1
|
Wilson MH, Hensley MR, Shen MC, Lu HY, Quinlivan VH, Busch-Nentwich EM, Rawls JF, Farber SA. Zebrafish are resilient to the loss of major diacylglycerol acyltransferase enzymes. J Biol Chem 2024; 300:107973. [PMID: 39510175 PMCID: PMC11663968 DOI: 10.1016/j.jbc.2024.107973] [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: 06/04/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
In zebrafish, maternally deposited yolk is the source of nutrients for embryogenesis prior to digestive system maturation. Yolk nutrients are processed and secreted to the growing organism by an extra-embryonic tissue, the yolk syncytial layer (YSL). The export of lipids from the YSL occurs through the production of triacylglycerol-rich lipoproteins. Here we report that mutations in the triacylglycerol synthesis enzyme, diacylglycerol acyltransferase-2 (Dgat2), cause yolk sac opacity due to aberrant accumulation of cytoplasmic lipid droplets in the YSL. Although triacylglycerol synthesis continues, it is not properly coupled to lipoprotein production as dgat2 mutants produce fewer, smaller, ApoB-containing lipoproteins. Unlike DGAT2-null mice, which are lipopenic and die soon after birth, zebrafish dgat2 mutants are viable, fertile, and exhibit normal mass and adiposity. Residual Dgat activity cannot be explained by the activity of other known Dgat isoenzymes, as dgat1a;dgat1b;dgat2 triple mutants continue to produce YSL lipid droplets and remain viable as adults. Further, the newly identified diacylglycerol acyltransferase, Tmem68, is also not responsible for the residual triacylglycerol synthesis activity. Unlike overexpression of Dgat1a and Dgat1b, monoacylglycerol acyltransferase-3 (Mogat3b) overexpression does not rescue yolk opacity, suggesting it does not possess Dgat activity in the YSL. However, mogat3b;dgat2 double mutants exhibit increased yolk opacity and often have structural alterations of the yolk extension. Quadruple mogat3b;dgat1a;dgat1b;dgat2 mutants either have severely reduced viability and stunted growth or do not survive past 3 days post fertilization, depending on the dgat2 mutant allele present. Our study highlights the remarkable ability of vertebrates to synthesize triacylglycerol through multiple biosynthetic pathways.
Collapse
Affiliation(s)
- Meredith H Wilson
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | - Monica R Hensley
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | - Meng-Chieh Shen
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | - Hsiu-Yi Lu
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University, Durham, North Carolina, USA
| | - Vanessa H Quinlivan
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | | | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University, Durham, North Carolina, USA
| | - Steven A Farber
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA.
| |
Collapse
|
2
|
Xie Z, Li Y, Cheng L, Huang Y, Rao W, Shi H, Li J. Potential therapeutic strategies for MASH: from preclinical to clinical development. LIFE METABOLISM 2024; 3:loae029. [PMID: 39872142 PMCID: PMC11749562 DOI: 10.1093/lifemeta/loae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 01/03/2025]
Abstract
Current treatment paradigms for metabolic dysfunction-associated steatohepatitis (MASH) are based primarily on dietary restrictions and the use of existing drugs, including anti-diabetic and anti-obesity medications. Given the limited number of approved drugs specifically for MASH, recent efforts have focused on promising strategies that specifically target hepatic lipid metabolism, inflammation, fibrosis, or a combination of these processes. In this review, we examined the pathophysiology underlying the development of MASH in relation to recent advances in effective MASH therapy. Particularly, we analyzed the effects of lipogenesis inhibitors, nuclear receptor agonists, glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists, fibroblast growth factor mimetics, and combinatorial therapeutic approaches. We summarize these targets along with their preclinical and clinical candidates with the ultimate goal of optimizing the therapeutic prospects for MASH.
Collapse
Affiliation(s)
- Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yufeng Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Long Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yidan Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanglin Rao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Honglu Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
3
|
Meng W, Brigance R, Mignone J, Negash L, Zhao G, Ahmad S, Wang W, Moore F, Ye XY, Sun JH, Mathur A, Li YX, Azzara A, Ma Z, Chu CH, Cullen MJ, Rooney S, Harvey S, Kopcho L, Abell L, O'Malley K, Keim W, Dierks EA, Chang S, Foster KA, Harden D, Dabros M, Goti V, De Oliveira C, Krishna G, Pelleymounter MA, Whaley J, Robl JA, Cheng D, Devasthale P. Discovery of 12 (BMS-986172) as a Highly Potent MGAT2 Inhibitor that Achieved Targeted Efficacious Exposures at a Low Human Dose for the Treatment of Metabolic Disorders. J Med Chem 2023; 66:13135-13147. [PMID: 37724542 DOI: 10.1021/acs.jmedchem.3c01147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
A series of dihydropyridinone (DHP) compounds was prepared and evaluated for MGAT2 activity. The efforts led to the identification of novel tetrazolones with potent MGAT2 inhibitory activity and favorable in vitro profiles. Further tests of select analogues in mouse models revealed significant reduction in food intake and body weight. Subsequent studies in MGAT2 knockout mice with the lead candidate 12 (BMS-986172) showed on-target- and mechanism-based pharmacology. Moreover, its favorable pharmacokinetic (PK) profile and the lack of species variability in the glucuronidation potential resulted in a greater confidence level in the projection of a low dose for achieving targeted efficacious exposures in humans. Consistent with these projections, PK data from a phase 1 trial confirmed that targeted efficacious exposures could be achieved at a low dose in humans, which supported compound 12 as our second and potentially superior development candidate for the treatment of various metabolic disorders.
Collapse
|
4
|
Chalhoub G, Jamnik A, Pajed L, Kolleritsch S, Hois V, Bagaric A, Prem D, Tilp A, Kolb D, Wolinski H, Taschler U, Züllig T, Rechberger GN, Fuchs C, Trauner M, Schoiswohl G, Haemmerle G. Carboxylesterase 2a deletion provokes hepatic steatosis and insulin resistance in mice involving impaired diacylglycerol and lysophosphatidylcholine catabolism. Mol Metab 2023; 72:101725. [PMID: 37059417 PMCID: PMC10148186 DOI: 10.1016/j.molmet.2023.101725] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/16/2023] Open
Abstract
OBJECTIVE Hepatic triacylglycerol accumulation and insulin resistance are key features of NAFLD. However, NAFLD development and progression are rather triggered by the aberrant generation of lipid metabolites and signaling molecules including diacylglycerol (DAG) and lysophosphatidylcholine (lysoPC). Recent studies showed decreased expression of carboxylesterase 2 (CES2) in the liver of NASH patients and hepatic DAG accumulation was linked to low CES2 activity in obese individuals. The mouse genome encodes several Ces2 genes with Ces2a showing highest expression in the liver. Herein we investigated the role of mouse Ces2a and human CES2 in lipid metabolism in vivo and in vitro. METHODS Lipid metabolism and insulin signaling were investigated in mice lacking Ces2a and in a human liver cell line upon pharmacological CES2 inhibition. Lipid hydrolytic activities were determined in vivo and from recombinant proteins. RESULTS Ces2a deficient mice (Ces2a-ko) are obese and feeding a high-fat diet (HFD) provokes severe hepatic steatosis and insulin resistance together with elevated inflammatory and fibrotic gene expression. Lipidomic analysis revealed a marked rise in DAG and lysoPC levels in the liver of Ces2a-ko mice fed HFD. Hepatic lipid accumulation in Ces2a deficiency is linked to lower DAG and lysoPC hydrolytic activities in liver microsomal preparations. Moreover, Ces2a deficiency significantly increases hepatic expression and activity of MGAT1, a PPAR gamma target gene, suggesting aberrant lipid signaling upon Ces2a deficiency. Mechanistically, we found that recombinant Ces2a and CES2 show significant hydrolytic activity towards lysoPC (and DAG) and pharmacological inhibition of CES2 in human HepG2 cells largely phenocopies the lipid metabolic changes present in Ces2a-ko mice including reduced lysoPC and DAG hydrolysis, DAG accumulation and impaired insulin signaling. CONCLUSIONS Ces2a and CES2 are critical players in hepatic lipid signaling likely via the hydrolysis of DAG and lysoPC at the ER.
Collapse
Affiliation(s)
- Gabriel Chalhoub
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Alina Jamnik
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Laura Pajed
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Victoria Hois
- Division of Endocrinology and Diabetology, Medical University of Graz, Austria
| | - Antonia Bagaric
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Dominik Prem
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anna Tilp
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Thomas Züllig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Claudia Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schoiswohl
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria.
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| |
Collapse
|
5
|
Active Glycogen Synthase in the Liver Prevents High-Fat Diet-Induced Glucose Intolerance, Decreases Food Intake, and Lowers Body Weight. Int J Mol Sci 2023; 24:ijms24032574. [PMID: 36768897 PMCID: PMC9917303 DOI: 10.3390/ijms24032574] [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: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Many lines of evidence demonstrate a correlation between liver glycogen content and food intake. We previously demonstrated that mice overexpressing protein targeting to glycogen (PTG) specifically in the liver-which have increased glycogen content in this organ-are protected from high-fat diet (HFD)-induced obesity by reduced food intake. However, the use of PTG to increase liver glycogen implies certain limitations. PTG stimulates glycogen synthesis but also inhibits the enzyme responsible for glycogen degradation. Furthermore, as PTG is a regulatory subunit of protein phosphatase 1 (PP1), which regulates many cellular functions, its overexpression could have side effects beyond the regulation of glycogen metabolism. Therefore, it is necessary to determine whether the direct activation of glycogen synthesis, without affecting its degradation or other cellular functions, has the same effects. To this end, we generated mice overexpressing a non-inactivatable form of glycogen synthase (GS) specifically in the liver (9A-MGSAlb mice). Control and 9a-MGSAlb mice were fed a standard diet (SD) or HFD for 16 weeks. Glucose tolerance and feeding behavior were analyzed. 9A-MGSAlb mice showed an increase in hepatic glycogen in fed and fasting conditions. When fed an HFD, these animals preserved their hepatic energy state, had a reduced food intake, and presented a lower body weight and fat mass than control animals, without changes in energy expenditure. Furthermore, 9A-MGSAlb animals showed improved glucose tolerance when fed an SD or HFD. Moreover, liver triacylglycerol levels that were increased after HFD feeding were lower in these mice. These results confirm that increased liver glycogen stores contribute to decreased appetite and improve glucose tolerance in mice fed an HFD. On the basis of our findings, strategies to preserve hepatic glycogen stores emerge as potential treatments for obesity and hyperglycemia.
Collapse
|
6
|
Wang Y, Zeng F, Zhao Z, He L, He X, Pang H, Huang F, Chang P. Transmembrane Protein 68 Functions as an MGAT and DGAT Enzyme for Triacylglycerol Biosynthesis. Int J Mol Sci 2023; 24:ijms24032012. [PMID: 36768334 PMCID: PMC9916437 DOI: 10.3390/ijms24032012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Triacylglycerol (TG) biosynthesis is an important metabolic process for intracellular storage of surplus energy, intestinal dietary fat absorption, attenuation of lipotoxicity, lipid transportation, lactation and signal transduction in mammals. Transmembrane protein 68 (TMEM68) is an endoplasmic reticulum (ER)-anchored acyltransferase family member of unknown function. In the current study we show that overexpression of TMEM68 promotes TG accumulation and lipid droplet (LD) formation in a conserved active sites-dependent manner. Quantitative targeted lipidomic analysis showed that diacylglycerol (DG), free fatty acid (FFA) and TG levels were increased by TMEM68 expression. In addition, TMEM68 overexpression affected the levels of several glycerophospholipids, such as phosphatidylcholine, phosphatidylethanolamine and phosphatidylinositol, as well as sterol ester contents. TMEM68 exhibited monoacylglycerol acyltransferase (MGAT) and diacylglycerol acyltransferase (DGAT) activities dependent on the conserved active sites in an in vitro assay. The expression of lipogenesis genes, including DGATs, fatty acid synthesis-related genes and peroxisome proliferator-activated receptor γ was upregulated in TMEM68-overexpressing cells. These results together demonstrate for the first time that TMEM68 functions as an acyltransferase and affects lipogenic gene expression, glycerolipid metabolism and TG storage in mammalian cells.
Collapse
|
7
|
Tu H, Yin X, Wen J, Wu W, Zhai B, Li J, Jiang H. Glutaminase 1 blockade alleviates nonalcoholic steatohepatitis via promoting proline metabolism. Biochem Biophys Res Commun 2022; 634:1-9. [DOI: 10.1016/j.bbrc.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 09/23/2022] [Accepted: 10/01/2022] [Indexed: 12/01/2022]
|
8
|
Cheng D, Zinker BA, Luo Y, Shipkova P, De Oliveira CH, Krishna G, Brown EA, Boehm SL, Tirucherai GS, Gu H, Ma Z, Chu CH, Onorato JM, Kopcho LM, Ammar R, Smith J, Devasthale P, Lawrence RM, Stryker SA, Dierks EA, Azzara AV, Carayannopoulos L, Charles ED, Lentz KA, Gordon DA. MGAT2 inhibitor decreases liver fibrosis and inflammation in murine NASH models and reduces body weight in human adults with obesity. Cell Metab 2022; 34:1732-1748.e5. [PMID: 36323235 DOI: 10.1016/j.cmet.2022.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/14/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
Abstract
Monoacylglycerol acyltransferase 2 (MGAT2) is an important enzyme highly expressed in the human small intestine and liver for the regulation of triglyceride absorption and homeostasis. We report that treatment with BMS-963272, a potent and selective MGAT2 inhibitor, decreased inflammation and fibrosis in CDAHFD and STAM, two murine nonalcoholic steatohepatitis (NASH) models. In high-fat-diet-treated cynomolgus monkeys, in contrast to a selective diacylglycerol acyltransferase 1 (DGAT1) inhibitor, BMS-963272 did not cause diarrhea. In a Phase 1 multiple-dose trial of healthy human adults with obesity (NCT04116632), BMS-963272 was safe and well tolerated with no treatment discontinuations due to adverse events. Consistent with the findings in rodent models, BMS-963272 elevated plasma long-chain dicarboxylic acid, indicating robust pharmacodynamic biomarker modulation; increased gut hormones GLP-1 and PYY; and decreased body weight in human subjects. These data suggest MGAT2 inhibition is a promising therapeutic opportunity for NASH, a disease with high unmet medical needs.
Collapse
Affiliation(s)
- Dong Cheng
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA.
| | - Bradley A Zinker
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Yi Luo
- Translational Medicine, Bristol Myers Squibb, Lawrenceville, NJ 08543, USA
| | - Petia Shipkova
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | | | - Gopal Krishna
- ICF Early Clinical Development, Bristol Myers Squibb, Summit, NJ 07901, USA
| | - Elizabeth A Brown
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Stephanie L Boehm
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | | | - Huidong Gu
- Translational Medicine, Bristol Myers Squibb, Lawrenceville, NJ 08543, USA
| | - Zhengping Ma
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Ching-Hsuen Chu
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Joelle M Onorato
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Lisa M Kopcho
- Leads Discovery and Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Ron Ammar
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Julia Smith
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Pratik Devasthale
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - R Michael Lawrence
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Steven A Stryker
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Elizabeth A Dierks
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - Anthony V Azzara
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | | | - Edgar D Charles
- Global Drug Development, Bristol Myers Squibb, Lawrenceville, NJ 08543, USA
| | - Kimberley A Lentz
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ 08543, USA
| | - David A Gordon
- Departments of Discovery Biology Cardiovascular and Fibrosis, Bristol Myers Squibb, Princeton, NJ 08543, USA
| |
Collapse
|
9
|
Singer JM, Shew TM, Ferguson D, Renkemeyer MK, Pietka TA, Hall AM, Finck BN, Lutkewitte AJ. Monoacylglycerol O-acyltransferase 1 lowers adipocyte differentiation capacity in vitro but does not affect adiposity in mice. Obesity (Silver Spring) 2022; 30:2122-2133. [PMID: 36321276 PMCID: PMC9634674 DOI: 10.1002/oby.23538] [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: 02/16/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Monoacylglycerol O-acyltransferase 1 (Mogat1), a lipogenic enzyme that converts monoacylglycerol to diacylglycerol, is highly expressed in adipocytes and may regulate lipolysis by re-esterifying fatty acids released during times when lipolytic rates are low. However, the role of Mogat1 in regulating adipocyte fat storage during differentiation and diet-induced obesity is relatively understudied. METHODS Here, adipocyte-specific Mogat1 knockout mice were generated and subjected to a high-fat diet to determine the effects of Mogat1 deficiency on diet-induced obesity. Mogat1 floxed mice were also used to develop preadipocyte cell lines wherein Mogat1 could be conditionally knocked out to study adipocyte differentiation in vitro. RESULTS In preadipocytes, it was found that Mogat1 knockout at the onset of preadipocyte differentiation prevented the accumulation of glycerolipids and reduced the differentiation capacity of preadipocytes. However, the loss of adipocyte Mogat1 did not affect weight gain or fat mass induced by a high-fat diet in mice. Furthermore, loss of Mogat1 in adipocytes did not affect plasma lipid or glucose concentrations or insulin tolerance. CONCLUSIONS These data suggest Mogat1 may play a role in adipocyte differentiation in vitro but not adipose tissue expansion in response to nutrient overload in mice.
Collapse
Affiliation(s)
- Jason M. Singer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Trevor M. Shew
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Daniel Ferguson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - M. Katie Renkemeyer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Terri A. Pietka
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Angela M. Hall
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Brian N. Finck
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Andrew J. Lutkewitte
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
10
|
Han H, Wang M, Zhong R, Yi B, Schroyen M, Zhang H. Depletion of Gut Microbiota Inhibits Hepatic Lipid Accumulation in High-Fat Diet-Fed Mice. Int J Mol Sci 2022; 23:ijms23169350. [PMID: 36012616 PMCID: PMC9408850 DOI: 10.3390/ijms23169350] [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: 07/27/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 02/07/2023] Open
Abstract
Dysregulated lipid metabolism is a key pathology in metabolic diseases and the liver is a critical organ for lipid metabolism. The gut microbiota has been shown to regulate hepatic lipid metabolism in the host. However, the underlying mechanism by which the gut microbiota influences hepatic lipid metabolism has not been elucidated. Here, a gut microbiota depletion mouse model was constructed with an antibiotics cocktail (Abx) to study the mechanism through which intestinal microbiota regulates hepatic lipid metabolism in high-fat diet (HFD)-fed mice. Our results showed that the Abx treatment effectively eradicated the gut microbiota in these mice. Microbiota depletion reduced the body weight and fat deposition both in white adipose tissue and liver. In addition, microbiota depletion reduced serum levels of glucose, total cholesterol (TC), low-density lipoproteins (LDL), insulin, and leptin in HFD-fed mice. Importantly, the depletion of gut microbiota in HFD-fed mice inhibited excessive hepatic lipid accumulation. Mechanistically, RNA-seq results revealed that gut microbiota depletion changed the expression of hepatic genes involved in cholesterol and fatty acid metabolism, such as Cd36, Mogat1, Cyp39a1, Abcc3, and Gpat3. Moreover, gut microbiota depletion reduced the abundance of bacteria associated with abnormal metabolism and inflammation, including Lachnospiraceae, Coriobacteriaceae_UCG-002, Enterorhabdus, Faecalibaculum, and Desulfovibrio. Correlation analysis showed that there was strong association between the altered gut microbiota abundance and the serum cholesterol level. This study indicates that gut microbiota ameliorates HFD-induced hepatic lipid metabolic dysfunction, which might be associated with genes participating in cholesterol and fatty acid metabolism in the liver.
Collapse
Affiliation(s)
- Hui Han
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, 4000 Gembloux, Belgium
| | - Mengyu Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bao Yi
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence:
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, 4000 Gembloux, Belgium
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
11
|
Gökçe G, Bayraktar M. Assessment of the association of the MOGAT1 and MOGAT3 gene with growth traits in different growth stages in Holstein calves. Arch Anim Breed 2022; 65:301-308. [PMID: 36035878 PMCID: PMC9400126 DOI: 10.5194/aab-65-301-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/22/2022] [Indexed: 11/11/2022] Open
Abstract
The members of the monoacylglycerol acyltransferase (MOGAT) family are essential candidate genes that influence economic traits associated with triglyceride synthesis, dietary fat absorption, and storage in livestock. In addition, the MOGAT gene family may also play an essential function in human polygenic diseases, like type 2 diabetes and obesity. The present study was conducted on Holstein calves to find the association between MOGAT1, MOGAT3/g.A229G, and MOGAT3/g.G1627A and growth traits. The polymerase chain reaction–restriction fragment length polymorphism (PCR-RFLP) method was performed for genotyping the MOGAT1, MOGAT3/g.A229G, and MOGAT3/g.G1627A genes' locus using the TaqI, MspI, and BsuRI restriction enzyme. The allele frequency of A and G of the MOGAT1 locus was 0.79 and 0.21, respectively, while the genotype frequency was 0.65, 0.28, and 0.07 for AA, AG, and GG, respectively. While the allele and genotype frequencies of the MOGAT3/g.A229G locus were 00.57(A1), 0.43(G1), 0.35(A1A1), 0.45(A1G1), and 0.20(G1G1), the allele and genotype frequencies of the MOGAT3/g.G1627A locus were 0.49(A2), 0.51(G2), 0.25(A2A2), 0.49(A2G2), and 0.26(G2G2). Chi-square analysis showed that MOGAT3/g.G1627A distribution was at the Hardy–Weinberg disequilibrium (p < 0.05), and MOGAT1 and MOGAT3/g.A229G distribution was at the Hardy–Weinberg equilibrium (p > 0.05). In total, two statistical methods (general linear model (GLM) and PROC MIXED) were used to identify an association between gene locus and growth traits. An association analysis showed a statistically significant difference between the MOGAT1 and body weight, body length, and chest circumference, MOGAT3/g.A229G with average daily gain (ADG) and withers height, and MOGAT3/g.G1627A with body weight and body length (p < 0.05). The results confirmed that the MOGAT1, MOGAT3/g.A229G, and MOGAT3/g.G1627A locus are strong candidate genes that could be considered molecular markers for growth traits in cattle breeding.
Collapse
|
12
|
Chen G, Harwood JL, Lemieux MJ, Stone SJ, Weselake RJ. Acyl-CoA:diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog Lipid Res 2022; 88:101181. [PMID: 35820474 DOI: 10.1016/j.plipres.2022.101181] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the last reaction in the acyl-CoA-dependent biosynthesis of triacylglycerol (TAG). DGAT activity resides mainly in membrane-bound DGAT1 and DGAT2 in eukaryotes and bifunctional wax ester synthase-diacylglycerol acyltransferase (WSD) in bacteria, which are all membrane-bound proteins but exhibit no sequence homology to each other. Recent studies also identified other DGAT enzymes such as the soluble DGAT3 and diacylglycerol acetyltransferase (EaDAcT), as well as enzymes with DGAT activities including defective in cuticular ridges (DCR) and steryl and phytyl ester synthases (PESs). This review comprehensively discusses research advances on DGATs in prokaryotes and eukaryotes with a focus on their biochemical properties, physiological roles, and biotechnological and therapeutic applications. The review begins with a discussion of DGAT assay methods, followed by a systematic discussion of TAG biosynthesis and the properties and physiological role of DGATs. Thereafter, the review discusses the three-dimensional structure and insights into mechanism of action of human DGAT1, and the modeled DGAT1 from Brassica napus. The review then examines metabolic engineering strategies involving manipulation of DGAT, followed by a discussion of its therapeutic applications. DGAT in relation to improvement of livestock traits is also discussed along with DGATs in various other eukaryotic organisms.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada.
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Membrane Protein Disease Research Group, Edmonton T6G 2H7, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Randall J Weselake
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada
| |
Collapse
|
13
|
The monoacylglycerol acyltransferase pathway contributes to triacylglycerol synthesis in HepG2 cells. Sci Rep 2022; 12:4943. [PMID: 35322811 PMCID: PMC8943211 DOI: 10.1038/s41598-022-08946-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
The monoacylglycerol acyltransferase (MGAT) pathway has a well-established role in the small intestine where it facilitates the absorption of dietary fat. In enterocytes, MGAT participates in the resynthesis of triacylglycerol using substrates (monoacylglycerol and fatty acids) generated in the gut lumen from the breakdown of triacylglycerol consumed in the diet. MGAT activity is also present in the liver, but its role in triacylglycerol metabolism in this tissue remains unclear. The predominant MGAT isoforms present in human liver appear to be MGAT2 and MGAT3. The objective of this study was to use selective small molecule inhibitors of MGAT2 and MGAT3 to determine the contributions of these enzymes to triacylglycerol production in liver cells. We found that pharmacological inhibition of either enzyme had no effect on TG mass in HepG2 cells but did alter lipid droplet size and number. Inhibition of MGAT2 did result in decreased DG and TG synthesis and TG secretion. Interestingly, MGAT2 preferentially utilized 2-monoacylglycerol derived from free glycerol and not from exogenously added 2-monoacylglycerol. In contrast, inhibition of MGAT3 had very little effect on TG metabolism in HepG2 cells. Additionally, we demonstrated that the MGAT activity of DGAT1 only makes a minor contribution to TG synthesis in intact HepG2 cells. Our data demonstrated that the MGAT pathway has a role in hepatic lipid metabolism with MGAT2, more so than MGAT3, contributing to TG synthesis and secretion.
Collapse
|
14
|
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: 1.7] [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
|
15
|
Paul B, Lewinska M, Andersen JB. Lipid alterations in chronic liver disease and liver cancer. JHEP Rep 2022; 4:100479. [PMID: 35469167 PMCID: PMC9034302 DOI: 10.1016/j.jhepr.2022.100479] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are a complex and diverse group of molecules with crucial roles in many physiological processes, as well as in the onset, progression, and maintenance of cancers. Fatty acids and cholesterol are the building blocks of lipids, orchestrating these crucial metabolic processes. In the liver, lipid alterations are prevalent as a cause and consequence of chronic hepatitis B and C virus infections, alcoholic hepatitis, and non-alcoholic fatty liver disease and steatohepatitis. Recent developments in lipidomics have also revealed that dynamic changes in triacylglycerols, phospholipids, sphingolipids, ceramides, fatty acids, and cholesterol are involved in the development and progression of primary liver cancer. Accordingly, the transcriptional landscape of lipid metabolism suggests a carcinogenic role of increasing fatty acids and sterol synthesis. However, limited mechanistic insights into the complex nature of the hepatic lipidome have so far hindered the development of effective therapies.
Collapse
|
16
|
Zhang E, Zhao Y, Hu H. Impact of Sodium Glucose Cotransporter 2 Inhibitors on Nonalcoholic Fatty Liver Disease Complicated by Diabetes Mellitus. Hepatol Commun 2021; 5:736-748. [PMID: 34027265 PMCID: PMC8122372 DOI: 10.1002/hep4.1611] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023] Open
Abstract
Sodium glucose cotransporter 2 (SGLT2), a type of membrane protein highly expressed in the kidney, can regulate plasma glucose through the glomerular filtration process by reabsorption from the kidney. SGLT2 inhibitors, which are newly developed oral antidiabetic drugs, can play a role in liver diseases by inhibiting SGLT2-mediated renal glucose reabsorption and inducing glycosuria. Nonalcoholic fatty liver disease (NAFLD) is the most common type of liver disease, resulting in severe liver dysfunction. During the progression of NAFLD, there are some hallmark complications, including lipid metabolism disorders, inflammation induction, and hepatocyte death. Herein, we review several SGLT2 inhibitors that are capable of protecting individuals with NAFLD from severe complications by inhibiting de novo lipogenesis, oxidative responses, inflammation induction, and hepatocyte death.
Collapse
Affiliation(s)
- Enxiang Zhang
- Key Laboratory of Growth Regulation and Transformation Research of Zhejiang ProvinceSchool of Life SciencesWestlake Institute for Advanced StudyWestlake UniversityShilongshanHangzhouChina.,Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Food Science and Nutritional EngineeringChina Agricultural UniversityBeijingChina.,Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaMinneapolisMN
| | - Yang Zhao
- Department of CardiologyZhejiang Provincial People's HospitalHangzhouChina.,Cardiovascular DivisionDepartment of MedicineUniversity of MinnesotaMinneapolisMN
| | - Hongbo Hu
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Food Science and Nutritional EngineeringChina Agricultural UniversityBeijingChina
| |
Collapse
|
17
|
Lutkewitte AJ, Singer JM, Shew TM, Martino MR, Hall AM, He M, Finck BN. Multiple antisense oligonucleotides targeted against monoacylglycerol acyltransferase 1 (Mogat1) improve glucose metabolism independently of Mogat1. Mol Metab 2021; 49:101204. [PMID: 33676028 PMCID: PMC8027266 DOI: 10.1016/j.molmet.2021.101204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 03/01/2021] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE Monoacylglycerol acyltransferase (MGAT) enzymes catalyze the synthesis of diacylglycerol from monoacylglycerol. Previous work has suggested the importance of MGAT activity in the development of obesity-related hepatic insulin resistance. Indeed, antisense oligonucleotide (ASO)-mediated knockdown of Mogat1 mRNA, which encodes MGAT1, reduced hepatic MGAT activity and improved glucose tolerance and insulin resistance in high-fat diet (HFD)-fed mice. However, recent work has suggested that some ASOs may have off-target effects on body weight and metabolic parameters via activation of the interferon alpha/beta receptor 1 (IFNAR-1) pathway. METHODS Mice with whole-body Mogat1 knockout or a floxed allele for Mogat1 to allow for liver-specific Mogat1-knockout (by either a liver-specific transgenic or adeno-associated virus-driven Cre recombinase) were generated. These mice were placed on an HFD, and glucose metabolism and insulin sensitivity were assessed after 16 weeks on diet. In some experiments, mice were treated with control scramble or Mogat1 ASOs in the presence or absence of IFNAR-1 neutralizing antibody. RESULTS Genetic deletion of hepatic Mogat1, either acutely or chronically, did not improve hepatic steatosis, glucose tolerance, or insulin sensitivity in HFD-fed mice. Furthermore, constitutive Mogat1 knockout in all tissues actually exacerbated HFD-induced obesity, insulin sensitivity, and glucose intolerance on an HFD. Despite markedly reduced Mogat1 expression, liver MGAT activity was unaffected in all knockout mouse models. Mogat1 overexpression in hepatocytes increased liver MGAT activity and TAG content in low-fat-fed mice but did not cause insulin resistance. Multiple Mogat1 ASO sequences improved glucose tolerance in both wild-type and Mogat1 null mice, suggesting an off-target effect. Hepatic IFNAR-1 signaling was activated by multiple Mogat1 ASOs, but its blockade did not prevent the effects of either Mogat1 ASO on glucose homeostasis. CONCLUSION These results indicate that genetic loss of Mogat1 does not affect hepatic MGAT activity or metabolic homeostasis on HFD and show that multiple Mogat1 ASOs improve glucose metabolism through effects independent of targeting Mogat1 or activation of IFNAR-1 signaling.
Collapse
Affiliation(s)
- Andrew J Lutkewitte
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Jason M Singer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Trevor M Shew
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael R Martino
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Angela M Hall
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
18
|
Liss KH, Ek SE, Lutkewitte AJ, Pietka TA, He M, Skaria P, Tycksen E, Ferguson D, Blanc V, Graham MJ, Hall AM, McGill MR, McCommis KS, Finck BN. Monoacylglycerol Acyltransferase 1 Knockdown Exacerbates Hepatic Ischemia/Reperfusion Injury in Mice With Hepatic Steatosis. Liver Transpl 2021; 27:116-133. [PMID: 32916011 PMCID: PMC7785593 DOI: 10.1002/lt.25886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming the most common indication for liver transplantation. The growing prevalence of NAFLD not only increases the demand for liver transplantation, but it also limits the supply of available organs because steatosis predisposes grafts to ischemia/reperfusion injury (IRI) and many steatotic grafts are discarded. We have shown that monoacylglycerol acyltransferase (MGAT) 1, an enzyme that converts monoacylglycerol to diacylglycerol, is highly induced in animal models and patients with NAFLD and is an important mediator in NAFLD-related insulin resistance. Herein, we sought to determine whether Mogat1 (the gene encoding MGAT1) knockdown in mice with hepatic steatosis would reduce liver injury and improve liver regeneration following experimental IRI. Antisense oligonucleotides (ASO) were used to knockdown the expression of Mogat1 in a mouse model of NAFLD. Mice then underwent surgery to induce IRI. We found that Mogat1 knockdown reduced hepatic triacylglycerol accumulation, but it unexpectedly exacerbated liver injury and mortality following experimental ischemia/reperfusion surgery in mice on a high-fat diet. The increased liver injury was associated with robust effects on the hepatic transcriptome following IRI including enhanced expression of proinflammatory cytokines and chemokines and suppression of enzymes involved in intermediary metabolism. These transcriptional changes were accompanied by increased signs of oxidative stress and an impaired regenerative response. We have shown that Mogat1 knockdown in a mouse model of NAFLD exacerbates IRI and inflammation and prolongs injury resolution, suggesting that Mogat1 may be necessary for liver regeneration following IRI and that targeting this metabolic enzyme will not be an effective treatment to reduce steatosis-associated graft dysfunction or failure.
Collapse
Affiliation(s)
- Kim H.H. Liss
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Shelby E. Ek
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Terri A. Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Priya Skaria
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Eric Tycksen
- Department of Genome Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO
| | - Daniel Ferguson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Valerie Blanc
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Angela M. Hall
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mitchell R. McGill
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Kyle S. McCommis
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO
| | - Brian N. Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
Dayarathne LA, Ranaweera SS, Natraj P, Rajan P, Lee YJ, Han CH. Restoration of the adipogenic gene expression by naringenin and naringin in 3T3-L1 adipocytes. J Vet Sci 2021; 22:e55. [PMID: 34313040 PMCID: PMC8318791 DOI: 10.4142/jvs.2021.22.e55] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/29/2021] [Accepted: 06/20/2021] [Indexed: 11/23/2022] Open
Abstract
Background Naringenin and its glycoside naringin are well known citrus flavonoids with several therapeutic benefits. Although the anti-adipogenic effects of naringenin and naringin have been reported previously, the detailed mechanism underlying their anti-adipogenesis effects is poorly understood. Objectives This study examined the anti-adipogenic effects of naringenin and naringin by determining differential gene expression patterns in these flavonoids-treated 3T3-L1 adipocytes. Methods Lipid accumulation and triglyceride (TG) content were determined by Oil red O staining and TG assay. Glucose uptake was measured using a 2-[N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxy-d-glucose fluorescent d-glucose analog. The phosphorylation levels of AMP-activated protein kinase (AMPK) and acetyl Co-A carboxylase (ACC) were observed via Western blot analysis. Differential gene expressions in 3T3-L1 adipocytes were evaluated via RNA sequencing analysis. Results Naringenin and naringin inhibited both lipid accumulation and TG content, increased phosphorylation levels of both AMPK and ACC and decreased the expression level of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) in 3T3-L1 adipocytes. RNA sequencing analysis revealed that 32 up-regulated (> 2-fold) and 17 down-regulated (< 0.6-fold) genes related to lipid metabolism, including Acaca, Fasn, Scd1, Mogat1, Dgat, Lipin1, Cpt1a, and Lepr, were normalized to the control level in naringenin-treated adipocytes. In addition, 25 up-regulated (> 2-fold) and 25 down-regulated (< 0.6-fold) genes related to lipid metabolism, including Acaca, Fasn, Fabp5, Scd1, Srebf1, Hmgcs1, Cpt1c, Lepr, and Lrp1, were normalized to the control level by naringin. Conclusions The results indicate that naringenin and naringin have anti-adipogenic potentials that are achieved by normalizing the expression levels of lipid metabolism-related genes that were perturbed in differentiated 3T3-L1 cells.
Collapse
Affiliation(s)
| | | | - Premkumar Natraj
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Priyanka Rajan
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Young Jae Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Chang Hoon Han
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
20
|
Sato Y. Development of Lipid Nanoparticles for the Delivery of Macromolecules Based on the Molecular Design of pH-Sensitive Cationic Lipids. Chem Pharm Bull (Tokyo) 2021; 69:1141-1159. [PMID: 34853281 DOI: 10.1248/cpb.c21-00705] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Considerable efforts have been made on the development of lipid nanoparticles (LNPs) for delivering of nucleic acids in LNP-based medicines, including a first-ever short interfering RNA (siRNA) medicine, Onpattro, and the mRNA vaccines against the coronavirus disease 2019 (COVID-19), which have been approved and are currently in use worldwide. The successful rational design of ionizable cationic lipids was a major breakthrough that dramatically increased delivery efficiency in this field. The LNPs would be expected to be useful as a platform technology for the delivery of various therapeutic modalities for genome editing and even for undiscovered therapeutic mechanisms. In this review, the current progress of my research, including the molecular design of pH-sensitive cationic lipids, their applications for various tissues and cell types, and for delivering various macromolecules, including siRNA, antisense oligonucleotide, mRNA, and the clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) system will be described. Mechanistic studies regarding relationships between the physicochemical properties of LNPs, drug delivery, and biosafety are also summarized. Furthermore, current issues that need to be addressed for next generation drug delivery systems are discussed.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
21
|
Dyleva YA, Gruzdeva OV. [MicroRNA and obesity. A modern view of the problem (review of literature).]. Klin Lab Diagn 2020; 65:411-417. [PMID: 32762178 DOI: 10.18821/0869-2084-2020-65-7-411-417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The incidence of obesity is steadily increasing worldwide, reaching the epidemic. Obesity is associated with cardiometabolic diseases through the complex interactions between genetics and epigenetics predisposition, the environment, diet, and lifestyle. However, the molecular mechanisms and factors influencing these processes are not fully known. MicroRNAs are a new class of important regulatory determinants in many biological and pathological processes. There is increasing evidence of the role of miRNAs in the regulation of the functional activity of adipose tissue and the development of obesity. A change in the expression of MicroRNAs can lead to changes in the activity of genes that control a number of biological processes, including inflammation, lipid metabolism, and adipogenesis. Understanding the role of miRNAs in the regulation of adipogenesis and the development of obesity will establish therapeutic targets for the development of new and effective drugs, which will lead to a breakthrough in the fight against obesity and related diseases. This review presents current data on the role of miRNAs in the regulation of the functional activity of adipose tissue, including adipogenesis of white, beige and brown adipocytes, as well as the prerequisites for using miRNAs as biomarkers of obesity and the possibility of therapeutic use.
Collapse
Affiliation(s)
- Yu A Dyleva
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, 650002, Kemerovo, Russian Federation
| | - O V Gruzdeva
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, 650002, Kemerovo, Russian Federation
| |
Collapse
|
22
|
Molecular mechanisms of hepatic insulin resistance in nonalcoholic fatty liver disease and potential treatment strategies. Pharmacol Res 2020; 159:104984. [PMID: 32502637 DOI: 10.1016/j.phrs.2020.104984] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) in the general population is estimated at 25 %, and there is currently no effective treatment of NAFLD. Although insulin resistance (IR) is not the only factor causing the pathogenesis of NAFLD, hepatic IR has a cause-effective relationship with NAFLD. Improving hepatic IR is a potential therapeutic strategy to treat NAFLD. This review highlights the molecular mechanisms of hepatic IR in the development of NAFLD. Available data on potential drugs including glucagon-like peptide 1 receptor (GLP-1) agonists, peroxisome proliferator-activated receptor (PPAR-γ/α/δ) agonists, farnesoid X receptor (FXR) agonists, etc. are carefully discussed.
Collapse
|
23
|
de Conti A, Tryndyak V, Willett RA, Borowa-Mazgaj B, Watson A, Patton R, Khare S, Muskhelishvili L, Olson GR, Avigan MI, Cerniglia CE, Ross SA, Sanyal AJ, Beland FA, Rusyn I, Pogribny IP. Characterization of the variability in the extent of nonalcoholic fatty liver induced by a high-fat diet in the genetically diverse Collaborative Cross mouse model. FASEB J 2020; 34:7773-7785. [PMID: 32304142 DOI: 10.1096/fj.202000194r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Interindividual variability and sexual dimorphisms in the development of nonalcoholic fatty liver disease (NAFLD) are still poorly understood. In the present study, male and female strains of Collaborative Cross (CC) mice were fed a high-fat and high-sucrose (HF/HS) diet or a control diet for 12 weeks to investigate interindividual- and sex-specific variations in the development of NAFLD. The severity of liver steatosis varied between sexes and individual strains and was accompanied by an elevation of serum markers of insulin resistance, including increases in total cholesterol, low-density lipoproteins, high-density lipoproteins, phospholipids, and glucose. The development of NAFLD was associated with overexpression of the critical fatty acid uptake and de novo lipogenesis genes Pparg, Mogat1, Cd36, Acaab1, Fabp2, and Gdf15 in male and female mice. The expression of Pparg, Mogat1, and Cd36 was positively correlated with liver triglycerides in male mice, and Mogat1 and Cd36 expression were positively correlated with liver triglycerides in female mice. Our results indicate the value of CC mice in combination with HF/HS diet-induced alterations as an approach to study the susceptibility and interindividual variabilities in the pathogenesis of nonalcoholic fatty liver and early nonalcoholic steatohepatitis at the population level, uncovering of susceptible and resistant cohorts, and identifying sex-specific molecular determinants of disease susceptibility.
Collapse
Affiliation(s)
- Aline de Conti
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Volodymyr Tryndyak
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Rose A Willett
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Barbara Borowa-Mazgaj
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Anna Watson
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Ralph Patton
- Toxicologic Pathology Associates, Jefferson, AR, USA
| | - Sangeeta Khare
- Division of Microbiology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | | | - Greg R Olson
- Toxicologic Pathology Associates, Jefferson, AR, USA
| | - Mark I Avigan
- Office of Pharmacovigilance and Epidemiology, FDA-Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Carl E Cerniglia
- Division of Microbiology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Sharon A Ross
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Igor P Pogribny
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| |
Collapse
|
24
|
The Vitamin D Receptor Regulates Glycerolipid and Phospholipid Metabolism in Human Hepatocytes. Biomolecules 2020; 10:biom10030493. [PMID: 32213983 PMCID: PMC7175212 DOI: 10.3390/biom10030493] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
The vitamin D receptor (VDR) must be relevant to liver lipid metabolism because VDR deficient mice are protected from hepatosteatosis. Therefore, our objective was to define the role of VDR on the overall lipid metabolism in human hepatocytes. We developed an adenoviral vector for human VDR and performed transcriptomic and metabolomic analyses of cultured human hepatocytes upon VDR activation by vitamin D (VitD). Twenty percent of the VDR responsive genes were related to lipid metabolism, including MOGAT1, LPGAT1, AGPAT2, and DGAT1 (glycerolipid metabolism); CDS1, PCTP, and MAT1A (phospholipid metabolism); and FATP2, SLC6A12, and AQP3 (uptake of fatty acids, betaine, and glycerol, respectively). They were rapidly induced (4–6 h) upon VDR activation by 10 nM VitD or 100 µM lithocholic acid (LCA). Most of these genes were also upregulated by VDR/VitD in mouse livers in vivo. Ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS) metabolomics demonstrated intracellular accumulation of triglycerides, with concomitant decreases in diglycerides and phosphatidates, at 8 and 24 h upon VDR activation. Significant alterations in phosphatidylcholines, increases in lyso-phosphatidylcholines and decreases in phosphatidylethanolamines and phosphatidylethanolamine plasmalogens were also observed. In conclusion, active VitD/VDR signaling in hepatocytes triggers an unanticipated coordinated gene response leading to triglyceride synthesis and to important perturbations in glycerolipids and phospholipids.
Collapse
|
25
|
Ko CW, Qu J, Black DD, Tso P. Regulation of intestinal lipid metabolism: current concepts and relevance to disease. Nat Rev Gastroenterol Hepatol 2020; 17:169-183. [PMID: 32015520 DOI: 10.1038/s41575-019-0250-7] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Lipids entering the gastrointestinal tract include dietary lipids (triacylglycerols, cholesteryl esters and phospholipids) and endogenous lipids from bile (phospholipids and cholesterol) and from shed intestinal epithelial cells (enterocytes). Here, we comprehensively review the digestion, uptake and intracellular re-synthesis of intestinal lipids as well as their packaging into pre-chylomicrons in the endoplasmic reticulum, their modification in the Golgi apparatus and the exocytosis of the chylomicrons into the lamina propria and subsequently to lymph. We also discuss other fates of intestinal lipids, including intestinal HDL and VLDL secretion, cytosolic lipid droplets and fatty acid oxidation. In addition, we highlight the applicability of these findings to human disease and the development of therapeutics targeting lipid metabolism. Finally, we explore the emerging role of the gut microbiota in modulating intestinal lipid metabolism and outline key questions for future research.
Collapse
Affiliation(s)
- Chih-Wei Ko
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jie Qu
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Dennis D Black
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
26
|
Park SO, Zammit VA. In vivo monitoring of hepatic glycolipid distribution of n-6 ∕ n-3 in jugular-vein-cannulated rats as a nutritional research model for monogastric animal. Arch Anim Breed 2019; 62:437-446. [PMID: 31807655 PMCID: PMC6852779 DOI: 10.5194/aab-62-437-2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The metabolic distribution via blood from liver of glycerolipids by omega-6
to omega-3 fatty acid (n-6 / n-3) ratio in monogastric animal nutrition is
very important. In vivo monitoring technique using jugular-vein-cannulated
rats as a nutritional model for monogastric animal can yield important
insights into animal nutrition. This study was conducted to determine the
effect of different n-6 / n-3 ratios (71:1, 4:1, 15:1, 30:1) on
metabolic distribution of glycerolipids newly synthesized and secreted in
the liver of the rats and explore the mechanism involved. Regarding
14CO2 released from oxidation of glycerolipid metabolism, it was
the highest (22.5 %) in groups with a n-6 / n-3 ratio of 4:1 (P<0.05).
The control group showed the highest total glycerolipid level, followed by the
30:1, 15:1, and 4:1 groups in order (P<0.05). When secreted
triacylglycerol level of each group was compared with that of the control
group, the 4:1, 15:1, and 30:1 groups were decreased by 36.3 %, 20.9 %, and
13.3 %, respectively (P<0.05). Regarding the distribution of
phospholipid against total glycerolipid compared to the control group, the 4:1, 15:1, and 30:1 groups were 1.38, 1.29, and 1.17 times higher, respectively
(P<0.05). In the comparison of 14CO2 emission against
total glycerolipid compared with the control group, the 4:1, 15:1, and 30:1
groups were 1.61, 1.52, and 1.29 times higher, respectively
(P<0.05). These results demonstrate that a dietary n-6 / n-3 fatty acid
ratio of 4:1 could significantly decrease harmful lipid levels in the blood
by controlling the mechanism of metabolic distribution via blood from
triglyceride and phospholipid newly synthesized in the liver of cannulated
rat.
Collapse
Affiliation(s)
- Sang-O Park
- College of Animal Life Science, Kangwon National University, Chuncheon, Gangwon-do, 24419 Republic of Korea
| | - Victor A Zammit
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
27
|
Shi L, Liu L, Lv X, Ma Z, Yang Y, Li Y, Zhao F, Sun D, Han B. Polymorphisms and genetic effects of PRLR, MOGAT1, MINPP1 and CHUK genes on milk fatty acid traits in Chinese Holstein. BMC Genet 2019; 20:69. [PMID: 31419940 PMCID: PMC6698030 DOI: 10.1186/s12863-019-0769-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 08/06/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Our initial genome-wide association study (GWAS) identified 20 promising candidate genes for milk fatty acid (FA) traits in a Chinese Holstein population, including PRLR, MOGAT1, MINPP1 and CHUK genes. In this study, we performed whether they had significant genetic effects on milk FA traits in Chinese Holstein. RESULTS We re-sequenced the entire exons and 3000 bp of the 5' and 3' flanking regions, and identified 11 single nucleotide polymorphisms (SNPs), containing four in PRLR, two in MOGAT1, two in MINPP1, and three in CHUK. The SNP-based association analyses showed that all the 11 SNPs were significantly associated with at least one milk FA trait (P = 0.0456 ~ < 0.0001), and none of them had association with C11:0, C13:0, C15:0 and C16:0 (P > 0.05). By the linkage disequilibrium (LD) analyses, we found two, one, one, and one haplotype blocks in PRLR, MOGAT1, MINPP1, and CHUK, respectively, and each haplotype block was significantly associated with at least one milk FA trait (P = 0.0456 ~ < 0.0001). Further, g.38949011G > A in PRLR, and g.111599360A > G and g.111601747 T > A in MOGAT1 were predicted to alter the transcription factor binding sites (TFBSs). A missense mutation, g.39115344G > A, could change the PRLR protein structure. The g.20966385C > G of CHUK varied the binding sequences for microRNAs. Therefore, we deduced the five SNPs as the potential functional mutations. CONCLUSION In summary, we first detected the genetic effects of PRLR, MOGAT1, MINPP1 and CHUK genes on milk FA traits, and researched the potential functional mutations. These data provided the basis for further investigation on function validation of the four genes in Chinese Holstein.
Collapse
Affiliation(s)
- Lijun Shi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 China
| | - Lin Liu
- Beijing Dairy Cattle Center, Beijing, 100192 China
| | - Xiaoqing Lv
- Beijing Dairy Cattle Center, Beijing, 100192 China
| | - Zhu Ma
- Beijing Dairy Cattle Center, Beijing, 100192 China
| | - Yuze Yang
- Beijing General Station of Animal Husbandry, Beijing, 100101 China
| | - Yanhua Li
- Beijing Dairy Cattle Center, Beijing, 100192 China
| | - Feng Zhao
- Beijing Dairy Cattle Center, Beijing, 100192 China
| | - Dongxiao Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 China
| | - Bo Han
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193 China
| |
Collapse
|
28
|
Liu H, Zhang H, Wang X, Yu X, Hu C, Zhang X. Alterations of DNA methylation profile in proximal jejunum potentially contribute to the beneficial effects of gastric bypass in a diabetic rat model. Surg Obes Relat Dis 2019; 15:1291-1298. [DOI: 10.1016/j.soard.2019.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/08/2019] [Accepted: 05/19/2019] [Indexed: 10/26/2022]
|
29
|
Lutkewitte AJ, McCommis KS, Schweitzer GG, Chambers KT, Graham MJ, Wang L, Patti GJ, Hall AM, Finck BN. Hepatic monoacylglycerol acyltransferase 1 is induced by prolonged food deprivation to modulate the hepatic fasting response. J Lipid Res 2019; 60:528-538. [PMID: 30610082 PMCID: PMC6399500 DOI: 10.1194/jlr.m089722] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/05/2018] [Indexed: 01/14/2023] Open
Abstract
During prolonged fasting, the liver plays a central role in maintaining systemic energy homeostasis by producing glucose and ketones in processes fueled by oxidation of fatty acids liberated from adipose tissue. In mice, this is accompanied by transient hepatic accumulation of glycerolipids. We found that the hepatic expression of monoacylglycerol acyltransferase 1 (Mogat1), an enzyme with monoacylglycerol acyltransferase (MGAT) activity that produces diacyl-glycerol from monoacylglycerol, was significantly increased in the liver of fasted mice compared with mice given ad libitum access to food. Basal and fasting-induced expression of Mogat1 was markedly diminished in the liver of mice lacking the transcription factor PPARα. Suppressing Mogat1 expression in liver and adipose tissue with antisense oligonucleotides (ASOs) reduced hepatic MGAT activity and triglyceride content compared with fasted controls. Surprisingly, the expression of many other PPARα target genes and PPARα activity was also decreased in mice given Mogat1 ASOs. When mice treated with control or Mogat1 ASOs were gavaged with the PPARα ligand, WY-14643, and then fasted for 18 h, WY-14643 administration reversed the effects of Mogat1 ASOs on PPARα target gene expression and liver triglyceride content. In conclusion, Mogat1 is a fasting-induced PPARα target gene that may feed forward to regulate liver PPARα activity during food deprivation.
Collapse
Affiliation(s)
- Andrew J Lutkewitte
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| | - Kyle S McCommis
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| | - George G Schweitzer
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| | - Kari T Chambers
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| | | | - Lingjue Wang
- Department of Chemistry, Washington University, St. Louis, MO
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Angela M Hall
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| | - Brian N Finck
- Center for Human Nutrition Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
30
|
Mizuno N, Seko Y, Kataoka S, Okuda K, Furuta M, Takemura M, Taketani H, Hara T, Umemura A, Nishikawa T, Yamaguchi K, Moriguchi M, Itoh Y. Increase in the skeletal muscle mass to body fat mass ratio predicts the decline in transaminase in patients with nonalcoholic fatty liver disease. J Gastroenterol 2019; 54:160-170. [PMID: 29948305 DOI: 10.1007/s00535-018-1485-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/11/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND The aim of this retrospective study was to determine the effect of skeletal muscle and body fat on liver function in patients with nonalcoholic fatty liver disease (NAFLD) diagnosed by liver biopsy. METHODS Among the 219 patients with NAFLD enrolled in this study was a cohort of 139 patients who had their body composition measured with Inbody720 at baseline and at ≥ 1 year postbaseline, to elucidate the relationship between liver function and changes in skeletal muscle and body fat mass. Multivariate analysis was used to identify factors influencing low skeletal muscle mass index (SMI, defined as 7 kg/m2 in men, and 5.7 kg/m2 in women) and the skeletal muscle mass to body fat mass ratio (SF ratio). RESULTS Of the 219 patients enrolled, 27 (12.3%) had a low SMI. Patient age (> 70 years) and female gender were identified as risk factors for low SMI. Hepatic fibrosis was not associated with SMI. In the cohort followed up at baseline and 12 months later, transaminase activity, body fat mass, and SMI significantly decreased over time. Changes in the SF ratio were significantly associated with changes in liver function. An increase in the SF ratio [hazard ratio (HR) 10.99 in men, 6.849 in women] was a predictor of reduced ALT, independent of age and other backgrounds. CONCLUSIONS In the patients with NAFLD, SMI was decreased, even in the early stages of NAFLD. Therapeutic strategies for NAFLD require a reduction in body fat mass and the maintenance of skeletal muscle is also needed.
Collapse
Affiliation(s)
- Naoki Mizuno
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuya Seko
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Seita Kataoka
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Keiichiroh Okuda
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mitsuhiro Furuta
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Masashi Takemura
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroyoshi Taketani
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tasuku Hara
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Umemura
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Taichiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kanji Yamaguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Michihisa Moriguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
31
|
Mu W, Cheng XF, Liu Y, Lv QZ, Liu GL, Zhang JG, Li XY. Potential Nexus of Non-alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus: Insulin Resistance Between Hepatic and Peripheral Tissues. Front Pharmacol 2019; 9:1566. [PMID: 30692925 PMCID: PMC6339917 DOI: 10.3389/fphar.2018.01566] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022] Open
Abstract
The liver is the central metabolic organ and plays a pivotal role in regulating homeostasis of glucose and lipid metabolism. Aberrant liver metabolism promotes insulin resistance, which is reported to be a common characteristic of metabolic diseases such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). There is a complex and bidirectional relationship between NAFLD and T2DM. NAFLD patients with hepatic insulin resistance generally share a high risk of impaired fasting glucose associated with early diabetes; most patients with T2DM experience non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), and other more severe liver complications such as cirrhosis and hepatocellular carcinoma (HCC). Additionally, hepatic insulin resistance, which is caused by diacylglycerol-mediated activation of protein kinase C epsilon (PKC𝜀), may be the critical pathological link between NAFLD and T2DM. Therefore, this review aims to illuminate current insights regarding the complex and strong association between NAFLD and T2DM and summarize novel and emerging targets for the treatment of hepatic insulin resistance based on established mechanistic knowledge.
Collapse
Affiliation(s)
- Wan Mu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Fang Cheng
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian-Zhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Influence of Liver Condition and Copper on Selective Parameters of Post-Mortem Dog Tissue Samples. Animals (Basel) 2018; 8:ani8120237. [PMID: 30551591 PMCID: PMC6316235 DOI: 10.3390/ani8120237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 11/17/2022] Open
Abstract
One of the liver functions is copper storage, which can be toxic when in excess. The objective of this retrospective study was to determine the relationship between hepatic copper and pathology conditions in stored samples from 55 post-mortem dogs (37 Beagles, 12 Labrador Retrievers, and 6 Labrador Mixes). The analyses evaluated data from blood chemistry and complete blood count (CBC) that were measured immediately before euthanasia, and liver biopsies which were harvested at necropsy and frozen at -80 °C. Slides for microscopic evaluation were prepared, and liver copper and plasma metabolites were measured. Hepatic copper was correlated (p ≤ 0.001) with monoacylglycerols, 13-HODE + 9-HODE (13-hydroxy-9,11-octadecadienoic acid + 9-hydroxy-10,12-octadecadienoic acid), and stearoyl-arachidonoyl-glycerophosphocholine. This indicates lipid metabolism modification and cell membrane oxidation. However, hepatic copper was not related to liver histopathology severity or altered liver biomarkers. The severity of liver pathology was positively correlated (p ≤ 0.05) with liver enzymes, bile salts, and glycerophosphocholines, suggesting cholestasis and altered lipid and amino acid metabolism. Liver neoplasia had increased (p ≤ 0.05) metabolites derived from nucleotides, along with an increase (p ≤ 0.05) in α-ketoglutarate from the energy and amino acid metabolism (p ≤ 0.05), suggesting rapid cell division. This study offers further insight regarding changes in metabolism due to hepatic tissue damage.
Collapse
|
33
|
Devasthale P, Cheng D. Monoacylglycerol Acyltransferase 2 (MGAT2) Inhibitors for the Treatment of Metabolic Diseases and Nonalcoholic Steatohepatitis (NASH). J Med Chem 2018; 61:9879-9888. [PMID: 29986142 DOI: 10.1021/acs.jmedchem.8b00864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Monoacylglycerol transferase 2 (MGAT2) is a pivotal enzyme in the monoacylglycerol pathway for triacylglycerol synthesis. The pathway for triacylglycerol synthesis has provided several attractive targets for drug discovery in the treatment of metabolic diseases. Marketed drugs that inhibit enzymes in this pathway include orlistat (pancreatic lipase inhibitor), lomitapide (mitochondrial transfer protein inhibitor), and mipomersen (apolipoprotein B synthesis inhibitor), but poor gastrointestinal (GI) tolerability or safety considerations have limited their use and indications. In addition, several inhibitors of diacylglycerol transferase 1 (DGAT1) have advanced to the clinic but were withdrawn due to poor GI tolerability. This report first discusses the biological rationale in support of inhibition of MGAT2 as a therapeutic approach that may offer a distinct and superior efficacy versus GI tolerability profile and then reviews advances in the discovery of small molecule MGAT2 inhibitors for the treatment of metabolic diseases and nonalcoholic steatohepatitis (NASH).
Collapse
|
34
|
Liss KHH, Lutkewitte AJ, Pietka T, Finck BN, Franczyk M, Yoshino J, Klein S, Hall AM. Metabolic importance of adipose tissue monoacylglycerol acyltransferase 1 in mice and humans. J Lipid Res 2018; 59:1630-1639. [PMID: 29853530 PMCID: PMC6121930 DOI: 10.1194/jlr.m084947] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/10/2018] [Indexed: 12/31/2022] Open
Abstract
Adipocyte triglyceride storage provides a reservoir of energy that allows the organism to survive times of nutrient scarcity, but excessive adiposity has emerged as a health problem in many areas of the world. Monoacylglycerol acyltransferase (MGAT) acylates monoacylglycerol to produce diacylglycerol; the penultimate step in triglyceride synthesis. However, little is known about MGAT activity in adipocytes, which are believed to rely primarily on another pathway for triglyceride synthesis. We show that expression of the gene that encodes MGAT1 is robustly induced during adipocyte differentiation and that its expression is suppressed in fat of genetically-obese mice and metabolically-abnormal obese human subjects. Interestingly, MGAT1 expression is also reduced in physiologic contexts where lipolysis is high. Moreover, knockdown or knockout of MGAT1 in adipocytes leads to higher rates of basal adipocyte lipolysis. Collectively, these data suggest that MGAT1 activity may play a role in regulating basal adipocyte FFA retention.
Collapse
Affiliation(s)
- Kim H H Liss
- Department of Pediatrics Washington University School of Medicine, St. Louis, MO 63110
| | | | - Terri Pietka
- Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian N Finck
- Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael Franczyk
- Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jun Yoshino
- Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Samuel Klein
- Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Angela M Hall
- Medicine, Washington University School of Medicine, St. Louis, MO 63110.
| |
Collapse
|
35
|
Ramon-Krauel M, Pentinat T, Bloks VW, Cebrià J, Ribo S, Pérez-Wienese R, Vilà M, Palacios-Marin I, Fernández-Pérez A, Vallejo M, Téllez N, Rodríguez MÀ, Yanes O, Lerin C, Díaz R, Plosch T, Tietge UJF, Jimenez-Chillaron JC. Epigenetic programming at the Mogat1 locus may link neonatal overnutrition with long-term hepatic steatosis and insulin resistance. FASEB J 2018; 32:fj201700717RR. [PMID: 29812971 DOI: 10.1096/fj.201700717rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Postnatal overfeeding increases the risk of chronic diseases later in life, including obesity, insulin resistance, hepatic steatosis, and type 2 diabetes. Epigenetic mechanisms might underlie the long-lasting effects associated with early nutrition. Here we aimed to explore the molecular pathways involved in early development of insulin resistance and hepatic steatosis, and we examined the potential contribution of DNA methylation and histone modifications to long-term programming of metabolic disease. We used a well-characterized mouse model of neonatal overfeeding and early adiposity by litter size reduction. Neonatal overfeeding led to hepatic insulin resistance very early in life that persisted throughout adulthood despite normalizing food intake. Up-regulation of monoacylglycerol O-acyltransferase ( Mogat) 1 conceivably mediates hepatic steatosis and insulin resistance through increasing intracellular diacylglycerol content. Early and sustained deregulation of Mogat1 was associated with a combination of histone modifications that might favor Mogat1 expression. In sum, postnatal overfeeding causes extremely rapid derangements of hepatic insulin sensitivity that remain relatively stable until adulthood. Epigenetic mechanisms, particularly histone modifications, could contribute to such long-lasting effects. Our data suggest that targeting hepatic monoacylglycerol acyltransferase activity during early life might provide a novel strategy to improve hepatic insulin sensitivity and prevent late-onset insulin resistance and fatty liver disease.-Ramon-Krauel, M., Pentinat, T., Bloks, V. W., Cebrià, J., Ribo, S., Pérez-Wienese, R., Vilà, M., Palacios-Marin, I., Fernández-Pérez, A., Vallejo, M., Téllez, N., Rodríguez, M. À., Yanes, O., Lerin, C., Díaz, R., Plosch, T., Tietge, U. J. F., Jimenez-Chillaron, J. C. Epigenetic programming at the Mogat1 locus may link neonatal overnutrition with long-term hepatic steatosis and insulin resistance.
Collapse
Affiliation(s)
- Marta Ramon-Krauel
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Thais Pentinat
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Vincent W Bloks
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Judith Cebrià
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Silvia Ribo
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Ricky Pérez-Wienese
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Maria Vilà
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Ivonne Palacios-Marin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Antonio Fernández-Pérez
- Ciber de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid (CSIC/UAM), Madrid, Spain
| | - Mario Vallejo
- Ciber de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid (CSIC/UAM), Madrid, Spain
| | - Noèlia Téllez
- Ciber de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Bellvitge Biomedical Research Institute (IDIBELL) L'Hospitalet, Barcelona, Spain
| | - Miguel Àngel Rodríguez
- Ciber de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, Spain
| | - Oscar Yanes
- Ciber de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, Spain
| | - Carles Lerin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Rubén Díaz
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Torsten Plosch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | |
Collapse
|
36
|
Lee YJ, Kim JW. Monoacylglycerol O-acyltransferase 1 (MGAT1) localizes to the ER and lipid droplets promoting triacylglycerol synthesis. BMB Rep 2018; 50:367-372. [PMID: 28347400 PMCID: PMC5584744 DOI: 10.5483/bmbrep.2017.50.7.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Indexed: 01/14/2023] Open
Abstract
Monoacylglycerol acyltransferase 1 (MGAT) is a microsomal enzyme that catalyzes the synthesis of diacylglycerol (DAG) and triacylglycerol (TAG). However, the subcellular localization and catalytic function domain of this enzyme is poorly understood. In this report, we identified that murine MGAT1 localizes to the endoplasmic reticulum (ER) under normal conditions, whereas MGAT1 co-localize to the lipid droplets (LD) under conditions of enriching fatty acids, contributing to TAG synthesis and LD expansion. For the enzyme activity, both the N-terminal transmembrane domain and catalytic HPHG motif are required. We also show that the transmembrane domain of MGAT1 consists of two hydrophobic regions in the N-terminus, and the consensus sequence FLXLXXXn, a putative neutral lipid-binding domain, exists in the first transmembrane domain. Finally, MGAT1 interacts with DGAT2, which serves to synergistically increase the TAG biosynthesis and LD expansion, leading to enhancement of lipid accumulation in the liver and fat.
Collapse
Affiliation(s)
- Yoo Jeong Lee
- Division of Metabolic Disease, Center for Biomedical Sciences, National Institutes of Health, Cheongju 28159, Korea
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Integrated Genomic Research Center for Metabolic Regulation, Institute of Genetic Science, Yonsei University College of Medicine; Brain Korea 21 PLUS Project for Medical Science, Yonsei University; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
37
|
Myers CE, Hoelzinger DB, Truong TN, Chew LA, Myles A, Chaudhuri L, Egan JB, Liu J, Gendler SJ, Cohen PA. Chemotherapy can induce weight normalization of morbidly obese mice despite undiminished ingestion of high fat diet. Oncotarget 2018; 8:5426-5438. [PMID: 28076839 PMCID: PMC5354920 DOI: 10.18632/oncotarget.14576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/05/2016] [Indexed: 02/06/2023] Open
Abstract
Morbidly obese patients who accomplish substantial weight loss often display a long-term decline in their resting metabolism, causing even relatively restrained caloric intake to trigger a relapse to the obese state. Paradoxically, we observed that morbidly obese mice receiving chemotherapy for cancer experienced spontaneous weight reduction despite unabated ingestion of their high fat diet (HFD). This response to chemotherapy could also be achieved in morbidly obese mice without cancer. Optimally dosed methotrexate (MTX) or cyclophosphamide (CY) enabled the mice to completely and safely normalize their body weight despite continued consumption of obesogenic quantities of HFD. Weight reduction was not attributable to decreased HFD intake, enhanced energy expenditure or malabsorption. MTX or CY dosing significantly depleted both adipose tissue and preadipocyte progenitors. Remarkably, however, despite continued high fat feeding, a compensatory increase in hepatocyte lipid storage was not observed, but rather the opposite. Gene microarray liver analyses demonstrated that HFD mice receiving MTX or CY experienced significantly inhibited lipogenesis and lipid storage, whereas Enho (energy homeostasis) gene expression was significantly upregulated. Further metabolic studies employing a human hepatocellular line revealed that MTX treatment preserved robust oxidative phosphorylation, but also promoted mitochondrial uncoupling with a surge in proton leak. This is the first report that certain optimally dosed chemotherapeutic agents can induce weight loss in morbidly obese mice without reduced dietary intake, apparently by depleting stores of adipocytes and their progenitors, curtailment of lipogenesis, and inconspicuous disposal of incoming dietary lipid via a steady state partial uncoupling of mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Cheryl E Myers
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Tiffany N Truong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Lindsey A Chew
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Arpita Myles
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Sandra J Gendler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Peter A Cohen
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
38
|
Kawanishi N, Takagi K, Lee HC, Nakano D, Okuno T, Yokomizo T, Machida S. Endurance exercise training and high-fat diet differentially affect composition of diacylglycerol molecular species in rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2018; 314:R892-R901. [PMID: 29443549 PMCID: PMC6032301 DOI: 10.1152/ajpregu.00371.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin resistance of peripheral muscle is implicated in the etiology of metabolic syndrome in obesity. Although accumulation of glycerolipids, such as triacylglycerol and diacylglycerol (DAG), in muscle contributes to insulin resistance in obese individuals, endurance-trained athletes also have higher glycerolipid levels but normal insulin sensitivity. We hypothesized that the difference in insulin sensitivity of skeletal muscle between athletes and obese individuals stems from changes in fatty acid composition of accumulated lipids. Here, we evaluated the effects of intense endurance exercise and high-fat diet (HFD) on the accumulation and composition of lipid molecular species in rat skeletal muscle using a lipidomic approach. Sprague-Dawley female rats were randomly assigned to three groups and received either normal diet (ND) in sedentary conditions, ND plus endurance exercise training, or HFD in sedentary conditions. Rats were fed ND or HFD between 4 and 12 wk of age. Rats in the exercise group ran on a treadmill for 120 min/day, 5 days/wk, for 8 wk. Soleus muscle lipidomic profiles were obtained using liquid chromatography/tandem mass spectrometry. Total DAG levels, particularly those of palmitoleate-containing species, were increased in muscle by exercise training. However, whereas the total DAG level in the muscle was also increased by HFD, the levels of DAG molecular species containing palmitoleate were decreased by HFD. The concentration of phosphatidylethanolamine molecular species containing palmitoleate was increased by exercise but decreased by HFD. Our results indicate that although DAG accumulation was similar levels in trained and sedentary obese rats, specific changes in molecular species containing palmitoleate were opposite.
Collapse
Affiliation(s)
- Noriaki Kawanishi
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan.,Research Fellow of the Japan Society for the Promotion of Sciences , Tokyo , Japan.,Institute of Health and Sports Science and Medicine, Juntendo University, Inzai, Japan.,Faculty of Advanced Engineering, Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Kana Takagi
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan
| | - Hyeon-Cheol Lee
- Department of Biochemistry, Graduate School of Medicine, Juntendo University , Tokyo , Japan
| | - Daiki Nakano
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Graduate School of Medicine, Juntendo University , Tokyo , Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Graduate School of Medicine, Juntendo University , Tokyo , Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan
| |
Collapse
|
39
|
Sanchez-Gurmaches J, Tang Y, Jespersen NZ, Wallace M, Martinez Calejman C, Gujja S, Li H, Edwards YJK, Wolfrum C, Metallo CM, Nielsen S, Scheele C, Guertin DA. Brown Fat AKT2 Is a Cold-Induced Kinase that Stimulates ChREBP-Mediated De Novo Lipogenesis to Optimize Fuel Storage and Thermogenesis. Cell Metab 2018; 27:195-209.e6. [PMID: 29153407 PMCID: PMC5762420 DOI: 10.1016/j.cmet.2017.10.008] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/30/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
Brown adipose tissue (BAT) is a therapeutic target for metabolic diseases; thus, understanding its metabolic circuitry is clinically important. Many studies of BAT compare rodents mildly cold to those severely cold. Here, we compared BAT remodeling between thermoneutral and mild-cold-adapted mice, conditions more relevant to humans. Although BAT is renowned for catabolic β-oxidative capacity, we find paradoxically that the anabolic de novo lipogenesis (DNL) genes encoding ACLY, ACSS2, ACC, and FASN were among the most upregulated by mild cold and that, in humans, DNL correlates with Ucp1 expression. The regulation and function of adipocyte DNL and its association with thermogenesis are not understood. We provide evidence suggesting that AKT2 drives DNL in adipocytes by stimulating ChREBPβ transcriptional activity and that cold induces the AKT2-ChREBP pathway in BAT to optimize fuel storage and thermogenesis. These data provide insight into adipocyte DNL regulation and function and illustrate the metabolic flexibility of thermogenesis.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yuefeng Tang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Naja Zenius Jespersen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Martina Wallace
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Camila Martinez Calejman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sharvari Gujja
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huawei Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yvonne J K Edwards
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH-Zürich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Søren Nielsen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Scheele
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
40
|
Samuel VT, Shulman GI. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metab 2018; 27:22-41. [PMID: 28867301 PMCID: PMC5762395 DOI: 10.1016/j.cmet.2017.08.002] [Citation(s) in RCA: 509] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/01/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022]
Abstract
NAFLD is closely linked with hepatic insulin resistance. Accumulation of hepatic diacylglycerol activates PKC-ε, impairing insulin receptor activation and insulin-stimulated glycogen synthesis. Peripheral insulin resistance indirectly influences hepatic glucose and lipid metabolism by increasing flux of substrates that promote lipogenesis (glucose and fatty acids) and gluconeogenesis (glycerol and fatty acid-derived acetyl-CoA, an allosteric activator of pyruvate carboxylase). Weight loss with diet or bariatric surgery effectively treats NAFLD, but drugs specifically approved for NAFLD are not available. Some new pharmacological strategies act broadly to alter energy balance or influence pathways that contribute to NAFLD (e.g., agonists for PPAR γ, PPAR α/δ, FXR and analogs for FGF-21, and GLP-1). Others specifically inhibit key enzymes involved in lipid synthesis (e.g., mitochondrial pyruvate carrier, acetyl-CoA carboxylase, stearoyl-CoA desaturase, and monoacyl- and diacyl-glycerol transferases). Finally, a novel class of liver-targeted mitochondrial uncoupling agents increases hepatocellular energy expenditure, reversing the metabolic and hepatic complications of NAFLD.
Collapse
Affiliation(s)
- Varman T Samuel
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Veterans Affairs Medical Center, West Haven, CT 06516, USA.
| | - Gerald I Shulman
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
41
|
Poursharifi P, Madiraju SRM, Prentki M. Monoacylglycerol signalling and ABHD6 in health and disease. Diabetes Obes Metab 2017; 19 Suppl 1:76-89. [PMID: 28880480 DOI: 10.1111/dom.13008] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/24/2017] [Accepted: 05/11/2017] [Indexed: 12/14/2022]
Abstract
Lipid metabolism dysregulation underlies chronic pathologies such as obesity, diabetes and cancer. Besides their role in structure and energy storage, lipids are also important signalling molecules regulating multiple biological functions. Thus, understanding the precise lipid metabolism enzymatic steps that are altered in some pathological conditions is helpful for designing better treatment strategies. Several monoacylglycerol (MAG) species are only recently being recognized as signalling lipid molecules in different tissues. Recent studies indicated the importance of the ubiquitously expressed serine hydrolase α/β-hydrolase domain 6 (ABHD6), which is a MAG hydrolase, in regulating signalling competent MAG in both central and peripheral tissues. The central and peripheral function of the endocannabinoid 2-arachidonoylglycerol, which is a 2-MAG, and its breakdown by both ABHD6 and classical MAG lipase has been well documented. ABHD6 and its substrate MAG appear to be involved in the regulation of various physiological and pathological processes including insulin secretion, adipose browning, food intake, neurotransmission, autoimmune disorders, neurological and metabolic diseases as well as cancer. Diverse cellular targets such as mammalian unc13-1 (Munc13-1), PPARs, GPR119 and CB1/2 receptors, for MAG-mediated signalling processes have been proposed in different cell types. The purpose of this review is to provide a comprehensive summary of the current state of knowledge regarding ABHD6/MAG signalling and its possible therapeutic implications.
Collapse
Affiliation(s)
- Pegah Poursharifi
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Sri Ramachandra Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| |
Collapse
|
42
|
Elam MB, Majumdar G, Mozhui K, Gerling IC, Vera SR, Fish-Trotter H, Williams RW, Childress RD, Raghow R. Patients experiencing statin-induced myalgia exhibit a unique program of skeletal muscle gene expression following statin re-challenge. PLoS One 2017; 12:e0181308. [PMID: 28771594 PMCID: PMC5542661 DOI: 10.1371/journal.pone.0181308] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/29/2017] [Indexed: 01/21/2023] Open
Abstract
Statins, the 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase inhibitors, are widely prescribed for treatment of hypercholesterolemia. Although statins are generally well tolerated, up to ten percent of statin-treated patients experience myalgia symptoms, defined as muscle pain without elevated creatinine phosphokinase (CPK) levels. Myalgia is the most frequent reason for discontinuation of statin therapy. The mechanisms underlying statin myalgia are not clearly understood. To elucidate changes in gene expression associated with statin myalgia, we compared profiles of gene expression in skeletal muscle biopsies from patients with statin myalgia who were undergoing statin re-challenge (cases) versus those of statin-tolerant controls. A robust separation of case and control cohorts was revealed by Principal Component Analysis of differentially expressed genes (DEGs). To identify putative gene expression and metabolic pathways that may be perturbed in skeletal muscles of patients with statin myalgia, we subjected DEGs to Ingenuity Pathways (IPA) and DAVID (Database for Annotation, Visualization and Integrated Discovery) analyses. The most prominent pathways altered by statins included cellular stress, apoptosis, cell senescence and DNA repair (TP53, BARD1, Mre11 and RAD51); activation of pro-inflammatory immune response (CXCL12, CST5, POU2F1); protein catabolism, cholesterol biosynthesis, protein prenylation and RAS-GTPase activation (FDFT1, LSS, TP53, UBD, ATF2, H-ras). Based on these data we tentatively conclude that persistent myalgia in response to statins may emanate from cellular stress underpinned by mechanisms of post-inflammatory repair and regeneration. We also posit that this subset of individuals is genetically predisposed to eliciting altered statin metabolism and/or increased end-organ susceptibility that lead to a range of statin-induced myopathies. This mechanistic scenario is further bolstered by the discovery that a number of single nucleotide polymorphisms (e.g., SLCO1B1, SLCO2B1 and RYR2) associated with statin myalgia and myositis were observed with increased frequency among patients with statin myalgia.
Collapse
Affiliation(s)
- Marshall B. Elam
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- * E-mail: (MBE); (RR)
| | - Gipsy Majumdar
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Khyobeni Mozhui
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Ivan C. Gerling
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Santiago R. Vera
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Hannah Fish-Trotter
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Richard D. Childress
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Rajendra Raghow
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- * E-mail: (MBE); (RR)
| |
Collapse
|
43
|
Icli B, Feinberg MW. MicroRNAs in dysfunctional adipose tissue: cardiovascular implications. Cardiovasc Res 2017; 113:1024-1034. [PMID: 28505257 PMCID: PMC5852642 DOI: 10.1093/cvr/cvx098] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/20/2017] [Accepted: 05/12/2017] [Indexed: 12/16/2022] Open
Abstract
In this review, we focus on the emerging role of microRNAs, non-coding RNAs that regulate gene expression and signaling pathways, in dysfunctional adipose tissue. We highlight current paradigms of microRNAs involved in adipose differentiation and function in depots such as white, brown, and beige adipose tissues and potential implications of microRNA dysregulation in human disease such as obesity, inflammation, microvasculature dysfunction, and related cardiovascular diseases. We highlight accumulating studies indicating that adipocyte-derived microRNAs may not only serve as biomarkers of cardiometabolic disease, but also may directly regulate gene expression of other tissues. Finally, we discuss the future prospects, challenges, and emerging strategies for microRNA delivery and targeting for therapeutic applications in cardiovascular disease states associated with adipocyte dysfunction.
Collapse
Affiliation(s)
- Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB-742F, Boston, MA 02115, USA
| | - Mark W. Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB-742F, Boston, MA 02115, USA
| |
Collapse
|
44
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017. [DOI: 10.1210/er.2016-1122.2017.1.test] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017; 38:145-168. [PMID: 28323921 PMCID: PMC5460677 DOI: 10.1210/er.2016-1122] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/13/2017] [Indexed: 12/11/2022]
Abstract
Chronic, low-grade systemic inflammation and impaired microvascular function are critical hallmarks in the development of insulin resistance. Accordingly, insulin resistance is a major risk factor for type 2 diabetes and cardiovascular disease. Accumulating studies demonstrate that restoration of impaired function of the diabetic macro- and microvasculature may ameliorate a range of cardiovascular disease states and diabetes-associated complications. In this review, we focus on the emerging role of microRNAs (miRNAs), noncoding RNAs that fine-tune target gene expression and signaling pathways, in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury. We highlight current pathophysiological paradigms of miRNAs and their targets involved in regulating the diabetic microvasculature in a range of diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type I or type II diabetes. Finally, we discuss emerging delivery platforms for manipulating miRNA expression or function as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Pharmacology and Pharmacy, University of Hong Kong, Pokfulam, Hong Kong SAR, China, and
| | - Xinghui Sun
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| | - Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mark W. Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
46
|
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
|
47
|
Kajimoto K, Suemitsu E, Sato Y, Sakurai Y, Harashima H. Liver-Specific Silencing of Lipin1 Reduces Fat Mass as Well as Hepatic Triglyceride Biosynthesis in Mice. Biol Pharm Bull 2017; 39:1653-1661. [PMID: 27725442 DOI: 10.1248/bpb.b16-00353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lipin1, a bifunctional protein, regulates fatty acid utilization in the triglyceride biosynthesis pathway. In the current study, using a liver-specific in vivo short interfering RNA (siRNA) delivery system, we examined the pathological and physiological roles of hepatic Lipin1 in the development of insulin resistance and the maintenance of systemic energy homeostasis. Liver-specific silencing of Lipin1 expression was achieved by the systemic administration of siRNA against Lpin1 mRNA (siLpin1)-loaded lipid nanoparticles (LNPs) to wild type mice at 3-4 d intervals for 25 d. The siLpin1-treated mice showed normal blood glucose levels and insulin sensitivity, however, triglyceride (TG) levels were reduced in liver and peripheral blood of them. The knockdown of hepatic Lipin1 in mice led to marked decrease in adipose tissue mass and adipocyte diameters in epididymal and inguinal fat depots without the undesired silencing of Lipin1 in adipose tissue. In summary, we report for the first time that the down-regulation of hepatic Lipin1 expression leads to less adiposity as well as a decrease in TG level in the liver and blood circulation, without any alterations in the glucose tolerance and blood glucose levels. Our findings may provide new insights into the physiological roles of hepatic Lipin1 in systemic energy homeostasis.
Collapse
|
48
|
Erion DM, Park HJ, Lee HY. The role of lipids in the pathogenesis and treatment of type 2 diabetes and associated co-morbidities. BMB Rep 2017; 49:139-48. [PMID: 26728273 PMCID: PMC4915228 DOI: 10.5483/bmbrep.2016.49.3.268] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Indexed: 12/25/2022] Open
Abstract
In the past decade, the incidence of type 2 diabetes (T2D) has rapidly increased, along with the associated cardiovascular complications. Therefore, understanding the pathophysiology underlying T2D, the associated complications and the impact of therapeutics on the T2D development has critical importance for current and future therapeutics. The prevailing feature of T2D is hyperglycemia due to excessive hepatic glucose production, insulin resistance, and insufficient secretion of insulin by the pancreas. These contribute to increased fatty acid influx into the liver and muscle causing accumulation of lipid metabolites. These lipid metabolites cause dyslipidemia and non-alcoholic fatty liver disease, which ultimately contributes to the increased cardiovascular risk in T2D. Therefore, understanding the mechanisms of hepatic insulin resistance and the specific role of liver lipids is critical in selecting and designing the most effective therapeutics for T2D and the associated co-morbidities, including dyslipidemia and cardiovascular disease. Herein, we review the effects and molecular mechanisms of conventional anti-hyperglycemic and lipid-lowering drugs on glucose and lipid metabolism. [BMB Reports 2016; 49(3): 139-148].
Collapse
Affiliation(s)
- Derek M Erion
- Takeda Pharmaceuticals 350 Massachusetts Ave. Cambridge, MA, 02139, USA
| | - Hyun-Jun Park
- Department of Molecular Medicine, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Korea
| | - Hui-Young Lee
- Department of Molecular Medicine and Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
49
|
Wolf Greenstein A, Majumdar N, Yang P, Subbaiah PV, Kineman RD, Cordoba-Chacon J. Hepatocyte-specific, PPARγ-regulated mechanisms to promote steatosis in adult mice. J Endocrinol 2017; 232:107-121. [PMID: 27799461 PMCID: PMC5120553 DOI: 10.1530/joe-16-0447] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/25/2016] [Indexed: 12/15/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is the target for thiazolidinones (TZDs), drugs that improve insulin sensitivity and fatty liver in humans and rodent models, related to a reduction in hepatic de novo lipogenesis (DNL). The systemic effects of TZDs are in contrast to reports suggesting hepatocyte-specific activation of PPARγ promotes DNL, triacylglycerol (TAG) uptake and fatty acid (FA) esterification. As these hepatocyte-specific effects of PPARγ could counterbalance the positive therapeutic actions of systemic delivery of TZDs, the current study used a mouse model of adult-onset, liver (hepatocyte)-specific PPARγ knockdown (aLivPPARγkd). This model has advantages over existing congenital knockout models, by avoiding compensatory changes related to embryonic knockdown, thus better modeling the impact of altering PPARγ on adult physiology, where metabolic diseases most frequently develop. The impact of aLivPPARγkd on hepatic gene expression and endpoints in lipid metabolism was examined after 1 or 18 weeks (Chow-fed) or after 14 weeks of low- or high-fat (HF) diet. aLivPPARγkd reduced hepatic TAG content but did not impact endpoints in DNL or TAG uptake. However, aLivPPARγkd reduced the expression of the FA translocase (Cd36), in 18-week Chow- and HF-fed mice, associated with increased NEFA after HF feeding. Also, aLivPPARγkd dramatically reduced Mogat1 expression, that was reflected by an increase in hepatic monoacylglycerol (MAG) levels, indicative of reduced MOGAT activity. These results, coupled with previous reports, suggest that Cd36-mediated FA uptake and MAG pathway-mediated FA esterification are major targets of hepatocyte PPARγ, where loss of this control explains in part the protection against steatosis observed after aLivPPARγkd.
Collapse
Affiliation(s)
- Abigail Wolf Greenstein
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Biologic Resources LaboratoryUniversity of Illinois at Chicago, Chicago, Illinois, USA
| | - Neena Majumdar
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Peng Yang
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Papasani V Subbaiah
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rhonda D Kineman
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jose Cordoba-Chacon
- Research and Development DivisionJesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
- Section of EndocrinologyDiabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
50
|
Sankella S, Garg A, Agarwal AK. Characterization of the Mouse and Human Monoacylglycerol O-Acyltransferase 1 (Mogat1) Promoter in Human Kidney Proximal Tubule and Rat Liver Cells. PLoS One 2016; 11:e0162504. [PMID: 27611931 PMCID: PMC5017789 DOI: 10.1371/journal.pone.0162504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/23/2016] [Indexed: 12/22/2022] Open
Abstract
Monoacylglycerol acyltransferase 1 (Mogat1) catalyzes the conversion of monoacylglycerols (MAG) to diacylglycerols (DAG), the precursor of several physiologically important lipids such as phosphatidylcholine, phosphatidylethanolamine and triacylglycerol (TAG). Expression of Mogat1 is tissue restricted and it is highly expressed in the kidney, stomach and adipose tissue but minimally in the normal adult liver. To understand the transcriptional regulation of Mogat1, we characterized the mouse and human Mogat1 promoters in human kidney proximal tubule-2 (HK-2) cells. In-silico analysis revealed several peroxisome proliferator response element (PPRE) binding sites in the promoters of both human and mouse Mogat1. These sites responded to all three peroxisome proliferator activated receptor (PPAR) isoforms such that their respective agonist or antagonist activated or inhibited the expression of Mogat1. PPRE site mutagenesis revealed that sites located at -592 and -2518 are very effective in decreasing luciferase reporter gene activity. Chromatin immunoprecipitation (ChIP) assay using PPARα antibody further confirmed the occupancy of these sites by PPARα. While these assays revealed the core promoter elements necessary for Mogat1 expression, there are additional elements required to regulate its tissue specific expression. Chromosome conformation capture (3C) assay revealed additional cis-elements located ~10–15 kb upstream which interact with the core promoter. These chromosomal regions are responsive to both PPARα agonist and antagonist.
Collapse
Affiliation(s)
- Shireesha Sankella
- Division of Nutrition and Metabolic Diseases, Department of Internal Medicine and Center for Human Nutrition, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, United States of America
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Department of Internal Medicine and Center for Human Nutrition, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, United States of America
| | - Anil K. Agarwal
- Division of Nutrition and Metabolic Diseases, Department of Internal Medicine and Center for Human Nutrition, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, United States of America
- * E-mail:
| |
Collapse
|