diet. Endocrinology 2024; 165:bqae072. [PMID: 38878275 DOI: 10.1210/endocr/bqae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Indexed: 07/05/2024]
Abstract
Genes regulating body fat are shared with high fidelity by mice and humans, indicating that mouse knockout (KO) phenotyping might identify valuable antiobesity drug targets. Male Mrs2 magnesium transporter (Mrs2) KO mice were recently reported as thin when fed a high-fat diet (HFD). They also exhibited increased energy expenditure (EE)/body weight and had beiged adipocytes that, along with isolated hepatocytes, demonstrated increased oxygen consumption, suggesting that increased EE drove the thin phenotype. Here we provide our data on these and additional assays in Mrs2 KO mice. We generated Mrs2 KO mice by homologous recombination. HFD-fed male and female Mrs2 KO mice had significantly less body fat, measured by quantitative magnetic resonance, than wild-type (WT) littermates. HFD-fed Mrs2 KO mice did not demonstrate increased EE by indirect calorimetry and could not maintain body temperature at 4 °C, consistent with their decreased brown adipose tissue stores but despite increased beige white adipose tissue. Instead, when provided a choice between HFD and low-fat diet (LFD), Mrs2 KO mice showed a significant 15% decrease in total energy intake resulting from significantly lower HFD intake that offset numerically increased LFD intake. Food restriction studies performed using WT mice suggested that this decrease in energy intake could explain the loss of body fat. Oral glucose tolerance test studies revealed significantly improved insulin sensitivity in Mrs2 KO mice. We conclude that HFD-fed Mrs2 KO mice are thin with improved insulin sensitivity, and that this favorable metabolic phenotype is driven by hypophagia. Further evaluation is warranted to determine the suitability of MRS2 as a drug target for antiobesity therapeutics.
Collapse
Affiliation(s)
| | - Deon D Doree
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | |
Collapse
|
3
|
Zhan M, Liu X, Xia X, Yang Y, Xie Y, Zhang L, Lin C, Zhu J, Ding W, Xu S. Promotion of neuroinflammation by the glymphatic system: a new insight into ethanol extracts from Alisma orientale in alleviating obesity-associated cognitive impairment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155147. [PMID: 37864890 DOI: 10.1016/j.phymed.2023.155147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/20/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Obesity is one of the critical risk factors for cognitive dysfunction. The glymphatic system (GS) plays a key role in the pathogenesis of cognitive deficits. Alisma orientale has been shown to have anti-inflammatory and antihyperlipidemic effects, whereas its effects and underlying mechanisms on obesity-associated cognitive impairment (OACI) are unclear. PURPOSE This work aims to decipher the mechanism of ethanol extracts from Alisma orientale (EEAO) in restoring cognitive impairment in HFD-induced obese mice through a GS approach. METHODS The restoration of abnormal glucose/lipid metabolism and excess adipose deposition by EEAO were assayed by biochemical analysis and visually displayed by a micro-CT scanner and Oil Red O staining. Biochemical assays and Western blotting (WB) were used to measure cerebral blood flow (CBF), free fatty acid (FFAs) levels and the structural integrity of the blood-brain barrier (BBB). Microglial activation and neuroinflammation were assessed with immunohistochemistry staining, ELISA and WB. Moreover, GS function was determined by immunofluorescence staining, fluorescence tracer imaging and WB. Finally, the neuropathological features and cognitive functions were detested with immunohistochemistry staining, immunofluorescence and Morris Water Maze. RESULTS EEAO not only alleviated body weight, cerebral lipid accumulation and serum FFAs in HFD-induced obese mice, but also increased CBF and BBB integrity. EEAO suppressed microglial activation and lipid deposition in the hippocampus and reduced the level of inflammatory cytokines including IL-6, IL-1β and TNF-α in brain tissue. Interestingly, long-term HFD-induced GS dysfunction was significantly restored after EEAO intervention, and neuropathological lesions and cognitive deficits were also markedly rescued. CONCLUSION EEAO rescued the cognitive deficits of OACI by inhibiting neuroinflammation and restoring GS dysfunction, indicating a potential remedy for OACI.
Collapse
Affiliation(s)
- Meng Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiao Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunqiao Lin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiushuang Zhu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
4
|
Wang Y, Song L, Ning M, Hu J, Cai H, Song W, Gong D, Liu L, Smith J, Li H, Huang Y. Identification of alternative splicing events related to fatty liver formation in duck using full-length transcripts. BMC Genomics 2023; 24:92. [PMID: 36858953 PMCID: PMC9976415 DOI: 10.1186/s12864-023-09160-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is one of most common diseases in the world. Recently, alternative splicing (AS) has been reported to play a key role in NAFLD processes in mammals. Ducks can quickly form fatty liver similar to human NAFLD after overfeeding and restore to normal liver in a short time, suggesting that ducks are an excellent model to unravel molecular mechanisms of lipid metabolism for NAFLD. However, how alternative splicing events (ASEs) affect the fatty liver process in ducks is still unclear. RESULTS Here we identify 126,277 unique transcripts in liver tissue from an overfed duck (77,237 total transcripts) and its sibling control (69,618 total transcripts). We combined these full-length transcripts with Illumina RNA-seq data from five pairs of overfed ducks and control individuals. Full-length transcript sequencing provided us with structural information of transcripts and Illumina RNA-seq data reveals the expressional profile of each transcript. We found, among these unique transcripts, 30,618 were lncRNAs and 1,744 transcripts including 155 lncRNAs and 1,589 coding transcripts showed significantly differential expression in liver tissues between overfed ducks and control individuals. We also detected 27,317 ASEs and 142 of them showed significant relative abundance changes in ducks under different feeding conditions. Full-length transcript profiles together with Illumina RNA-seq data demonstrated that 10 genes involving in lipid metabolism had ASEs with significantly differential abundance in normally fed (control) and overfed ducks. Among these genes, protein products of five genes (CYP4F22, BTN, GSTA2, ADH5, and DHRS2 genes) were changed by ASEs. CONCLUSIONS This study presents an example of how to identify ASEs related to important biological processes, such as fatty liver formation, using full-length transcripts alongside Illumina RNA-seq data. Based on these data, we screened out ASEs of lipid-metabolism related genes which might respond to overfeeding. Our future ability to explore the function of genes showing AS differences between overfed ducks and their sibling controls, using genetic manipulations and co-evolutionary studies, will certainly extend our knowledge of genes related to the non-pathogenic fatty liver process.
Collapse
Affiliation(s)
- Yiming Wang
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China
| | - Linfei Song
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China
| | - Mengfei Ning
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China
| | - Jiaxiang Hu
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China
| | - Han Cai
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China
| | - Weitao Song
- Department of Waterfowl Breeding and Production, Jiangsu Institute of Poultry Science, No. 58 Cangjie Road, Hanjiang District, Yangzhou, 349019093, China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Huifang Li
- Department of Waterfowl Breeding and Production, Jiangsu Institute of Poultry Science, No. 58 Cangjie Road, Hanjiang District, Yangzhou, 349019093, China.
| | - Yinhua Huang
- State Key Laboratory for Agrobiotechnology, College of Biology Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Hai Dian District, Beijing, 100193, China.
| |
Collapse
|
5
|
Otaibi AA, Mubarak SA, Qarni AA, Hawwari A, Bakillah A, Iqbal J. ATP-Binding Cassette Protein ABCC10 Deficiency Prevents Diet-Induced Obesity but Not Atherosclerosis in Mice. Int J Mol Sci 2022; 23:ijms232213813. [PMID: 36430292 PMCID: PMC9694421 DOI: 10.3390/ijms232213813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022] Open
Abstract
Excess plasma lipid levels are a risk factor for various cardiometabolic disorders. Studies have shown that improving dyslipidemia lowers the progression of these disorders. In this study, we investigated the role of ATP-binding cassette transporter C10 (ABCC10) in regulating lipid metabolism. Our data indicate that deletion of the Abcc10 gene in male mice results in lower plasma and intestinal triglycerides by around 38% and 36%, respectively. Furthermore, deletion of ABCC10 ameliorates diet-induced obesity in mice and leads to a better response during insulin and glucose tolerance tests. Unexpectedly, ABCC10 deficiency does not affect triglyceride levels or atherosclerosis in ApoE-deficient mice. In addition, our studies demonstrate low oleate uptake by enterocytes (~25-30%) and less absorption (~37%) of triglycerides in the small intestine of ABCC10 knockout mice. Deletion of the Abcc10 gene also alters several lipid metabolism genes in the intestine, suggesting that ABCC10 regulates dietary fat absorption, which may contribute to diet-induced obesity in mice.
Collapse
|
6
|
Xia X, Xie Y, Gong Y, Zhan M, He Y, Liang X, Jin Y, Yang Y, Ding W. Electroacupuncture promoted intestinal defensins and rescued the dysbiotic cecal microbiota of high-fat diet-induced obese mice. Life Sci 2022; 309:120961. [PMID: 36116529 DOI: 10.1016/j.lfs.2022.120961] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 10/31/2022]
Abstract
Obesity is currently one of the most important challenges to public health worldwide. Acupuncture has been widely used to treat obesity. However, whether acupuncture regulates intestinal innate immunity via intestinal microbiota against obesity remains to be elucidated. In this study, electroacupuncture (EA) effectively reduced body weight and fat accumulation in obese mice persistently fed a high-fat diet. Full-length 16S rDNA sequencing showed dysbiotic microbiota in the cecum of obese mice. The composition and function of the cecal microbiota of obese mice were markedly restored after EA treatment. After 21 d of EA intervention, the expression of defensin alpha 5 (Defa5) was restored to healthy controls, whereas fat digestion and absorption genes including fabp1 were markedly decreased in the jejunum of obese mice. The Defa5 levels were positively correlated with the family Lachnospiraceae and negatively correlated with obesity indexes. EA also reduced tissue inflammation, ameliorated misaligned glucose tolerance, and inhibited key genes for intestinal lipid absorption. In summary, EA exerted an anti-obesity effect by promoting intestinal defensins, rescuing dysbiotic cecal microbiota, and reducing lipid absorption in a synergistic mode. We present for the first time the key role of alpha defensins in the relationship between gut microbiota and disease during electroacupuncture treatment of obesity. The mucosal innate immunity seems to have a stronger ability to shape the microbiota than dietary factors.
Collapse
Affiliation(s)
- Xiuwen Xia
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China.
| | - Ya Xie
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Yanju Gong
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Meng Zhan
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Yan He
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Xinyu Liang
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Yue Jin
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Youjun Yang
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China
| | - Weijun Ding
- Chengdu University of Traditional Chinese Medicine, Wenjiang District, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Burchat N, Akal T, Ntambi JM, Trivedi N, Suresh R, Sampath H. SCD1 is nutritionally and spatially regulated in the intestine and influences systemic postprandial lipid homeostasis and gut-liver crosstalk. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159195. [PMID: 35718096 PMCID: PMC11287785 DOI: 10.1016/j.bbalip.2022.159195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/21/2022] [Accepted: 06/01/2022] [Indexed: 01/27/2023]
Abstract
Stearoyl-CoA desaturase-1 is an endoplasmic reticulum (ER)-membrane resident protein that inserts a double bond into saturated fatty acids, converting them into their monounsaturated counterparts. Previous studies have demonstrated an important role for SCD1 in modulating tissue and systemic health. Specifically, lack of hepatic or cutaneous SCD1 results in significant reductions in tissue esterified lipids. While the intestine is an important site of lipid esterification and assimilation into the body, the regulation of intestinal SCD1 or its impact on lipid composition in the intestine and other tissues has not been investigated. Here we report that unlike other lipogenic enzymes, SCD1 is enriched in the distal small intestine and in the colon of chow-fed mice and is robustly upregulated by acute refeeding of a high-sucrose diet. We generated a mouse model lacking SCD1 specifically in the intestine (iKO mice). These mice have significant reductions not only in intestinal lipids, but also in plasma triacylglycerols, diacylglycerols, cholesterol esters, and free cholesterol. Additionally, hepatic accumulation of diacylglycerols is significantly reduced in iKO mice. Comprehensive targeted lipidomic profiling revealed a consistent reduction in the myristoleic (14:1) to myristic (14:0) acid ratios in intestine, liver, and plasma of iKO mice. Consistent with the reduction of the monounsaturated fatty acid myristoleic acid in hepatic lipids of chow fed iKO mice, hepatic expression of Pgc-1α, Sirt1, and related fatty acid oxidation genes were reduced in chow-fed iKO mice. Further, lack of intestinal SCD1 reduced expression of de novo lipogenic genes in distal intestine of chow-fed mice and in the livers of mice fed a lipogenic high-sucrose diet. Taken together, these studies reveal a novel pattern of expression of SCD1 in the intestine. They also demonstrate that intestinal SCD1 modulates lipid content and composition of not only intestinal tissues, but also that of plasma and liver. Further, these data point to intestinal SCD1 as a modulator of gut-liver crosstalk, potentially through the production of novel signaling lipids such as myristoleic acid. These data have important implications to understanding how intestinal SCD1 may modulate risk for post-prandial lipemia, hepatic steatosis, and related pathologies.
Collapse
Affiliation(s)
- Natalie Burchat
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, United States of America
| | - Tasleenpal Akal
- Department of Nutritional Sciences, Rutgers University, United States of America
| | - James M Ntambi
- Departments of Biochemistry and Nutritional Sciences, University of Wisconsin-Madison, United States of America
| | - Nirali Trivedi
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, United States of America
| | - Ranjita Suresh
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, United States of America
| | - Harini Sampath
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, United States of America; Department of Nutritional Sciences, Rutgers University, United States of America.
| |
Collapse
|
8
|
Ghanem M, Lewis GF, Xiao C. Recent advances in cytoplasmic lipid droplet metabolism in intestinal enterocyte. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159197. [PMID: 35820577 DOI: 10.1016/j.bbalip.2022.159197] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Processing of dietary fats in the intestine is a highly regulated process that influences whole-body energy homeostasis and multiple physiological functions. Dysregulated lipid handling in the intestine leads to dyslipidemia and atherosclerotic cardiovascular disease. In intestinal enterocytes, lipids are incorporated into lipoproteins and cytoplasmic lipid droplets (CLDs). Lipoprotein synthesis and CLD metabolism are inter-connected pathways with multiple points of regulation. This review aims to highlight recent advances in the regulatory mechanisms of lipid processing in the enterocyte, with particular focus on CLDs. In-depth understanding of the regulation of lipid metabolism in the enterocyte may help identify therapeutic targets for the treatment and prevention of metabolic disorders.
Collapse
Affiliation(s)
- Murooj Ghanem
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology, University of Toronto, and University Health Network, Toronto, ON, Canada
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
9
|
Stone SJ. Mechanisms of intestinal triacylglycerol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159151. [PMID: 35296424 DOI: 10.1016/j.bbalip.2022.159151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
Abstract
Triacylglycerols are a major source of stored energy that are obtained either from the diet or can be synthesized to some extent by most tissues. Alterations in pathways of triacylglycerol metabolism can result in their excessive accumulation leading to obesity, insulin resistance, cardiovascular disease and nonalcoholic fatty liver disease. Most tissues in mammals synthesize triacylglycerols via the glycerol 3-phosphate pathway. However, in the small intestine the monoacylglycerol acyltransferase pathway is the predominant pathway for triacylglycerol biosynthesis where it participates in the absorption of dietary triacylglycerol. In this review, the enzymes that are part of both the glycerol 3-phosphate and monoacylglycerol acyltransferase pathways and their contributions to intestinal triacylglycerol metabolism are reviewed. The potential of some of the enzymes involved in triacylglycerol synthesis in the small intestine as possible therapeutic targets for treating metabolic disorders associated with elevated triacylglycerol is briefly discussed.
Collapse
Affiliation(s)
- Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
10
|
Zhou YG, Yin RX, Huang F, Wu JZ, Chen WX, Cao XL. DGAT2-MOGAT2 SNPs and Gene-Environment Interactions on Serum Lipid Profiles and the Risk of Ischemic Stroke. Front Cardiovasc Med 2021; 8:685970. [PMID: 34901200 PMCID: PMC8654148 DOI: 10.3389/fcvm.2021.685970] [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: 03/26/2021] [Accepted: 10/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The genetic susceptibility to ischemic stroke (IS) is still not well-understood. Recent genome-wide association studies (GWASes) found that several single nucleotide polymorphisms (SNPs) in the Diacylglycerol acyltransferase 2 gene (DGAT2) and monoacylglycerol O-acyltransferase 2 (MOGAT2) cluster were associated with serum lipid levels. However, the association between the DGAT2-MOGAT2 SNPs and serum lipid phenotypes has not yet been verified in the Chinese people. Therefore, the present study was to determine the DGAT2-MOGAT2 SNPs and gene-environment interactions on serum lipid profiles and the risk of IS. Methods: Genotyping of 5 SNPs (DGAT2 rs11236530, DGAT2 rs3060, MOGAT2 rs600626, MOGAT2 rs609379, and MOGAT2 rs10899104) in 544 IS patients and 561 healthy controls was performed by the next-generation sequencing technologies. The association between genotypes and serum lipid data was determined by analysis of covariance, and a corrected P-value was adopted after Bonferroni correction. Unconditional logistic regression analysis was performed to assess the association between genotypes and the risk of IS after adjustment of potential confounders. Results: The rs11236530A allele was associated with increased risk of IS (CA/AA vs. CC, OR = 1.45, 95%CI = 1.12-1.88, P = 0.0044), whereas the rs600626G-rs609379A-rs10899104G haplotype was associated with decreased risk of IS (adjusted OR = 0.67, 95% CI = 0.48-0.93, P = 0.018). The rs11236530A allele carriers had lower high-density lipoprotein cholesterol (HDL-C) concentrations than the rs11236530A allele non-carriers (P < 0.001). The interactions of rs11236530-smoking, rs3060-smoking and rs10899104-smoking influenced serum apolipoprotein B levels, whereas the interactions of rs11236530- and rs3060-alcohol affected serum HDL-C levels (P I < 0.004-0.001). The interaction of rs600626G-rs609379A-rs10899104G-alcohol (OR = 0.41, 95% CI = 0.22-0.76) and rs600626G-rs609379C-rs10899104T-alcohol (OR = 0.12, 95% CI = 0.04-0.36) decreased the risk of IS (P I < 0.0001). Conclusions: The rs11236530A allele was associated with decreased serum HDL-C levels in controls and increased risk of IS in patient group. The rs600626G-rs609379A-rs10899104G haplotype, the rs600626G-rs 609379A-rs10899104G-alcohol and rs600626G-rs609379C-rs10899104T-alcohol interactions were associated with decreased risk of IS. The rs11236530 SNP may be a genetic marker for IS in our study populations.
Collapse
Affiliation(s)
- Yong-Gang Zhou
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Feng Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Jin-Zhen Wu
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Wu-Xian Chen
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xiao-Li Cao
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Gutiérrez-Guerrero YT, Ibarra-Laclette E, Martínez del Río C, Barrera-Redondo J, Rebollar EA, Ortega J, León-Paniagua L, Urrutia A, Aguirre-Planter E, Eguiarte LE. Genomic consequences of dietary diversification and parallel evolution due to nectarivory in leaf-nosed bats. Gigascience 2020; 9:giaa059. [PMID: 32510151 PMCID: PMC7276932 DOI: 10.1093/gigascience/giaa059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The New World leaf-nosed bats (Phyllostomids) exhibit a diverse spectrum of feeding habits and innovations in their nutrient acquisition and foraging mechanisms. However, the genomic signatures associated with their distinct diets are unknown. RESULTS We conducted a genomic comparative analysis to study the evolutionary dynamics related to dietary diversification and specialization. We sequenced, assembled, and annotated the genomes of five Phyllostomid species: one insect feeder (Macrotus waterhousii), one fruit feeder (Artibeus jamaicensis), and three nectar feeders from the Glossophaginae subfamily (Leptonycteris yerbabuenae, Leptonycteris nivalis, and Musonycteris harrisoni), also including the previously sequenced vampire Desmodus rotundus. Our phylogenomic analysis based on 22,388 gene families displayed differences in expansion and contraction events across the Phyllostomid lineages. Independently of diet, genes relevant for feeding strategies and food intake experienced multiple expansions and signatures of positive selection. We also found adaptation signatures associated with specialized diets: the vampire exhibited traits associated with a blood diet (i.e., coagulation mechanisms), whereas the nectarivore clade shares a group of positively selected genes involved in sugar, lipid, and iron metabolism. Interestingly, in fruit-nectar-feeding Phyllostomid and Pteropodids bats, we detected positive selection in two genes: AACS and ALKBH7, which are crucial in sugar and fat metabolism. Moreover, in these two proteins we found parallel amino acid substitutions in conserved positions exclusive to the tribe Glossophagini and to Pteropodids. CONCLUSIONS Our findings illuminate the genomic and molecular shifts associated with the evolution of nectarivory and shed light on how nectar-feeding bats can avoid the adverse effects of diets with high glucose content.
Collapse
Affiliation(s)
- Yocelyn T Gutiérrez-Guerrero
- Departamento de Ecología Evolutiva, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| | - Enrique Ibarra-Laclette
- Red de Estudios Moleculares Avanzados, Instituto de Ecología AC, 91070 Xalapa, Veracruz, Mexico
| | | | - Josué Barrera-Redondo
- Departamento de Ecología Evolutiva, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| | - Eria A Rebollar
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, 62210 Cuernavaca, Morelos, Mexico
| | - Jorge Ortega
- Departamento de Zoología, Laboratorio de Bioconservación y Manejo, Posgrado en Ciencias Quimicobiológicas, Instituto Politécnico Nacional-ENCB, 11340 Mexico City, Mexico
| | - Livia León-Paniagua
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| | - Araxi Urrutia
- Departamento de Ecología Funcional, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| | - Erika Aguirre-Planter
- Departamento de Ecología Evolutiva, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| | - Luis E Eguiarte
- Departamento de Ecología Evolutiva, Instituto de Ecología, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, 04510 Coyoacán, Mexico City, Mexico
| |
Collapse
|
12
|
Mochida T, Take K, Maki T, Nakakariya M, Adachi R, Sato K, Kitazaki T, Takekawa S. Inhibition of MGAT2 modulates fat-induced gut peptide release and fat intake in normal mice and ameliorates obesity and diabetes in ob/ob mice fed on a high-fat diet. FEBS Open Bio 2019; 10:316-326. [PMID: 31837122 PMCID: PMC7050258 DOI: 10.1002/2211-5463.12778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/12/2019] [Accepted: 12/11/2019] [Indexed: 11/29/2022] Open
Abstract
Monoacylglycerol O‐acyltransferase 2 (MGAT2) is one of the key enzymes responsible for triglyceride (TG) re‐synthesis in the small intestine. We have previously demonstrated that pharmacological inhibition of MGAT2 has beneficial effects on obesity and metabolic disorders in mice. Here, we further investigate the effects of MGAT2 inhibition on (a) fat‐induced gut peptide release and fat intake in normal mice and (b) metabolic disorders in high‐fat diet (HFD)‐fed ob/ob mice, a model of severe obesity and type 2 diabetes mellitus, using an orally bioavailable MGAT2 inhibitor Compound B (CpdB). CpdB inhibited elevation of plasma TG in mice challenged with an oil‐supplemented liquid meal. Oil challenge stimulated the secretion of two gut anorectic hormones (peptide tyrosine–tyrosine and glucagon‐like peptide‐1) into the bloodstream, and these responses were augmented in mice pretreated with CpdB. In a two‐choice test using an HFD and a low‐fat diet, CpdB selectively inhibited intake of the HFD in normal mice. Administration of CpdB to HFD‐fed ob/ob mice for 5 weeks suppressed food intake and body weight gain and inhibited elevation of glycated hemoglobin. These results indicate that pharmacological MGAT2 inhibition modulates fat‐induced gut peptide release and fat intake in normal mice and improves obesity and diabetes in HFD‐fed ob/ob mice and thus may have potential for development into a treatment of obesity and its related metabolic diseases.
Collapse
Affiliation(s)
- Taisuke Mochida
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kazumi Take
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Toshiyuki Maki
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Ryutaro Adachi
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kenjiro Sato
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Tomoyuki Kitazaki
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Shiro Takekawa
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
13
|
Vergès B, Duvillard L, Pais de Barros JP, Bouillet B, Baillot-Rudoni S, Rouland A, Sberna AL, Petit JM, Degrace P, Demizieux L. Liraglutide Reduces Postprandial Hyperlipidemia by Increasing ApoB48 (Apolipoprotein B48) Catabolism and by Reducing ApoB48 Production in Patients With Type 2 Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2018; 38:2198-2206. [DOI: 10.1161/atvbaha.118.310990] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Objective—
Treatment with liraglutide, a GLP-1 (glucagon-like peptide-1) agonist, has been shown to reduce postprandial lipidemia, an important feature of diabetic dyslipidemia. However, the underlying mechanisms for this effect remain unknown. This prompted us to study the effect of liraglutide on the metabolism of ApoB48 (apolipoprotein B48).
Approach and Results—
We performed an in vivo kinetic study with stable isotopes (D
8
-valine) in the fed state in 10 patients with type 2 diabetes mellitus before treatment and 6 months after the initiation of treatment with liraglutide (1.2 mg/d). We also evaluated, in mice, the effect of a 1-week liraglutide treatment on postload triglycerides and analysed in vitro on jejunum, the direct effect of liraglutide on the expression of genes involved in the biosynthesis of chylomicron. In diabetic patients, liraglutide treatment induced a dramatic reduction of ApoB48 pool (65±38 versus 162±87 mg;
P
=0.005) because of a significant decrease in ApoB48 production rate (3.02±1.33 versus 6.14±4.27 mg kg
-1
d
-1
;
P
=0.009) and a significant increase in ApoB48 fractional catabolic rate (5.12±1.35 versus 3.69±0.75 pool d
-1
;
P
=0.005). One-week treatment with liraglutide significantly reduced postload plasma triglycerides in mice and liraglutide, in vitro, reduced the expression of ApoB48, DGAT1 (diacylglycerol O-acyltransferase 1), and MTP (microsomal transfer protein) genes.
Conclusions—
We show that treatment with liraglutide induces a significant reduction of the ApoB48 pool because of both a reduction of ApoB48 production and an increase in ApoB48 catabolism. In vitro, liraglutide reduces the expression of genes involved in chylomicron synthesis. These effects might benefit cardiovascular health.
Clinical Trial Registration—
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT02721888.
Collapse
Affiliation(s)
- Bruno Vergès
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| | - Laurence Duvillard
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| | - Jean Paul Pais de Barros
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
- Lipidomic Analytical Platform, Bâtiment B3, Dijon, France (J.P.P.d.B.)
| | - Benjamin Bouillet
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| | - Sabine Baillot-Rudoni
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
| | - Alexia Rouland
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
| | - Anne-Laure Sberna
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
| | - Jean-Michel Petit
- From the Department of Endocrinology-Diabetology (B.V., B.B, S.B.-R., A.R., A.-L.S., J.M.P.)
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| | - Pascal Degrace
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| | - Laurent Demizieux
- Department of Biochemistry (L.D.), University Hospital, Dijon, France
- INSERM LNR UMR1231, University of Burgundy and Franche-Comté, Dijon, France (B.V., L.D., J.P.P.d.B., B.B., J.-M.P., P.D., L.D.)
| |
Collapse
|
14
|
Bertaggia E, Jensen KK, Castro-Perez J, Xu Y, Di Paolo G, Chan RB, Wang L, Haeusler RA. Cyp8b1 ablation prevents Western diet-induced weight gain and hepatic steatosis because of impaired fat absorption. Am J Physiol Endocrinol Metab 2017; 313:E121-E133. [PMID: 28377401 PMCID: PMC5582885 DOI: 10.1152/ajpendo.00409.2016] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 01/05/2023]
Abstract
Bile acids (BAs) are cholesterol derivatives that regulate lipid metabolism, through their dual abilities to promote lipid absorption and activate BA receptors. However, different BA species have varying abilities to perform these functions. Eliminating 12α-hydroxy BAs in mice via Cyp8b1 knockout causes low body weight and improved glucose tolerance. The goal of this study was to determine mechanisms of low body weight in Cyp8b1-/- mice. We challenged Cyp8b1-/- mice with a Western-type diet and assessed body weight and composition. We measured energy expenditure, fecal calories, and lipid absorption and performed lipidomic studies on feces and intestine. We investigated the requirement for dietary fat in the phenotype using a fat-free diet. Cyp8b1-/- mice were resistant to Western diet-induced body weight gain, hepatic steatosis, and insulin resistance. These changes were associated with increased fecal calories, due to malabsorption of hydrolyzed dietary triglycerides. This was reversed by treating the mice with taurocholic acid, the major 12α-hydroxylated BA species. The improvements in body weight and steatosis were normalized by feeding mice a fat-free diet. The effects of BA composition on intestinal lipid handling are important for whole body energy homeostasis. Thus modulating BA composition is a potential tool for obesity or diabetes therapy.
Collapse
Affiliation(s)
- Enrico Bertaggia
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Kristian K Jensen
- Diabetes Department, Merck Research Laboratories, Kenilworth, New Jersey; and
| | - Jose Castro-Perez
- Diabetes Department, Merck Research Laboratories, Kenilworth, New Jersey; and
| | - Yimeng Xu
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University, New York, New York
- Denali Therapeutics, Incorporated, South San Francisco, California
| | - Robin B Chan
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Liangsu Wang
- Diabetes Department, Merck Research Laboratories, Kenilworth, New Jersey; and
| | - Rebecca A Haeusler
- Department of Pathology and Cell Biology, Columbia University, New York, New York;
| |
Collapse
|
15
|
Qi J, Masucci JA, Lang W, Connelly MA, Caldwell GW, Petrounia I, Kirkpatrick J, Barnakov AN, Struble G, Miller R, Dzordzorine K, Kuo GH, Gaul M, Pocai A, Lee S. Novel LC/MS/MS and High-Throughput Mass Spectrometric Assays for Monoacylglycerol Acyltransferase Inhibitors. SLAS DISCOVERY 2017; 22:433-439. [PMID: 28328322 DOI: 10.1177/2472555217690101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoacylglycerol acyltransferase enzymes (MGAT1, MGAT2, and MGAT3) convert monoacylglycerol to diacylglycerol (DAG). MGAT1 and MGAT2 are both implicated in obesity-related metabolic diseases. Conventional MGAT enzyme assays use radioactive substrates, wherein the product of the MGAT-catalyzed reaction is usually resolved by time-consuming thin layer chromatography (TLC) analysis. Furthermore, microsomal membrane preparations typically contain endogenous diacylglycerol acyltransferase (DGAT) from the host cells, and these DGAT activities can further acylate DAG to form triglyceride (TG). Our mass spectrometry (liquid chromatography-tandem mass spectrometry, or LC/MS/MS) MGAT2 assay measures human recombinant MGAT2-catalyzed formation of didecanoyl-glycerol from 1-decanoyl-rac-glycerol and decanoyl-CoA, to produce predominantly 1,3-didecanoyl-glycerol. Unlike 1,2-DAG, 1,3-didecanoyl-glycerol is proved to be not susceptible to further acylation to TG. 1,3-Didecanoyl-glycerol product can be readily solubilized and directly subjected to high-throughput mass spectrometry (HTMS) without further extraction in a 384-well format. We also have established the LC/MS/MS MGAT activity assay in the intestinal microsomes from various species. Our assay is proved to be highly sensitive, and thus it allows measurement of endogenous MGAT activity in cell lysates and tissue preparations. The implementation of the HTMS MGAT activity assay has facilitated the robust screening and evaluation of MGAT inhibitors for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jenson Qi
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| | - John A Masucci
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Wensheng Lang
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Margery A Connelly
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Gary W Caldwell
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Ioanna Petrounia
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Jennifer Kirkpatrick
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Alexander N Barnakov
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Geoffrey Struble
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Robyn Miller
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Keli Dzordzorine
- 2 Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Gee-Hong Kuo
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Michael Gaul
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Alessandro Pocai
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Seunghun Lee
- 1 Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, Spring House, PA, USA
| |
Collapse
|
16
|
Pharmacological characterization of a series of aryl-sulfonamide derivatives that potently and selectively inhibit monoacylglycerol acyltransferase 2. Eur J Pharmacol 2016; 791:569-577. [PMID: 27658346 DOI: 10.1016/j.ejphar.2016.09.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/08/2016] [Accepted: 09/14/2016] [Indexed: 11/20/2022]
Abstract
Monoacylglycerol acyltransferase (MGAT) 2 is an endoplasmic reticulum membrane enzyme that catalyzes the synthesis of diacylglycerol (DAG) from fatty acyl-CoA and monoacylglycerol as substrates. It is important for the resynthesis of triacylglycerol in the intestine. We have identified a series of aryl-sulfonamide MGAT2 inhibitors and demonstrated pharmacological inhibition of MGAT2 improved hyperlipidemia, obesity, and diabetes in animal models. However, its mechanism of action has not been elucidated in molecular and cellular levels. In the present study, we have characterized a series of aryl-sulfonamide derivatives that potently and selectively inhibit human MGAT2 and determined their pharmacological profiles. Analyses on the molecular mechanism of a representative aryl-sulfonamide MGAT2 inhibitor revealed a reversible inhibitory activity and a binding activity to MGAT2. The aryl-sulfonamide derivatives exhibited potent inhibitory activities against both human and mouse intestinal MGAT activities, which were correlated to those determined using recombinant human and mouse MGAT enzymes. We have developed a cellular assay using Liquid Chromatography-Mass Spectrometry and confirmed that the aryl-sulfonamide derivatives suppressed DAG synthesis in the cellular context. We have thus elucidated their pharmacological profiles and provided the fundamental clues for understanding the molecular and cellular actions of the aryl-sulfonamide MGAT2 inhibitors.
Collapse
|
17
|
Qi J, Lang W, Connelly MA, Du F, Liang Y, Caldwell GW, Martin T, Hansen MK, Kuo GH, Gaul MD, Pocai A, Lee S. Metabolic tracing of monoacylglycerol acyltransferase-2 activity in vitro and in vivo. Anal Biochem 2016; 524:68-75. [PMID: 27665677 DOI: 10.1016/j.ab.2016.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 01/24/2023]
Abstract
Monoacylglycerol acyltransferase 2 (MGAT2) catalyzes the synthesis of diacylglycerol (DAG) from free fatty acids (FFA) and sn-monoacylglycerol (MG), the two major hydrolysis products of dietary fat. To demonstrate MGAT2-mediated cellular activity of triglyceride (TG) synthesis, we utilized 1-oleoyl-glycerol-d5 as a substrate to trace MGAT2-driven 1-oleoyl-glycerol-d5 incorporation into TG in HEK293 cells stably expressing human MGAT2. The oleoyl-glycerol-d5 incorporated major TG species were then quantified by liquid chromatography electrospray ionization tandem mass spectrometry (LC/ESI/MS/MS) in a 96-well format. Conventional MGAT2 target-engagement in vivo assays measure the elevation of total plasma TG by orally dosing a bolus of TG oil. We developed a novel LC/ESI/MS/MS-based fat absorption assay to assess the ability of MGAT2 inhibitors to inhibit fat absorption in CD1 mice by a meal tolerance test consisting of a mixture of liquid Boost plus® and 0.59 g/kg U13C-TG oil. The newly resynthesized plasma heavy TGs containing three 13C in the glycerol backbone and two U13C-acyl-chains, which represented the digested, absorbed and resynthesized TGs, were then quantitated by LC/ESI/MS/MS. With this assay, we identified a potent MGAT2 inhibitor that blocked MGAT2-mediated activity in vitro and in vivo. The use of 1-oleoyl-glycerol-d5 and U13C-TG oil followed by LC/ESI/MS/MS detection of stable-isotopic labeled DAG, TG, or glycerol provides a wide range of applications to study pathophysiological regulation of the monoacylglycerol pathway and MGAT2 activity.
Collapse
Affiliation(s)
- Jenson Qi
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA.
| | - Wensheng Lang
- Discovery Sciences, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Margery A Connelly
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Fuyong Du
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Yin Liang
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Gary W Caldwell
- Discovery Sciences, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Tonya Martin
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Michael K Hansen
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Gee-Hong Kuo
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Michael D Gaul
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Alessandro Pocai
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| | - Seunghun Lee
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477-0776, USA
| |
Collapse
|
18
|
D'Aquila T, Hung YH, Carreiro A, Buhman KK. Recent discoveries on absorption of dietary fat: Presence, synthesis, and metabolism of cytoplasmic lipid droplets within enterocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:730-47. [PMID: 27108063 DOI: 10.1016/j.bbalip.2016.04.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023]
Abstract
Dietary fat provides essential nutrients, contributes to energy balance, and regulates blood lipid concentrations. These functions are important to health, but can also become dysregulated and contribute to diseases such as obesity, diabetes, cardiovascular disease, and cancer. Within enterocytes, the digestive products of dietary fat are re-synthesized into triacylglycerol, which is either secreted on chylomicrons or stored within cytoplasmic lipid droplets (CLDs). CLDs were originally thought to be inert stores of neutral lipids, but are now recognized as dynamic organelles that function in multiple cellular processes in addition to lipid metabolism. This review will highlight recent discoveries related to dietary fat absorption with an emphasis on the presence, synthesis, and metabolism of CLDs within this process.
Collapse
Affiliation(s)
- Theresa D'Aquila
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Alicia Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Pharmacological Inhibition of Monoacylglycerol O-Acyltransferase 2 Improves Hyperlipidemia, Obesity, and Diabetes by Change in Intestinal Fat Utilization. PLoS One 2016; 11:e0150976. [PMID: 26938273 PMCID: PMC4777574 DOI: 10.1371/journal.pone.0150976] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 02/22/2016] [Indexed: 02/02/2023] Open
Abstract
Monoacylglycerol O-acyltransferase 2 (MGAT2) catalyzes the synthesis of diacylglycerol (DG), a triacylglycerol precursor and potential peripheral target for novel anti-obesity therapeutics. High-throughput screening identified lead compounds with MGAT2 inhibitory activity. Through structural modification, a potent, selective, and orally bioavailable MGAT2 inhibitor, compound A (compA), was discovered. CompA dose-dependently inhibited postprandial increases in plasma triglyceride (TG) levels. Metabolic flux analysis revealed that compA inhibited triglyceride/diacylglycerol resynthesis in the small intestine and increased free fatty acid and acyl-carnitine with shorter acyl chains than originally labelled fatty acid. CompA decreased high-fat diet (HFD) intake in C57BL/6J mice. MGAT2-null mice showed a similar phenotype as compA-treated mice and compA did not suppress a food intake in MGAT2 KO mice, indicating that the anorectic effects were dependent on MGAT2 inhibition. Chronic administration of compA significantly prevented body weight gain and fat accumulation in mice fed HFD. MGAT2 inhibition by CompA under severe diabetes ameliorated hyperglycemia and fatty liver in HFD-streptozotocin (STZ)-treated mice. Homeostatic model assessments (HOMA-IR) revealed that compA treatment significantly improved insulin sensitivity. The proximal half of the small intestine displayed weight gain following compA treatment. A similar phenomenon has been observed in Roux-en-Y gastric bypass-treated animals and some studies have reported that this intestinal remodeling is essential to the anti-diabetic effects of bariatric surgery. These results clearly demonstrated that MGAT2 inhibition improved dyslipidemia, obesity, and diabetes, suggesting that compA is an effective therapeutic for obesity-related metabolic disorders.
Collapse
|
20
|
da Silva RP, Kelly KB, Lewis ED, Leonard KA, Goruk S, Curtis JM, Vine DF, Proctor SD, Field CJ, Jacobs RL. Choline deficiency impairs intestinal lipid metabolism in the lactating rat. J Nutr Biochem 2015; 26:1077-83. [DOI: 10.1016/j.jnutbio.2015.04.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 10/23/2022]
|
21
|
Sato K, Takahagi H, Kubo O, Hidaka K, Yoshikawa T, Kamaura M, Nakakariya M, Amano N, Adachi R, Maki T, Take K, Takekawa S, Kitazaki T, Maekawa T. Optimization of a novel series of N-phenylindoline-5-sulfonamide-based acyl CoA:monoacylglycerol acyltransferase-2 inhibitors: Mitigation of CYP3A4 time-dependent inhibition and phototoxic liabilities. Bioorg Med Chem 2015; 23:4544-4560. [DOI: 10.1016/j.bmc.2015.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 12/20/2022]
|
22
|
Park JS, Park SB, Jeon HJ, Jung WH, Kim HY, Kang SK, Ahn JH, Rhee SD, Kim KR, Kim KY. Optimization of Enzyme Activity Assay for Screening of Acryl CoA: Monoacylglycerol Acyltransferase 2 Inhibitors. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ji Seon Park
- Alternative Toxicological Methods Research; Alternative Toxicological Methods Research Center; Daejeon 305-600 Republic of Korea
| | - Sung Bum Park
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
- Department of Toxicology, College of Pharmacy; Chungnam National University; Daejeon 305-764 Republic of Korea
| | - Hyo-Jin Jeon
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Won Hoon Jung
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Hee Youn Kim
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Seung Kyu Kang
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Jin Hee Ahn
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
- Department of Medicinal and Pharmaceutical Chemistry; University of Science and Technology; Daejeon 305-333 Republic of Korea
| | - Sang Dal Rhee
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Kwang Rok Kim
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
| | - Ki Young Kim
- Division of Drug Discovery Research; Korea Research Institute of Chemical Technology; Daejeon 305-600 Republic of Korea
- Department of Medicinal and Pharmaceutical Chemistry; University of Science and Technology; Daejeon 305-333 Republic of Korea
| |
Collapse
|
23
|
Sato K, Takahagi H, Yoshikawa T, Morimoto S, Takai T, Hidaka K, Kamaura M, Kubo O, Adachi R, Ishii T, Maki T, Mochida T, Takekawa S, Nakakariya M, Amano N, Kitazaki T. Discovery of a Novel Series of N-Phenylindoline-5-sulfonamide Derivatives as Potent, Selective, and Orally Bioavailable Acyl CoA:Monoacylglycerol Acyltransferase-2 Inhibitors. J Med Chem 2015; 58:3892-909. [DOI: 10.1021/acs.jmedchem.5b00178] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Kenjiro Sato
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroki Takahagi
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi Yoshikawa
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shinji Morimoto
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takafumi Takai
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kousuke Hidaka
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Osamu Kubo
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Ryutaro Adachi
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tsuyoshi Ishii
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Toshiyuki Maki
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Taisuke Mochida
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shiro Takekawa
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masanori Nakakariya
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Nobuyuki Amano
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomoyuki Kitazaki
- Pharmaceutical
Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
24
|
JTP-103237, a novel monoacylglycerol acyltransferase inhibitor, modulates fat absorption and prevents diet-induced obesity. Eur J Pharmacol 2015; 758:72-81. [PMID: 25857225 DOI: 10.1016/j.ejphar.2015.03.072] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 01/06/2023]
Abstract
Monoacylglycerol acyltransferase 2 (MGAT2) plays an important role in intestinal fat absorption. We discovered the novel MGAT2 inhibitor, JTP-103237, and evaluated its pharmacological profile. JTP-103237 selectively inhibited MGAT2 without remarkable species differences and reduced absorbed lipids in circulation. After lipid administration, JTP-103237 slightly but significantly decreased triglyceride content in proximal small intestine and significantly increased the lipids content in the distal small intestine. In addition, JTP-103237 significantly increased MGAT substrate (monoacylglycerol and fatty acid) content in the small intestine. JTP-103237 increased plasma peptide YY levels after lipid loading and reduced food intake in a dietary fat-dependent manner. After chronic treatment, JTP-103237 significantly decreased body weight and increased O2 consumption in the early dark phase in high fat diet induced obese (DIO) mice. Moreover, JTP-103237 improved glucose tolerance and decreased fat weight and hepatic triglyceride content in DIO mice. Our findings indicate that JTP-103237 prevents diet-induced obesity by inhibiting intestinal MGAT2 and has unique properties as a drug for the treatment of obesity.
Collapse
|
25
|
Huang B, Wang Z, Park JH, Ryu OH, Choi MK, Lee JY, Kang YH, Lim SS. Anti-diabetic effect of purple corn extract on C57BL/KsJ db/db mice. Nutr Res Pract 2015; 9:22-9. [PMID: 25671064 PMCID: PMC4317475 DOI: 10.4162/nrp.2015.9.1.22] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND/OBJECTIVES Recently, anthocyanins have been reported to have various biological activities. Furthermore, anthocyanin-rich purple corn extract (PCE) ameliorated insulin resistance and reduced diabetes-associated mesanginal fibrosis and inflammation, suggesting that it may have benefits for the prevention of diabetes and diabetes complications. In this study, we determined the anthocyanins and non-anthocyanin component of PCE by HPLC-ESI-MS and investigated its anti-diabetic activity and mechanisms using C57BL/KsJ db/db mice. MATERIALS/METHODS The db/db mice were divided into four groups: diabetic control group (DC), 10 or 50 mg/kg PCE (PCE 10 or PCE 50), or 10 mg/kg pinitol (pinitol 10) and treated with drugs once per day for 8 weeks. During the experiment, body weight and blood glucose levels were measured every week. At the end of treatment, we measured several diabetic parameters. RESULTS Compared to the DC group, Fasting blood glucose levels were 68% lower in PCE 50 group and 51% lower in the pinitol 10 group. Furthermore, the PCE 50 group showed 2- fold increased C-peptide and adiponectin levels and 20% decreased HbA1c levels, than in the DC group. In pancreatic islets morphology, the PCE- or pinitol-treated mice showed significant prevention of pancreatic β-cell damage and higher insulin content. Microarray analyses results indicating that gene and protein expressions associated with glycolysis and fatty acid metabolism in liver and fat tissues. In addition, purple corn extract increased the phosphorylation of AMP-activated protein kinase (AMPK) and decreased phosphoenolpyruvate carboxykinase (PEPCK), glucose 6-phosphatase (G6pase) genes in liver, and also increased glucose transporter 4 (GLUT4) expressions in skeletal muscle. CONCLUSIONS Our results suggested that PCE exerted anti-diabetic effects through protection of pancreatic β-cells, increase of insulin secretion and AMPK activation in the liver of C57BL/KsJ db/db mice.
Collapse
Affiliation(s)
- Bo Huang
- College of Food Science and Engineering, Liaoning Medical University, Jinzhou 121000, China
| | - Zhiqiang Wang
- Department of Food Science and Nutrition and Center for Aging and HealthCare, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon 200-702, Korea
| | - Jong Hyuk Park
- Institute of Natural Medicine, Hallym University Medical School, Gangwon 200-702, Korea
| | - Ok Hyun Ryu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University, Gangwon 200-702, Korea
| | - Moon Ki Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University, Gangwon 200-702, Korea
| | - Jae-Yong Lee
- Institute of Natural Medicine, Hallym University Medical School, Gangwon 200-702, Korea. ; Department of Biochemistry, School of Medicine, Hallym University, Gangwon 200-702, Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition and Center for Aging and HealthCare, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon 200-702, Korea
| | - Soon Sung Lim
- Institute of Natural Medicine, Hallym University Medical School, Gangwon 200-702, Korea. ; Department of Food Science and Nutrition and Center for Aging and HealthCare, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon 200-702, Korea
| |
Collapse
|
26
|
Mul JD, Begg DP, Haller AM, Pressler JW, Sorrell J, Woods SC, Farese RV, Seeley RJ, Sandoval DA. MGAT2 deficiency and vertical sleeve gastrectomy have independent metabolic effects in the mouse. Am J Physiol Endocrinol Metab 2014; 307:E1065-72. [PMID: 25315695 PMCID: PMC4254982 DOI: 10.1152/ajpendo.00376.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Vertical sleeve gastrectomy (VSG) is currently one of the most effective treatments for obesity. Despite recent developments, the underlying mechanisms that contribute to the metabolic improvements following bariatric surgery remain unresolved. VSG reduces postprandial intestinal triglyceride (TG) production, but whether the effects of VSG on intestinal metabolism are related to metabolic outcomes has yet to be established. The lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (Mogat2; MGAT2) plays a crucial role in the assimilation of dietary fat in the intestine and in regulation of adiposity stores as well. Given the phenotypic similarities between VSG-operated and MGAT2-deficient animals, we reasoned that this enzyme could also have a key role in mediating the metabolic benefits of VSG. However, VSG reduced body weight and fat mass and improved glucose metabolism similarly in whole body MGAT2-deficient (Mogat2(-/-)) mice and wild-type littermates. Furthermore, along with an increase in energy expenditure, surgically naive Mogat2(-/-) mice had altered macronutrient preference, shifting preference away from fat and toward carbohydrates, and increased locomotor activity. Collectively, these data suggest that the beneficial effects of VSG on body weight and glucose metabolism are independent of MGAT2 activity and rather that they are separate from the effects of MGAT2 deficiency. Because MGAT2 inhibitors are proposed as a pharmacotherapeutic option for obesity, our data suggest that, in addition to increasing energy expenditure, shifting macronutrient preference away from fat could be another important mechanism by which these compounds could contribute to weight loss.
Collapse
Affiliation(s)
- Joram D Mul
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio
| | - Denovan P Begg
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio; School of Psychology, University of New South Wales, Sydney, New South Wales, Australia
| | - April M Haller
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio
| | - Josh W Pressler
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio
| | - Joyce Sorrell
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio
| | - Stephen C Woods
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio
| | - Robert V Farese
- Gladstone Institute of Cardiovascular Disease, San Francisco, California; Departments of Medicine, Biochemistry, and Biophysics, University of California, San Francisco, California; and
| | - Randy J Seeley
- North Campus Research Complex, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Darleen A Sandoval
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| |
Collapse
|
27
|
Abdel-Magid AF. MoGAT-2 Inhibitors May Provide Effective Treatment for Hypertriglyceridemia. ACS Med Chem Lett 2014; 5:840-1. [PMID: 25147598 DOI: 10.1021/ml500217g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Indexed: 11/30/2022] Open
|
28
|
Zhang J, Xu D, Nie J, Cao J, Zhai Y, Tong D, Shi Y. Monoacylglycerol acyltransferase-2 is a tetrameric enzyme that selectively heterodimerizes with diacylglycerol acyltransferase-1. J Biol Chem 2014; 289:10909-10918. [PMID: 24573674 DOI: 10.1074/jbc.m113.530022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acyl-CoA:monoacylglycerol acyltransferases (MGATs) and diacylglycerol acyltransferases (DGATs) catalyze the two consecutive steps in the synthesis of triacylglycerol, a key process required for dietary fat absorption into the enterocytes of the small intestine. In this report, we investigated the tendency of MGAT2 to form an enzyme complex with DGAT1 and DGAT2 in intact cells. We demonstrated that in addition to the 38-kDa monomer of the MGAT2 enzyme predicted by its peptide sequence, a 76-kDa moiety was detected in SDS-PAGE without reducing agent and heat inactivation. The 76-kDa MGAT2 moiety was greatly enhanced by treatment with a cross-linking reagent in intact cells. Additionally, the cross-linking reagent dose-dependently yielded a band corresponding to the tetramer (152 kDa) in SDS-PAGE, suggesting that the MGAT2 enzyme primarily functions as a homotetrameric protein and as a tetrameric protein. Likewise, DGAT1 also forms a homodimer under nondenaturing conditions. When co-expressed in COS-7 cells, MGAT2 heterodimerized with DGAT1 without treatment with a cross-linking reagent. MGAT2 also co-eluted with DGAT1 on a gel filtration column, suggesting that the two enzymes form a complex in intact cells. In contrast, MGAT2 did not heterodimerize with DGAT2 when co-expressed in COS-7 cells, despite high sequence homology between the two enzymes. Furthermore, systematic deletion analysis demonstrates that N-terminal amino acids 35-80 of DGAT1, but not a signal peptide at the N terminus of MGAT2, is required for the heterodimerization. Finally, co-expression of MGAT2 with DGAT1 significantly increased lipogenesis in COS-7 cells, indicating the functional importance of the dimerization.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Dan Xu
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jia Nie
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Jingsong Cao
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Yonggong Zhai
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yuguang Shi
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033.
| |
Collapse
|
29
|
Buttet M, Traynard V, Tran TTT, Besnard P, Poirier H, Niot I. From fatty-acid sensing to chylomicron synthesis: role of intestinal lipid-binding proteins. Biochimie 2013; 96:37-47. [PMID: 23958439 DOI: 10.1016/j.biochi.2013.08.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
Today, it is well established that the development of obesity and associated diseases results, in part, from excessive lipid intake associated with a qualitative imbalance. Among the organs involved in lipid homeostasis, the small intestine is the least studied even though it determines lipid bioavailability and largely contributes to the regulation of postprandial hyperlipemia (triacylglycerols (TG) and free fatty acids (FFA)). Several Lipid-Binding Proteins (LBP) are expressed in the small intestine. Their supposed intestinal functions were initially based on what was reported in other tissues, and took no account of the physiological specificity of the small intestine. Progressively, the identification of regulating factors of intestinal LBP and the description of the phenotype of their deletion have provided new insights into cellular and molecular mechanisms involved in fat absorption. This review will discuss the physiological contribution of each LBP in the main steps of intestinal absorption of long-chain fatty acids (LCFA): uptake, trafficking and reassembly into chylomicrons (CM). Moreover, current data indicate that the small intestine is able to adapt its lipid absorption capacity to the fat content of the diet, especially through the coordinated induction of LBP. This adaptation requires the existence of a mechanism of intestinal lipid sensing. Emerging data suggest that the membrane LBP CD36 may operate as a lipid receptor that triggers an intracellular signal leading to the modulation of the expression of LBP involved in CM formation. This event could be the starting point for the optimized synthesis of large CM, which are efficiently degraded in blood. Better understanding of this intestinal lipid sensing might provide new approaches to decrease the prevalence of postprandial hypertriglyceridemia, which is associated with cardiovascular diseases, insulin resistance and obesity.
Collapse
Affiliation(s)
- Marjorie Buttet
- Physiologie de la Nutrition et Toxicologie Team (NUTox), UMR U866 INSERM, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France
| | | | | | | | | | | |
Collapse
|
30
|
Li B, Matter EK, Hoppert HT, Grayson BE, Seeley RJ, Sandoval DA. Identification of optimal reference genes for RT-qPCR in the rat hypothalamus and intestine for the study of obesity. Int J Obes (Lond) 2013; 38:192-7. [PMID: 23736358 DOI: 10.1038/ijo.2013.86] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 03/28/2013] [Accepted: 04/28/2013] [Indexed: 02/02/2023]
Abstract
BACKGROUND Obesity has a complicated metabolic pathology, and defining the underlying mechanisms of obesity requires integrative studies with molecular end points. Real-time quantitative PCR (RT-qPCR) is a powerful tool that has been widely utilized. However, the importance of using carefully validated reference genes in RT-qPCR seems to have been overlooked in obesity-related research. The objective of this study was to select a set of reference genes with stable expressions to be used for RT-qPCR normalization in rats under fasted vs re-fed and chow vs high-fat diet (HFD) conditions. DESIGN Male long-Evans rats were treated under four conditions: chow/fasted, chow/re-fed, HFD/fasted and HFD/re-fed. Expression stabilities of 13 candidate reference genes were evaluated in the rat hypothalamus, duodenum, jejunum and ileum using the ReFinder software program. The optimal number of reference genes needed for RT-qPCR analyses was determined using geNorm. RESULTS Using geNorm analysis, we found that it was sufficient to use the two most stably expressed genes as references in RT-qPCR analyses for each tissue under specific experimental conditions. B2M and RPLP0 in the hypothalamus, RPS18 and HMBS in the duodenum, RPLP2 and RPLP0 in the jejunum and RPS18 and YWHAZ in the ileum were the most suitable pairs for a normalization study when the four aforementioned experimental conditions were considered. CONCLUSIONS Our study demonstrates that gene expression levels of reference genes commonly used in obesity-related studies, such as ACTB or RPS18, are altered by changes in acute or chronic energy status. These findings underline the importance of using reference genes that are stable in expression across experimental conditions when studying the rat hypothalamus and intestine, because these tissues have an integral role in the regulation of energy homeostasis. It is our hope that this study will raise awareness among obesity researchers on the essential need for reference gene validation in gene expression studies.
Collapse
Affiliation(s)
- B Li
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - E K Matter
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - H T Hoppert
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - B E Grayson
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - R J Seeley
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - D A Sandoval
- Department of Internal Medicine, Division of Endocrinology University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
31
|
Schober G, Arnold M, Birtles S, Buckett LK, Pacheco-López G, Turnbull AV, Langhans W, Mansouri A. Diacylglycerol acyltransferase-1 inhibition enhances intestinal fatty acid oxidation and reduces energy intake in rats. J Lipid Res 2013; 54:1369-84. [PMID: 23449193 DOI: 10.1194/jlr.m035154] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Acyl CoA:diacylglycerol acyltransferase-1 (DGAT-1) catalyzes the final step in triacylglycerol (TAG) synthesis and is highly expressed in the small intestine. Because DGAT-1 knockout mice are resistant to diet-induced obesity, we investigated the acute effects of intragastric (IG) infusion of a small molecule diacylglycerol acyltransferase-1 inhibitor (DGAT-1i) on eating, circulating fat metabolites, indirect calorimetry, and hepatic and intestinal expression of key fat catabolism enzymes in male rats adapted to an 8 h feeding-16 h deprivation schedule. Also, the DGAT-1i effect on fatty acid oxidation (FAO) was investigated in enterocyte cell culture models. IG DGAT-1i infusions reduced energy intake compared with vehicle in high-fat diet (HFD)-fed rats, but scarcely in chow-fed rats. IG DGAT-1i also blunted the postprandial increase in serum TAG and increased β-hydroxybutyrate levels only in HFD-fed rats, in which it lowered the respiratory quotient and increased intestinal, but not hepatic, protein levels of Complex III of the mitochondrial respiratory chain and of mitochondrial hydroxymethylglutaryl-CoA synthase. Finally, the DGAT-1i enhanced FAO in CaCo2 (EC50 = 0.3494) and HuTu80 (EC50 = 0.00762) cells. Thus, pharmacological DGAT-1 inhibition leads to an increase in intestinal FAO and ketogenesis when dietary fat is available. This may contribute to the observed eating-inhibitory effect.
Collapse
Affiliation(s)
- Gudrun Schober
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, Zurich, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|