1
|
Chitraju C, Fischer AW, Ambaw YA, Wang K, Yuan B, Hui S, Walther TC, Farese RV. Mice lacking triglyceride synthesis enzymes in adipose tissue are resistant to diet-induced obesity. eLife 2023; 12:RP88049. [PMID: 37782317 PMCID: PMC10545428 DOI: 10.7554/elife.88049] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Triglycerides (TGs) in adipocytes provide the major stores of metabolic energy in the body. Optimal amounts of TG stores are desirable as insufficient capacity to store TG, as in lipodystrophy, or exceeding the capacity for storage, as in obesity, results in metabolic disease. We hypothesized that mice lacking TG storage in adipocytes would result in excess TG storage in cell types other than adipocytes and severe lipotoxicity accompanied by metabolic disease. To test this hypothesis, we selectively deleted both TG synthesis enzymes, DGAT1 and DGAT2, in adipocytes (ADGAT DKO mice). As expected with depleted energy stores, ADGAT DKO mice did not tolerate fasting well and, with prolonged fasting, entered torpor. However, ADGAT DKO mice were unexpectedly otherwise metabolically healthy and did not accumulate TGs ectopically or develop associated metabolic perturbations, even when fed a high-fat diet. The favorable metabolic phenotype resulted from activation of energy expenditure, in part via BAT (brown adipose tissue) activation and beiging of white adipose tissue. Thus, the ADGAT DKO mice provide a fascinating new model to study the coupling of metabolic energy storage to energy expenditure.
Collapse
Affiliation(s)
- Chandramohan Chitraju
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Alexander W Fischer
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Yohannes A Ambaw
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Kun Wang
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Bo Yuan
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Howard Hughes Medical InstituteBostonUnited States
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| |
Collapse
|
2
|
Chaves AB, Zheng D, Johnson JA, Bergman BC, Patinkin ZW, Zaegel V, Biagioni EM, Krassovskaia P, Broskey NT, May LE, Dabelea D, Houmard JA, Boyle KE. Infant Mesenchymal Stem Cell Insulin Action Is Associated With Maternal Plasma Free Fatty Acids, Independent of Obesity Status: The Healthy Start Study. Diabetes 2022; 71:1649-1659. [PMID: 35621990 PMCID: PMC9490356 DOI: 10.2337/db21-0812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022]
Abstract
Preclinical rodent and nonhuman primate models investigating maternal obesity have highlighted the importance of the intrauterine environment in the development of insulin resistance in offspring; however, it remains unclear if these findings can be translated to humans. To investigate possible intrauterine effects in humans, we isolated mesenchymal stem cells (MSCs) from the umbilical cord tissue of infants born to mothers of normal weight or mothers with obesity. Insulin-stimulated glycogen storage was determined in MSCs undergoing myogenesis in vitro. There was no difference in insulin action based on maternal obesity. However, maternal free fatty acid (FFA) concentration, cord leptin, and intracellular triglyceride content were positively correlated with insulin action. Furthermore, MSCs from offspring born to mothers with elevated FFAs displayed elevated activation of the mTOR signaling pathway. Taken together, these data suggest that infants born to mothers with elevated lipid availability have greater insulin action in MSCs, which may indicate upregulation of growth and lipid storage pathways during periods of maternal overnutrition.
Collapse
Affiliation(s)
- Alec B. Chaves
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Donghai Zheng
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Jonathan A. Johnson
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Bryan C. Bergman
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Zachary W. Patinkin
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY
| | - Vincent Zaegel
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Ericka M. Biagioni
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Polina Krassovskaia
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Nicholas T. Broskey
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Linda E. May
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Dana Dabelea
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
- The Lifecourse Epidemiology of Adiposity and Diabetes Center, Aurora, CO
| | - Joseph A. Houmard
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Kristen E. Boyle
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- The Lifecourse Epidemiology of Adiposity and Diabetes Center, Aurora, CO
| |
Collapse
|
3
|
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
|
4
|
Vujić N, Korbelius M, Sachdev V, Rainer S, Zimmer A, Huber A, Radović B, Kratky D. Intestine-specific DGAT1 deficiency improves atherosclerosis in apolipoprotein E knockout mice by reducing systemic cholesterol burden. Atherosclerosis 2020; 310:26-36. [PMID: 32882484 PMCID: PMC7116265 DOI: 10.1016/j.atherosclerosis.2020.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/25/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Background and aims Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is the rate-limiting enzyme catalyzing the final step of triglyceride synthesis by esterifying a diglyceride with a fatty acid. We have previously shown that apolipoprotein E-knockout (ApoE−/−) mice lacking Dgat1 have reduced intestinal cholesterol absorption and potentiated macrophage cholesterol efflux, and consequently, exhibit attenuated atherogenesis. However, he-matopoietic Dgat1 deficiency lacked beneficial effects on atherosclerosis. Due to our recent results on the critical role of intestinal Dgat1 in murine cholesterol homeostasis, we delineated whether intestinal Dgat1 deficiency regulates atherogenesis in mice. Methods We generated intestine-specific Dgat1−/− mice on the ApoE−/− background (iDgat1−/−ApoE−/−) and determined cholesterol homeostasis and atherosclerosis development. Results When fed a Western-type diet, iDgat1−/−ApoE−/− mice exhibited a substantial decrease in fasting plasma cholesterol content in ApoB-containing lipoproteins. Although lipid absorption was delayed, iDgat1−/−ApoE−/− mice had reduced acute and fractional cholesterol absorption coupled with an elevated fecal caloric loss. In line, increased appearance of i.v. administered [3H]cholesterol in duodena and stool of iDgat1−/−ApoE−/− animals suggested potentiated cholesterol elimination. Atherosclerotic lesions were markedly smaller with beneficial alterations in plaque composition as evidenced by reduced macrophage infiltration and necrotic core size despite unaltered collagen content, indicating improved plaque stability. Conclusions Disruption of Dgat1 activity solely in the small intestine of ApoE−/− mice strongly decreased plasma cholesterol levels by abrogating the assimilation of dietary cholesterol, partly by reduced absorption and increased excretion. Consequently, the reduced cholesterol burden significantly attenuated atherogenesis and improved the lesion phenotype in iDgat1−/−ApoE−/− mice.
Collapse
Affiliation(s)
- Nemanja Vujić
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Melanie Korbelius
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Vinay Sachdev
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Andreas Zimmer
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Anton Huber
- Institute of Chemistry, University of Graz, Graz, Austria
| | - Branislav Radović
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
5
|
Cao H. Identification of the major diacylglycerol acyltransferase mRNA in mouse adipocytes and macrophages. BMC BIOCHEMISTRY 2018; 19:11. [PMID: 30547742 PMCID: PMC6293574 DOI: 10.1186/s12858-018-0103-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023]
Abstract
Background Triacylglycerols (TAGs) are the major form of energy storage in eukaryotes. Diacylglycerol acyltransferases (DGATs) catalyze the final and rate-limiting step of TAG biosynthesis. Mammalian DGATs are classified into DGAT1 and DGAT2 subfamilies. It was unclear which DGAT was the major isoform expressed in animal cells. The objective was to identify the major DGAT mRNA expressed in cultured mouse adipocytes and macrophages and compared it to that expressed in tung tree seeds. Methods qPCR evaluated DGAT mRNA levels in mouse 3 T3-L1 adipocytes and RAW264.7 macrophages and tung tree seeds. Results TaqMan qPCR showed that DGAT2 mRNA levels were 10–30 fold higher than DGAT1 in adipocytes and macrophages, and DGAT mRNA levels in adipocytes were 50–100-fold higher than those in macrophages. In contrast, the anti-inflammatory tristetraprolin/zinc finger protein 36 (TTP/ZFP36) mRNA levels were 2–4-fold higher in macrophages than those in adipocytes and similar to DGAT1 in adipocytes but 100-fold higher than DGAT1 in macrophages. SYBR Green qPCR analyses confirmed TaqMan qPCR results. DGAT2 mRNA as the major DGAT mRNA in the mouse cells was similar to that in tung tree seeds where DGAT2 mRNA levels were 10–20-fold higher than DGAT1 or DGAT3. Conclusion The results demonstrated that DGAT2 mRNA was the major form of DGAT mRNA expressed in mouse adipocytes and macrophages and tung tree seeds.
Collapse
Affiliation(s)
- Heping Cao
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, New Orleans, LA, 70124, USA.
| |
Collapse
|
6
|
Cao H, Sethumadhavan K. Cottonseed Extracts and Gossypol Regulate Diacylglycerol Acyltransferase Gene Expression in Mouse Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6022-6030. [PMID: 29807418 DOI: 10.1021/acs.jafc.8b01240] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Plant bioactive polyphenols have been used for the prevention and treatment of various diseases since ancient history. Cotton ( Gossypium hirsutum L.) seeds are classified as glanded or glandless depending on the presence or absence of pigment glands, which contain polyphenolic gossypol. Diacylglycerol acyltransferases (DGATs) are integral membrane proteins that catalyze the last step of triacylglycerol biosynthesis in eukaryotes. Understanding the regulation of DGATs will provide information for therapeutic intervention for obesity and related diseases. However, little was known if DGAT gene expression was regulated by natural products. The objective of this study was to investigate the effects of cottonseed extracts and gossypol on DGAT gene expression in mouse RAW264.7 macrophages. Mouse cells were treated with different concentrations of cottonseed extracts, gossypol, and lipopolysaccharides (LPS) for various times. Quantitative polymerase chain reaction assay showed that coat extract of glanded seeds had a modest effect on DGAT1 and minimal effect on DGAT2 mRNA levels. Kernel extract of glanded seeds had a minimal effect on DGAT1 but increased DGAT2 mRNA levels more than 20-fold. Coat extract of glandless seeds and LPS had minimal effects on DGAT mRNA levels. Kernel extract of glandless seeds did not have much effect on DGAT1 and slightly increased DGAT2 mRNA levels. Gossypol increased DGAT1 and DGAT2 mRNA levels by up to three-fold and more than 80-fold, respectively. The coefficient correlations ( R2) between DGAT2 mRNA levels and glanded kernel extract and gossypol concentrations were 0.82-0.99. This study suggests that Dgat2 is an inducible gene rapidly responding to stimulators such as polyphenols whose protein product DGAT2 plays an important role in fat biosynthesis. We conclude that gossypol and ethanol extract from glanded cottonseed kernel are strong stimulators of DGAT2 gene expression and that they may be novel agents for intervention of lipid-related dysfunction via increasing DGAT2 gene expression in target tissues.
Collapse
Affiliation(s)
- Heping Cao
- U.S. Department of Agriculture, Agricultural Research Service , Southern Regional Research Center , New Orleans , Louisiana 70124 , United States
| | - Kandan Sethumadhavan
- U.S. Department of Agriculture, Agricultural Research Service , Southern Regional Research Center , New Orleans , Louisiana 70124 , United States
| |
Collapse
|
7
|
Farese RV, Sajan MP, Standaert ML. Insulin-Sensitive Protein Kinases (Atypical Protein Kinase C and Protein Kinase B/Akt): Actions and Defects in Obesity and Type II Diabetes. Exp Biol Med (Maywood) 2016; 230:593-605. [PMID: 16179727 DOI: 10.1177/153537020523000901] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Glucose transport into muscle is the initial process in glucose clearance and is uniformly defective in insulin-resistant conditions of obesity, metabolic syndrome, and Type II diabetes mellitus. Insulin regulates glucose transport by activating insulin receptor substrate-1 (IRS-1)-dependent phosphatidylinositol 3-kinase (PI3K) which, via increases in PI-3, 4, 5-triphosphate (PIP3), activates atypical protein kinase C (aPKC) and protein kinase B (PKB/Akt). Here, we review (i) the evidence that both aPKC and PKB are required for insulin-stimulated glucose transport, (ii) abnormalities in muscle aPKC/PKB activation seen in obesity and diabetes, and (iii) mechanisms for impaired aPKC activation in insulin-resistant conditions. In most cases, defective muscle aPKC/PKB activation reflects both impaired activation of IRS-1/PI3K, the upstream activator of aPKC and PKB in muscle and, in the case of aPKC, poor responsiveness to PIP3, the lipid product of PI3K. Interestingly, insulin-sensitizing agents (e.g., thiazolidinediones, metformin) improve aPKC activation by insulin in vivo and PIP3 in vitro, most likely by activating 5′-adenosine monophosphate-activated protein kinase, which favorably alters intracellular lipid metabolism. Differently from muscle, aPKC activation in the liver is dependent on IRS-2/PI3K rather than IRS-1/PI3K and, surprisingly, the activation of IRS-2/PI3K and aPKC is conserved in high-fat feeding, obesity, and diabetes. This conservation has important implications, as continued activation of hepatic aPKC in hyperinsulinemic states may increase the expression of sterol regulatory element binding protein-1c, which controls genes that increase hepatic lipid synthesis. On the other hand, the defective activation of IRS-1/PI3K and PKB, as seen in diabetic liver, undoubtedly and importantly contributes to increases in hepatic glucose output. Thus, the divergent activation of aPKC and PKB in the liver may explain why some hepatic actions of insulin (e.g., aPKC-dependent lipid synthesis) are increased while other actions (e.g., PKB-dependent glucose metabolism) are diminished. This may explain the paradox that the liver secretes excessive amounts of both very low density lipoprotein triglycerides and glucose in Type II diabetes. Previous reviews from our laboratory that have appeared in the Proceedings have provided essentials on phospholipid-signaling mechanisms used by insulin to activate several protein kinases that seem to be important in mediating the metabolic effects of insulin. During recent years, there have been many new advances in our understanding of how these lipid-dependent protein kinases function during insulin action and why they fail to function in states of insulin resistance. The present review will attempt to summarize what we believe are some of the more important advances.
Collapse
Affiliation(s)
- Robert V Farese
- James A. Haley Veterans Administration Hospital Research Service and Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
8
|
Vujic N, Porter Abate J, Schlager S, David T, Kratky D, Koliwad SK. Acyl-CoA:Diacylglycerol Acyltransferase 1 Expression Level in the Hematopoietic Compartment Impacts Inflammation in the Vascular Plaques of Atherosclerotic Mice. PLoS One 2016; 11:e0156364. [PMID: 27223895 PMCID: PMC4880185 DOI: 10.1371/journal.pone.0156364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/12/2016] [Indexed: 01/09/2023] Open
Abstract
The final step of triacylglycerol synthesis is catalyzed by acyl-CoA:diacylglycerol acyltransferases (DGATs). We have previously shown that ApoE-/-Dgat1-/- mice are protected from developing atherosclerosis in association with reduced foam cell formation. However, the role of DGAT1, specifically in myeloid and other hematopoietic cell types, in determining this protective phenotype is unknown. To address this question, we reconstituted the bone marrow of irradiated Ldlr-/-mice with that from wild-type (WT→ Ldlr-/-) and Dgat1-/-(Dgat1-/-→ Ldlr-/-) donor mice. We noted that DGAT1 in the hematopoietic compartment exerts a sex-specific effect on systemic cholesterol homeostasis. However, both male and female Dgat1-/-→ Ldlr-/-mice had higher circulating neutrophil and lower lymphocyte counts than control mice, suggestive of a classical inflammatory phenotype. Moreover, specifically examining the aortae of these mice revealed that Dgat1-/-→ Ldlr-/-mice have atherosclerotic plaques with increased macrophage content. This increase was coupled to a reduced plaque collagen content, leading to a reduced collagen-to-macrophage ratio. Together, these findings point to a difference in the inflammatory contribution to plaque composition between Dgat1-/-→ Ldlr-/-and control mice. By contrast, DGAT1 deficiency did not affect the transcriptional responses of cultured macrophages to lipoprotein treatment in vitro, suggesting that the alterations seen in the plaques of Dgat1-/-→ Ldlr-/-mice in vivo do not reflect a cell intrinsic effect of DGAT1 in macrophages. We conclude that although DGAT1 in the hematopoietic compartment does not impact the overall lipid content of atherosclerotic plaques, it exerts reciprocal effects on inflammation and fibrosis, two processes that control plaque vulnerability.
Collapse
Affiliation(s)
- Nemanja Vujic
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Jess Porter Abate
- Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Stefanie Schlager
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Tovo David
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Suneil K. Koliwad
- Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
9
|
Li Y, Xu S, Zhang X, Yi Z, Cichello S. Skeletal intramyocellular lipid metabolism and insulin resistance. BIOPHYSICS REPORTS 2015; 1:90-98. [PMID: 26942223 PMCID: PMC4762133 DOI: 10.1007/s41048-015-0013-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/10/2015] [Indexed: 12/24/2022] Open
Abstract
Lipids stored in skeletal muscle cells are known as intramyocellular lipid (IMCL). Disorders involving IMCL and its causative factor, circulatory free fatty acids (FFAs), induce a toxic state and ultimately result in insulin resistance (IR) in muscle tissue. On the other hand, intramuscular triglyceride (IMTG), the most abundant component of IMCL and an essential energy source for active skeletal muscle, is different from other IMCLs, as it is stored in lipid droplets and plays a pivotal role in skeletal muscle energy homeostasis. This review discusses the association of FFA-induced ectopic lipid accumulation and IR, with specific emphasis on the relationship between IMCL/IMTG metabolism and IR.
Collapse
Affiliation(s)
- Yiran Li
- Department of Biological Science and Biotechnology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China ; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China ; University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xuelin Zhang
- Capital University of Physical Education and Sport, Beijing, 100191 China
| | - Zongchun Yi
- Department of Biological Science and Biotechnology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Simon Cichello
- School of Life Sciences, La Trobe University, Melbourne, VIC 3086 Australia
| |
Collapse
|
10
|
Phosphorylation and function of DGAT1 in skeletal muscle cells. BIOPHYSICS REPORTS 2015; 1:41-50. [PMID: 26942218 PMCID: PMC4762128 DOI: 10.1007/s41048-015-0004-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/27/2015] [Indexed: 12/31/2022] Open
Abstract
Aberrant intramuscular triacylglycerol (TAG) storage in human skeletal muscle is closely related to insulin insensitivity. Excessive lipid storage can induce insulin resistance of skeletal muscle, and under severe conditions, lead to type 2 diabetes. The balance of interconversion between diacylglycerol and TAG greatly influences lipid storage and utilization. Diacylglycerol O-acyltransferase 1 (DGAT1) plays a key role in this process, but its activation and phosphorylation requires further dissection. In this study, 12 putative conserved phosphorylation sites of DGAT1 were identified by examining amino acid conservation of DGAT1 in different species. Another 12 putative phosphorylation sites were also found based on bioinformatics predictions and previous reports. Meanwhile, several phosphorylation sites of DGAT1 were verified by phosphorylation mass spectrometry analysis of purified DGAT1 from mouse myoblast C2C12 cells. Using single point mutations, a regulatory role of 3 putative phosphorylation sites was dissected. Finally, using truncation mutations, a potential domain of DGAT1 that was involved in the regulation of enzymatic activity was revealed. This study provides useful information for further understanding DGAT1 activity regulation.
Collapse
|
11
|
Discovery and optimization of adamantane carboxylic acid derivatives as potent diacylglycerol acyltransferase 1 inhibitors for the potential treatment of obesity and diabetes. Eur J Med Chem 2015. [DOI: 10.1016/j.ejmech.2015.06.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
12
|
He S, Hong Q, Lai Z, Yang DX, Ting PC, Kuethe JT, Cernak TA, Dykstra KD, Sperbeck DM, Wu Z, Yu Y, Yang GX, Jian T, Liu J, Guiadeen D, Krikorian AD, Sonatore LM, Wiltsie J, Liu J, Gorski JN, Chung CC, Gibson JT, Lisnock J, Xiao J, Wolff M, Tong SX, Madeira M, Karanam BV, Shen DM, Balkovec JM, Pinto S, Nargund RP, DeVita RJ. Discovery of a Potent and Selective DGAT1 Inhibitor with a Piperidinyl-oxy-cyclohexanecarboxylic Acid Moiety. ACS Med Chem Lett 2014; 5:1082-7. [PMID: 25349648 DOI: 10.1021/ml5003426] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 09/08/2014] [Indexed: 02/01/2023] Open
Abstract
We report the discovery of a novel series of DGAT1 inhibitors in the benzimidazole class with a piperdinyl-oxy-cyclohexanecarboxylic acid moiety. This novel series possesses significantly improved selectivity against the A2A receptor, no ACAT1 off-target activity at 10 μM, and higher aqueous solubility and free fraction in plasma as compared to the previously reported pyridyl-oxy-cyclohexanecarboxylic acid series. In particular, 5B was shown to possess an excellent selectivity profile by screening it against a panel of more than 100 biological targets. Compound 5B significantly reduces lipid excursion in LTT in mouse and rat, demonstrates DGAT1 mediated reduction of food intake and body weight in mice, is negative in a 3-strain Ames test, and appears to distribute preferentially in the liver and the intestine in mice. We believe this lead series possesses significant potential to identify optimized compounds for clinical development.
Collapse
Affiliation(s)
- Shuwen He
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qingmei Hong
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhong Lai
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - David X. Yang
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Pauline C. Ting
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jeffrey T. Kuethe
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Timothy A. Cernak
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kevin D. Dykstra
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Donald M. Sperbeck
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhicai Wu
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Yang Yu
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ginger X. Yang
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Tianying Jian
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jian Liu
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Deodial Guiadeen
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Arto D. Krikorian
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lisa M. Sonatore
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Judyann Wiltsie
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jinqi Liu
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Judith N. Gorski
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Christine C. Chung
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jack T. Gibson
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - JeanMarie Lisnock
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jianying Xiao
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Wolff
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Sharon X. Tong
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Maria Madeira
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Bindhu V. Karanam
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Dong-Ming Shen
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - James M. Balkovec
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Shirly Pinto
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ravi P. Nargund
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert J. DeVita
- Early Development and Discovery
Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
13
|
Identification and characterization of a type-2 diacylglycerol acyltransferase (DGAT2) from Rhodosporidium diobovatum. Antonie van Leeuwenhoek 2014; 106:1127-37. [DOI: 10.1007/s10482-014-0282-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/10/2014] [Indexed: 10/24/2022]
|
14
|
Sahebkar A, Chew GT, Watts GF. Recent advances in pharmacotherapy for hypertriglyceridemia. Prog Lipid Res 2014; 56:47-66. [PMID: 25083925 DOI: 10.1016/j.plipres.2014.07.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/10/2014] [Accepted: 07/18/2014] [Indexed: 12/20/2022]
Abstract
Elevated plasma triglyceride (TG) concentrations are associated with an increased risk of atherosclerotic cardiovascular disease (CVD), hepatic steatosis and pancreatitis. Existing pharmacotherapies, such as fibrates, n-3 polyunsaturated fatty acids (PUFAs) and niacin, are partially efficacious in correcting elevated plasma TG. However, several new TG-lowering agents are in development that can regulate the transport of triglyceride-rich lipoproteins (TRLs) by modulating key enzymes, receptors or ligands involved in their metabolism. Balanced dual peroxisome proliferator-activated receptor (PPAR) α/γ agonists, inhibitors of microsomal triglyceride transfer protein (MTTP) and acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1), incretin mimetics, and apolipoprotein (apo) B-targeted antisense oligonucleotides (ASOs) can all decrease the production and secretion of TRLs; inhibitors of cholesteryl ester transfer protein (CETP) and angiopoietin-like proteins (ANGPTLs) 3 and 4, monoclonal antibodies (Mabs) against proprotein convertase subtilisin/kexin type 9 (PCSK9), apoC-III-targeted ASOs, selective peroxisome proliferator-activated receptor modulators (SPPARMs), and lipoprotein lipase (LPL) gene replacement therapy (alipogene tiparvovec) enhance the catabolism and clearance of TRLs; dual PPAR-α/δ agonists and n-3 polyunsaturated fatty acids can lower plasma TG by regulating both TRL secretion and catabolism. Varying degrees of TG reduction have been reported with the use of these therapies, and for some agents such as CETP inhibitors and PCSK9 Mabs findings have not been consistent. Whether they reduce CVD events has not been established. Trials investigating the effect of CETP inhibitors (anacetrapib and evacetrapib) and PCSK9 Mabs (AMG-145 and REGN727/SAR236553) on CVD outcomes are currently in progress, although these agents also regulate LDL metabolism and, in the case of CETP inhibitors, HDL metabolism. Further to CVD risk reduction, these new treatments might also have a potential role in the management of diabetes and non-alcoholic fatty liver disease owing to their insulin-sensitizing action (PPAR-α/γ agonists) and potential capacity to decrease hepatic TG accumulation (PPAR-α/δ agonists and DGAT-1 inhibitors), but this needs to be tested in future trials. We summarize the clinical trial findings regarding the efficacy and safety of these novel therapies for hypertriglyceridemia.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Gerard T Chew
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Australia.
| |
Collapse
|
15
|
Naik R, Obiang-Obounou BW, Kim M, Choi Y, Lee HS, Lee K. Therapeutic Strategies for Metabolic Diseases: Small-Molecule Diacylglycerol Acyltransferase (DGAT) Inhibitors. ChemMedChem 2014; 9:2410-24. [DOI: 10.1002/cmdc.201402069] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Indexed: 11/07/2022]
|
16
|
Cao H, Shockey JM, Klasson KT, Chapital DC, Mason CB, Scheffler BE. Developmental regulation of diacylglycerol acyltransferase family gene expression in tung tree tissues. PLoS One 2013; 8:e76946. [PMID: 24146944 PMCID: PMC3795650 DOI: 10.1371/journal.pone.0076946] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/26/2013] [Indexed: 11/29/2022] Open
Abstract
Diacylglycerol acyltransferases (DGAT) catalyze the final and rate-limiting step of triacylglycerol (TAG) biosynthesis in eukaryotic organisms. DGAT genes have been identified in numerous organisms. Multiple isoforms of DGAT are present in eukaryotes. We previously cloned DGAT1 and DGAT2 genes of tung tree (Vernicia fordii), whose novel seed TAGs are useful in a wide range of industrial applications. The objective of this study was to understand the developmental regulation of DGAT family gene expression in tung tree. To this end, we first cloned a tung tree gene encoding DGAT3, a putatively soluble form of DGAT that possesses 11 completely conserved amino acid residues shared among 27 DGAT3s from 19 plant species. Unlike DGAT1 and DGAT2 subfamilies, DGAT3 is absent from animals. We then used TaqMan and SYBR Green quantitative real-time PCR, along with northern and western blotting, to study the expression patterns of the three DGAT genes in tung tree tissues. Expression results demonstrate that 1) all three isoforms of DGAT genes are expressed in developing seeds, leaves and flowers; 2) DGAT2 is the major DGAT mRNA in tung seeds, whose expression profile is well-coordinated with the oil profile in developing tung seeds; and 3) DGAT3 is the major form of DGAT mRNA in tung leaves, flowers and immature seeds prior to active tung oil biosynthesis. These results suggest that DGAT2 is probably the major TAG biosynthetic isoform in tung seeds and that DGAT3 gene likely plays a significant role in TAG metabolism in other tissues. Therefore, DGAT2 should be a primary target for tung oil engineering in transgenic organisms.
Collapse
Affiliation(s)
- Heping Cao
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, Commodity Utilization Research Unit, New Orleans, Louisiana, United States of America
- * E-mail:
| | - Jay M. Shockey
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, Commodity Utilization Research Unit, New Orleans, Louisiana, United States of America
| | - K. Thomas Klasson
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, Commodity Utilization Research Unit, New Orleans, Louisiana, United States of America
| | - Dorselyn C. Chapital
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, Commodity Utilization Research Unit, New Orleans, Louisiana, United States of America
| | - Catherine B. Mason
- U.S. Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, Commodity Utilization Research Unit, New Orleans, Louisiana, United States of America
| | - Brian E. Scheffler
- U.S. Department of Agriculture, Agricultural Research Service, Genomics and Bioinformatics Research Unit, Stoneville, Mississippi, United States of America
| |
Collapse
|
17
|
Cao H, Chapital DC, Howard OD, Deterding LJ, Mason CB, Shockey JM, Klasson KT. Expression and purification of recombinant tung tree diacylglycerol acyltransferase 2. Appl Microbiol Biotechnol 2012; 96:711-27. [PMID: 22270236 PMCID: PMC11338361 DOI: 10.1007/s00253-012-3869-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 12/19/2011] [Accepted: 12/23/2011] [Indexed: 12/11/2022]
Abstract
Diacylglycerol acyltransferases (DGATs) esterify sn-1,2-diacylglycerol with a long-chain fatty acyl-CoA, the last and rate-limiting step of triacylglycerol (TAG) biosynthesis in eukaryotic organisms. At least 74 DGAT2 sequences from 61 organisms have been identified, but the expression of any DGAT2 as a partial or full-length protein in Escherichia coli had not been reported. The main objective of this study was to express and purify recombinant DGAT2 (rDGAT2) from E. coli for antigen production with a minor objective to compare rDGAT2 expression in yeast. A plasmid was engineered to express tung tree DGAT2 fused to maltose binding protein and poly-histidine (His) affinity tags. Immunoblotting showed that rDGAT2 was detected in the soluble, insoluble, and membrane fractions. The rDGAT2 in the soluble fraction was partially purified by amylose resin, nickel-nitrilotriacetic agarose (Ni-NTA) beads, and tandem affinity chromatography. Multiple proteins co-purified with rDGAT2. Size exclusion chromatography estimated the size of the rDGAT2-enriched fraction to be approximately eight times the monomer size. Affinity-purified rDGAT2 fractions had a yellow tint and contained fatty acids. The rDGAT2 in the insoluble fraction was partially solubilized by seven detergents with SDS being the most effective. Recombinant DGAT2 was purified to near homogeneity by SDS solubilization and Ni-NTA affinity chromatography. Mass spectrometry identified rDGAT2 as a component in the bands corresponding to the monomer and dimer forms as observed by SDS-PAGE. Protein bands with monomer and dimer sizes were also observed in the microsomal membranes of Saccharomyces cerevisiae expressing hemagglutinin-tagged DGAT2. Nonradioactive assay showed TAG synthesis activity of DGAT2 from yeast but not E. coli. The results suggest that rDGAT2 is present as monomer and dimer forms on SDS-PAGE, associated with other proteins, lipids, and membranes, and that post-translational modification of rDGAT2 may be required for its enzymatic activity and/or the E. coli protein is misfolded.
Collapse
Affiliation(s)
- Heping Cao
- Commodity Utilization Research Unit, Southern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, 1100 Robert E. Lee Blvd, New Orleans, LA 70124, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Habegger KM, Matzke D, Ottaway N, Hembree J, Holland J, Raver C, Mansfeld J, Müller TD, Perez-Tilve D, Pfluger PT, Lee SJ, Diaz-Meco M, Moscat J, Leitges M, Tschöp MH, Hofmann SM. Role of adipose and hepatic atypical protein kinase C lambda (PKCλ) in the development of obesity and glucose intolerance. Adipocyte 2012; 1:203-214. [PMID: 23700535 PMCID: PMC3609106 DOI: 10.4161/adip.20891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PKCλ, an atypical member of the multifunctional protein kinase C family, has been implicated in the regulation of insulin-stimulated glucose transport and of the intracellular immune response. To further elucidate the role of this cellular regulator in diet-induced obesity and insulin resistance, we generated both liver (PKC-Alb) and adipose tissue (PKC-Ap2) specific knockout mice. Body weight, fat mass, food intake, glucose homeostasis and energy expenditure were evaluated in mice maintained on either chow or high fat diet (HFD). Ablation of PKCλ from the adipose tissue resulted in mice that were indistinguishable from their wild-type littermates. However, PKC-Alb mice were resistant to diet-induced obesity (DIO). Surprisingly this DIO resistance was not associated with either a reduction in caloric intake or an increase in energy expenditure as compared with their wild-type littermates. Furthermore, these mice displayed an improvement in glucose tolerance. When maintained on chow diet, these mice were similar to wild types in respect to body weight and fat mass, yet insulin sensitivity was impaired compared with wt littermates. Taken together these data suggest that hepatic PKCλ is modulating insulin-mediated glucose turnover and response to high fat diet feeding, thus offering a deeper understanding of an important target for anti-obesity therapeutics.
Collapse
|
19
|
Yeh VSC, Beno DWA, Brodjian S, Brune ME, Cullen SC, Dayton BD, Dhaon MK, Falls HD, Gao J, Grihalde N, Hajduk P, Hansen TM, Judd AS, King AJ, Klix RC, Larson KJ, Lau YY, Marsh KC, Mittelstadt SW, Plata D, Rozema MJ, Segreti JA, Stoner EJ, Voorbach MJ, Wang X, Xin X, Zhao G, Collins CA, Cox BF, Reilly RM, Kym PR, Souers AJ. Identification and preliminary characterization of a potent, safe, and orally efficacious inhibitor of acyl-CoA:diacylglycerol acyltransferase 1. J Med Chem 2012; 55:1751-7. [PMID: 22263872 DOI: 10.1021/jm201524g] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A high-throughput screen against human DGAT-1 led to the identification of a core structure that was subsequently optimized to afford the potent, selective, and orally bioavailable compound 14. Oral administration at doses ≥0.03 mg/kg significantly reduced postprandial triglycerides in mice following an oral lipid challenge. Further assessment in both acute and chronic safety pharmacology and toxicology studies demonstrated a clean profile up to high plasma levels, thus culminating in the nomination of 14 as clinical candidate ABT-046.
Collapse
Affiliation(s)
- Vince S C Yeh
- Global Pharmaceutical Research and Development, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6100, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Cao J, Zhou Y, Peng H, Huang X, Stahler S, Suri V, Qadri A, Gareski T, Jones J, Hahm S, Perreault M, McKew J, Shi M, Xu X, Tobin JF, Gimeno RE. Targeting Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) with small molecule inhibitors for the treatment of metabolic diseases. J Biol Chem 2011; 286:41838-41851. [PMID: 21990351 DOI: 10.1074/jbc.m111.245456] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is one of two known DGAT enzymes that catalyze the final step in triglyceride synthesis. Findings from genetically modified mice as well as pharmacological studies suggest that inhibition of DGAT1 is a promising strategy for the treatment of obesity and type 2 diabetes. Here we characterize a tool DGAT1 inhibitor compound, T863. We found that T863 is a potent inhibitor for both human and mouse DGAT1 in vitro, which acts on the acyl-CoA binding site of DGAT1 and inhibits DGAT1-mediated triacylglycerol formation in cells. In an acute lipid challenge model, oral administration of T863 significantly delayed fat absorption and resulted in lipid accumulation in the distal small intestine of mice, mimicking the effects of genetic ablation of DGAT1. In diet-induced obese mice, oral administration of T863 for 2 weeks caused weight loss, reduction in serum and liver triglycerides, and improved insulin sensitivity. In addition to the expected triglyceride-lowering activity, T863 also lowered serum cholesterol. Hepatic IRS2 protein was dramatically up-regulated in mice treated with T863, possibly contributing to improved insulin sensitivity. In differentiated 3T3-L1 adipocytes, T863 enhanced insulin-stimulated glucose uptake, suggesting a possible role for adipocytes to improve insulin sensitivity upon DGAT1 inhibition. These results reveal novel mechanistic insights into the insulin-sensitizing effects of DGAT1 inhibition in mouse models. Taken together, our study provides a comprehensive evaluation of a small molecule inhibitor for DGAT1 and suggests that pharmacological inhibition of DGAT1 holds promise in treating diverse metabolic disorders.
Collapse
Affiliation(s)
- Jingsong Cao
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140.
| | - Yingjiang Zhou
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Haibing Peng
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Xinyi Huang
- Screening Sciences, Pfizer, Collegeville, Pennsylvania 19426
| | - Shannon Stahler
- Screening Sciences, Pfizer, Collegeville, Pennsylvania 19426
| | - Vipin Suri
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Ariful Qadri
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Tiffany Gareski
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Juli Jones
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Seung Hahm
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Mylene Perreault
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - John McKew
- Exploratory Medicinal Chemistry, Pfizer, Cambridge, Massachusetts 02140
| | - Mengxiao Shi
- Discovery Pharmacokinetics, Pfizer, Andover, Massachusetts 01810
| | - Xin Xu
- Discovery Pharmacokinetics, Pfizer, Andover, Massachusetts 01810
| | - James F Tobin
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140
| | - Ruth E Gimeno
- Biocorrection Research Unit, Pfizer, Cambridge, Massachusetts 02140.
| |
Collapse
|
21
|
Cao H. Structure-function analysis of diacylglycerol acyltransferase sequences from 70 organisms. BMC Res Notes 2011; 4:249. [PMID: 21777418 PMCID: PMC3157451 DOI: 10.1186/1756-0500-4-249] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/21/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diacylglycerol acyltransferase families (DGATs) catalyze the final and rate-limiting step of triacylglycerol (TAG) biosynthesis in eukaryotic organisms. Understanding the roles of DGATs will help to create transgenic plants with value-added properties and provide clues for therapeutic intervention for obesity and related diseases. The objective of this analysis was to identify conserved sequence motifs and amino acid residues for better understanding of the structure-function relationship of these important enzymes. RESULTS 117 DGAT sequences from 70 organisms including plants, animals, fungi and human are obtained from database search using tung tree DGATs. Phylogenetic analysis separates these proteins into DGAT1 and DGAT2 subfamilies. These DGATs are integral membrane proteins with more than 40% of the total amino acid residues being hydrophobic. They have similar properties and amino acid composition except that DGAT1s are approximately 20 kDa larger than DGAT2s. DGAT1s and DGAT2s have 41 and 16 completely conserved amino acid residues, respectively, although only two of them are shared by all DGATs. These residues are distributed in 7 and 6 sequence blocks for DGAT1s and DGAT2s, respectively, and located at the carboxyl termini, suggesting the location of the catalytic domains. These conserved sequence blocks do not contain the putative neutral lipid-binding domain, mitochondrial targeting signal, or ER retrieval motif. The importance of conserved residues has been demonstrated by site-directed and natural mutants. CONCLUSIONS This study has identified conserved sequence motifs and amino acid residues in all 117 DGATs and the two subfamilies. None of the completely conserved residues in DGAT1s and DGAT2s is present in recently reported isoforms in the multiple sequences alignment, raising an important question how proteins with completely different amino acid sequences could perform the same biochemical reaction. The sequence analysis should facilitate studying the structure-function relationship of DGATs with the ultimate goal to identify critical amino acid residues for engineering superb enzymes in metabolic engineering and selecting enzyme inhibitors in therapeutic application for obesity and related diseases.
Collapse
Affiliation(s)
- Heping Cao
- Commodity Utilization Research Unit, Southern Regional Research Center, Agricultural Research Service, U,S, Department of Agriculture, 1100 Robert E, Lee Blvd,, New Orleans, Louisiana 70124, USA.
| |
Collapse
|
22
|
Liu L, Yu S, Khan RS, Ables GP, Bharadwaj KG, Hu Y, Huggins LA, Eriksson JW, Buckett LK, Turnbull AV, Ginsberg HN, Blaner WS, Huang LS, Goldberg IJ. DGAT1 deficiency decreases PPAR expression and does not lead to lipotoxicity in cardiac and skeletal muscle. J Lipid Res 2011; 52:732-44. [PMID: 21205704 DOI: 10.1194/jlr.m011395] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Diacylglycerol (DAG) acyl transferase 1 (Dgat1) knockout ((-/-)) mice are resistant to high-fat-induced obesity and insulin resistance, but the reasons are unclear. Dgat1(-/-) mice had reduced mRNA levels of all three Ppar genes and genes involved in fatty acid oxidation in the myocardium of Dgat1(-/-) mice. Although DGAT1 converts DAG to triglyceride (TG), tissue levels of DAG were not increased in Dgat1(-/-) mice. Hearts of chow-diet Dgat1(-/-) mice were larger than those of wild-type (WT) mice, but cardiac function was normal. Skeletal muscles from Dgat1(-/-) mice were also larger. Muscle hypertrophy factors phospho-AKT and phospho-mTOR were increased in Dgat1(-/-) cardiac and skeletal muscle. In contrast to muscle, liver from Dgat1(-/-) mice had no reduction in mRNA levels of genes mediating fatty acid oxidation. Glucose uptake was increased in cardiac and skeletal muscle in Dgat1(-/-) mice. Treatment with an inhibitor specific for DGAT1 led to similarly striking reductions in mRNA levels of genes mediating fatty acid oxidation in cardiac and skeletal muscle. These changes were reproduced in cultured myocytes with the DGAT1 inhibitor, which also blocked the increase in mRNA levels of Ppar genes and their targets induced by palmitic acid. Thus, loss of DGAT1 activity in muscles decreases mRNA levels of genes involved in lipid uptake and oxidation.
Collapse
Affiliation(s)
- Li Liu
- Division of Preventive Medicine and Nutrition, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhang XD, Yan JW, Yan GR, Sun XY, Ji J, Li YM, Hu YH, Wang HY. Pharmacological inhibition of diacylglycerol acyltransferase 1 reduces body weight gain, hyperlipidemia, and hepatic steatosis in db/db mice. Acta Pharmacol Sin 2010; 31:1470-7. [PMID: 21052084 DOI: 10.1038/aps.2010.104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIM To test whether pharmacological inhibition of Diacylglycerol acyltransferase 1 (DGAT1) by a small-molecule inhibitor H128 can improve metabolism disorders in leptin receptor-deficient db/db mice. METHODS To investigate the effect of H128 on intestinal fat absorption,db/db mice were acutely given a bolus of corn oil by gavage. The mice were further orally administered H128 (3 and 10 mg/kg) for 5 weeks. Blood glucose, lipids, insulin, ALT, and AST as well as hepatic triglycerides were measured. The insulin tolerance test was performed to evaluate insulin sensitivity. The expression of genes involved in fatty acid oxidation was detected by RT-PCR. RESULTS Oral administration of H128 (10 mg/kg) acutely inhibited intestinal fat absorption following a lipid challenge in db/db mice. Chronic treatment with H128 significantly inhibited body weight gain, decreased food intake, and induced a pronounced reduction of serum triglycerides. In addition, H128 treatment markedly ameliorated hepatic steatosis, characterized by decreased liver weight, lipid droplets, and triglyceride content as well as serum ALT and AST levels. Furthermore, H128 treatment increased the expression of the CPT1 and PPARα genes in liver, suggesting that H128 enhanced fatty acid oxidation in db/db mice. However, neither blood glucose nor insulin tolerance was affected by H128 treatment throughout the 5-week experimental period. CONCLUSION DGAT1 may be an effective therapeutic target for the treatment of obesity, hyperlipidemia and hepatic steatosis.
Collapse
|
24
|
King AJ, Judd AS, Souers AJ. Inhibitors of diacylglycerol acyltransferase: a review of 2008 patents. Expert Opin Ther Pat 2010; 20:19-29. [PMID: 20021283 DOI: 10.1517/13543770903499305] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Postprandial hypertriglyceridemia has been identified as a major independent risk factor for future cardiovascular events. Therefore, inhibition of triglyceride synthesis has enormous therapeutic potential in the treatment of metabolic disorders. Diacylglycerol acyltransferase (DGAT) enzymes catalyze the final and only committed step in triglyceride biosynthesis and have thus been identified as potential therapeutic targets to combat human cardio-metabolic diseases. OBJECTIVE/METHOD Significant interest in DGAT-1 inhibitors has emerged in the last several years. To provide a perspective on the exciting features of this enzyme for targeting metabolic diseases, a summary of the biology and pharmacology surrounding the DGAT enzymes is presented. Following this is a discussion of the various chemotypes that have been disclosed within relevant patent applications published in 2008. Specifically, the similarities and differences of the chemical structures and the biological data that are provided to support the corresponding claims are presented. CONCLUSION Small molecule and biologic-based DGAT inhibitors have been successfully used for the preclinical validation of DGAT enzymes as targets for the treatment of metabolic diseases. Given the advanced stage in which some of the chemical matter resides, it is expected that DGAT inhibitors will enter the clinic in the coming years.
Collapse
Affiliation(s)
- Andrew J King
- Global Pharmaceutical Research & Development, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL 60064, USA
| | | | | |
Collapse
|
25
|
Lee B, Fast AM, Zhu J, Cheng JX, Buhman KK. Intestine-specific expression of acyl CoA:diacylglycerol acyltransferase 1 reverses resistance to diet-induced hepatic steatosis and obesity in Dgat1-/- mice. J Lipid Res 2010; 51:1770-80. [PMID: 20147738 DOI: 10.1194/jlr.m002311] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mice deficient in acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1), a key enzyme in triacylglycerol (TG) biosynthesis, are resistant to high-fat (HF) diet-induced hepatic steatosis and obesity. DGAT1-deficient (Dgat1-/-) mice have no defect in quantitative absorption of dietary fat; however, they have abnormally high levels of TG stored in the cytoplasm of enterocytes, and they have a reduced postprandial triglyceridemic response. We generated mice expressing DGAT1 only in the intestine (Dgat1IntONLY) to determine whether this phenotype contributes to resistance to HF diet-induced hepatic steatosis and obesity in Dgat1-/- mice. Despite lacking DGAT1 in liver and adipose tissue, we found that Dgat1IntONLY mice are not resistant to HF diet-induced hepatic steatosis or obesity. The results presented demonstrate that intestinal DGAT1 stimulates dietary fat secretion out of enterocytes and that altering this cellular function alters the fate of dietary fat in specific tissues.
Collapse
Affiliation(s)
- Bonggi Lee
- Department of Foods and Nutrition, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
26
|
King AJ, Segreti JA, Larson KJ, Souers AJ, Kym PR, Reilly RM, Zhao G, Mittelstadt SW, Cox BF. Diacylglycerol acyltransferase 1 inhibition lowers serum triglycerides in the Zucker fatty rat and the hyperlipidemic hamster. J Pharmacol Exp Ther 2009; 330:526-31. [PMID: 19478132 DOI: 10.1124/jpet.109.154047] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Acyl CoA/diacylglycerol acyltransferase (DGAT) 1 is one of two known DGAT enzymes that catalyze the final and only committed step in triglyceride biosynthesis. The purpose of this study was to test the hypothesis that chronic inhibition of DGAT-1 with a small-molecule inhibitor will reduce serum triglyceride concentrations in both genetic and diet-induced models of hypertriglyceridemia. Zucker fatty rats and diet-induced dyslipidemic hamsters were dosed orally with A-922500 (0.03, 0.3, and 3-mg/kg), a potent and selective DGAT-1 inhibitor, for 14 days. Serum triglycerides were significantly reduced by the 3 mg/kg dose of the DGAT-1 inhibitor in both the Zucker fatty rat (39%) and hyperlipidemic hamster (53%). These serum triglyceride changes were accompanied by significant reductions in free fatty acid levels by 32% in the Zucker fatty rat and 55% in the hyperlipidemic hamster. In addition, high-density lipoprotein-cholesterol was significantly increased (25%) in the Zucker fatty rat by A-922500 administered at 3 mg/kg. This study provides the first report that inhibition of DGAT-1, the final and only committed step of triglyceride synthesis, with a selective small-molecule inhibitor, significantly reduces serum triglyceride levels in both genetic and diet-induced animal models of hypertriglyceridemia. The results of this study support further investigation of DGAT-1 inhibition as a novel therapeutic approach to the treatment of hypertriglyceridemia in humans, and they suggest that inhibition of triglyceride synthesis may have more diverse beneficial effects on serum lipid profiles beyond triglyceride lowering.
Collapse
Affiliation(s)
- Andrew J King
- Department of Integrative Pharmacology, Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, Illinois 60064, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shi Y, Cheng D. Beyond triglyceride synthesis: the dynamic functional roles of MGAT and DGAT enzymes in energy metabolism. Am J Physiol Endocrinol Metab 2009; 297:E10-8. [PMID: 19116371 PMCID: PMC3735925 DOI: 10.1152/ajpendo.90949.2008] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Monoacyglycerol acyltransferases (MGATs) and diacylglycerol acyltransferases (DGATs) catalyze two consecutive steps of enzyme reactions in the synthesis of triacylglycerols (TAGs). The metabolic complexity of TAG synthesis is reflected by the presence of multiple isoforms of MGAT and DGAT enzymes that differ in catalytic properties, subcellular localization, tissue distribution, and physiological functions. MGAT and DGAT enzymes play fundamental roles in the metabolism of monoacylglycerol (MAG), diacylglycerol (DAG), and triacylglycerol (TAG) that are involved in many aspects of physiological functions, such as intestinal fat absorption, lipoprotein assembly, adipose tissue formation, signal transduction, satiety, and lactation. The recent progress in the phenotypic characterization of mice deficient in MGAT and DGAT enzymes and the development of chemical inhibitors have revealed important roles of these enzymes in the regulation of energy homeostasis and insulin sensitivity. Consequently, selective inhibition of MGAT or DGAT enzymes by synthetic compounds may provide novel treatment for obesity and its related metabolic complications.
Collapse
Affiliation(s)
- Yuguang Shi
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr., Hershey, PA, USA.
| | | |
Collapse
|
28
|
Sajan MP, Standaert ML, Nimal S, Varanasi U, Pastoor T, Mastorides S, Braun U, Leitges M, Farese RV. The critical role of atypical protein kinase C in activating hepatic SREBP-1c and NFkappaB in obesity. J Lipid Res 2009; 50:1133-45. [PMID: 19202134 DOI: 10.1194/jlr.m800520-jlr200] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obesity is frequently associated with systemic insulin resistance, glucose intolerance, and hyperlipidemia. Impaired insulin action in muscle and paradoxical diet/insulin-dependent overproduction of hepatic lipids are important components of obesity, but their pathogenesis and inter-relationships between muscle and liver are uncertain. We studied two murine obesity models, moderate high-fat-feeding and heterozygous muscle-specific PKC-lambda knockout, in both of which insulin activation of atypical protein kinase C (aPKC) is impaired in muscle, but conserved in liver. In both models, activation of hepatic sterol receptor element binding protein-1c (SREBP-1c) and NFkappaB (nuclear factor-kappa B), major regulators of hepatic lipid synthesis and systemic insulin resistance, was chronically increased in the fed state. In support of a critical mediatory role of aPKC, in both models, inhibition of hepatic aPKC by adenovirally mediated expression of kinase-inactive aPKC markedly diminished diet/insulin-dependent activation of hepatic SREBP-1c and NFkappaB, and concomitantly improved hepatosteatosis, hypertriglyceridemia, hyperinsulinemia, and hyperglycemia. Moreover, in high-fat-fed mice, impaired insulin signaling to IRS-1-dependent phosphatidylinositol 3-kinase, PKB/Akt and aPKC in muscle and hyperinsulinemia were largely reversed. In obesity, conserved hepatic aPKC-dependent activation of SREBP-1c and NFkappaB contributes importantly to the development of hepatic lipogenesis, hyperlipidemia, and systemic insulin resistance. Accordingly, hepatic aPKC is a potential target for treating obesity-associated abnormalities.
Collapse
Affiliation(s)
- Mini P Sajan
- James A. Haley Veterans Hospital, Tampa, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yen CLE, Stone SJ, Koliwad S, Harris C, Farese RV. Thematic review series: glycerolipids. DGAT enzymes and triacylglycerol biosynthesis. J Lipid Res 2008; 49:2283-301. [PMID: 18757836 PMCID: PMC3837458 DOI: 10.1194/jlr.r800018-jlr200] [Citation(s) in RCA: 805] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 08/29/2008] [Indexed: 12/18/2022] Open
Abstract
Triacylglycerols (triglycerides) (TGs) are the major storage molecules of metabolic energy and FAs in most living organisms. Excessive accumulation of TGs, however, is associated with human diseases, such as obesity, diabetes mellitus, and steatohepatitis. The final and the only committed step in the biosynthesis of TGs is catalyzed by acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes. The genes encoding two DGAT enzymes, DGAT1 and DGAT2, were identified in the past decade, and the use of molecular tools, including mice deficient in either enzyme, has shed light on their functions. Although DGAT enzymes are involved in TG synthesis, they have distinct protein sequences and differ in their biochemical, cellular, and physiological functions. Both enzymes may be useful as therapeutic targets for diseases. Here we review the current knowledge of DGAT enzymes, focusing on new advances since the cloning of their genes, including possible roles in human health and diseases.
Collapse
Affiliation(s)
- Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI
53706
| | - Scot J. Stone
- Department of Biochemistry, University of Saskatchewan, Saskatoon,
Saskatchewan, Canada
| | - Suneil Koliwad
- Gladstone Institute of Cardiovascular Disease, University of California, San
Francisco, San Francisco, CA 94141
- Cardiovascular Research Institute, University of California, San
Francisco, San Francisco, CA 94141
- Department of Medicine, University of California, San Francisco,
San Francisco, CA 94141
| | - Charles Harris
- Gladstone Institute of Cardiovascular Disease, University of California, San
Francisco, San Francisco, CA 94141
- Cardiovascular Research Institute, University of California, San
Francisco, San Francisco, CA 94141
- Department of Medicine, University of California, San Francisco,
San Francisco, CA 94141
| | - Robert V. Farese
- Gladstone Institute of Cardiovascular Disease, University of California, San
Francisco, San Francisco, CA 94141
- Cardiovascular Research Institute, University of California, San
Francisco, San Francisco, CA 94141
- Department of Medicine, University of California, San Francisco,
San Francisco, CA 94141
- Department of Biochemistry and Biophysics, University of
California, San Francisco, San Francisco, CA 94141
| |
Collapse
|
30
|
Diacylglycerol acyltransferases: Potential roles as pharmacological targets. Pharmacol Ther 2008; 118:295-302. [PMID: 18508126 DOI: 10.1016/j.pharmthera.2008.03.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 03/17/2008] [Indexed: 12/29/2022]
Abstract
Triglyceride (TG) synthesis occurs in many cell-types, but only the adipocyte is specialised for TG storage. The increased incidence of obesity and its attendant pathologies have increased interest in pharmacological strategies aimed at inhibition of triglyceride synthesis. In the liver this would also appear to offer the advantages of the prevention of steatosis and/or dyslipidaemia. The two major enzymes that have DGAT activity appear to have specialised functions, that are most evident in triglyceride-secreting tissues. The presence of triglyceride in non-adipose cells can lead to (through lipolysis), or be a marker for, undesirable complications such as insulin resistance, or can be indicative of simultaneously high capacities for triglyceride synthesis, lipolysis and oxidation of fatty acids as in highly aerobic, trained muscle. Consequently, inhibition of triglyceride synthesis may not be a straightforward strategy, either in terms of its achievement pharmacologically or in its anticipated outcomes. The metabolic complexities of triglyceride synthesis, with particular reference to the diacylglycerol acyltransferases (DGATs) are considered in this short review.
Collapse
|
31
|
Timmers S, Schrauwen P, de Vogel J. Muscular diacylglycerol metabolism and insulin resistance. Physiol Behav 2007; 94:242-51. [PMID: 18207474 DOI: 10.1016/j.physbeh.2007.12.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 12/03/2007] [Accepted: 12/05/2007] [Indexed: 12/11/2022]
Abstract
Failure of insulin to elicit an increase in glucose uptake and metabolism in target tissues such as skeletal muscle is a major characteristic of non-insulin dependent type 2 diabetes mellitus. A strong correlation between intramyocellular triacylglycerol concentrations and the severity of insulin resistance has been found and led to the assumption that lipid oversupply to skeletal muscle contributes to reduced insulin action. However, the molecular mechanism that links intramyocellular lipid content with the generation of muscle insulin resistance is still unclear. It appears unlikely that the neutral lipid metabolite triacylglycerol directly impairs insulin action. Hence it is believed that intermediates in fatty acid metabolism, such as fatty acyl-CoA, ceramides or diacylglycerol (DAG) link fat deposition in the muscle to compromised insulin signaling. DAG is identified as a potential mediator of lipid-induced insulin resistance, as increased DAG levels are associated with protein kinase C activation and a reduction in both insulin-stimulated IRS-1 tyrosine phosphorylation and PI3 kinase activity. As DAG is an intermediate in the synthesis of triacylglycerol from fatty acids and glycerol, its level can be lowered by either improving the oxidation of cellular fatty acids or by accelerating the incorporation of fatty acids into triacylglycerol. This review discusses the evidence that implicates DAG being central in the development of muscular insulin resistance. Furthermore, we will discuss if and how modulation of skeletal muscle DAG levels could function as a possible therapeutic target for the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Silvie Timmers
- Department of Human Biology, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University, The Netherlands
| | | | | |
Collapse
|
32
|
Liu L, Zhang Y, Chen N, Shi X, Tsang B, Yu YH. Upregulation of myocellular DGAT1 augments triglyceride synthesis in skeletal muscle and protects against fat-induced insulin resistance. J Clin Invest 2007; 117:1679-89. [PMID: 17510710 PMCID: PMC1866250 DOI: 10.1172/jci30565] [Citation(s) in RCA: 260] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 03/12/2007] [Indexed: 01/03/2023] Open
Abstract
Increased fat deposition in skeletal muscle is associated with insulin resistance. However, exercise increases both intramyocellular fat stores and insulin sensitivity, a phenomenon referred to as "the athlete's paradox". In this study, we provide evidence that augmenting triglyceride synthesis in skeletal muscle is intrinsically connected with increased insulin sensitivity. Exercise increased diacylglycerol (DAG) acyltransferase (DGAT) activity in skeletal muscle. Channeling fatty acid substrates into TG resulted in decreased DAG and ceramide levels. Transgenic overexpression of DGAT1 in mouse skeletal muscle replicated these findings and protected mice against high-fat diet-induced insulin resistance. Moreover, in isolated muscle, DGAT1 deficiency exacerbated insulin resistance caused by fatty acids, whereas DGAT1 overexpression mitigated the detrimental effect of fatty acids. The heightened insulin sensitivity in the transgenic mice was associated with attenuated fat-induced activation of DAG-responsive PKCs and the stress mediator JNK1. Consistent with these changes, serine phosphorylation of insulin receptor substrate 1 was reduced, and Akt activation and glucose 4 membrane translocation were increased. In conclusion, upregulation of DGAT1 in skeletal muscle is sufficient to recreate the athlete's paradox and illustrates a mechanism of exercise-induced enhancement of muscle insulin sensitivity. Thus, increasing muscle DGAT activity may offer a new approach to prevent and treat insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Li Liu
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| | - Yiying Zhang
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| | - Nancy Chen
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| | - Xiaojing Shi
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| | - Bonny Tsang
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| | - Yi-Hao Yu
- Department of Medicine and
Department of Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
33
|
Wang SJY, Cornick C, O'Dowd J, Cawthorne MA, Arch JRS. Improved glucose tolerance in acyl CoA:diacylglycerol acyltransferase 1-null mice is dependent on diet. Lipids Health Dis 2007; 6:2. [PMID: 17239230 PMCID: PMC1794239 DOI: 10.1186/1476-511x-6-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 01/19/2007] [Indexed: 12/22/2022] Open
Abstract
Background Mice that lack acyl CoA:diacylglycerol acyltransferase (Dgat1-/- mice) are reported to have a reduced body fat content and improved glucose tolerance and insulin sensitivity. Studies so far have focussed on male null mice fed a high fat diet and there are few data on heterozygotes. We compared male and female Dgat1-/-, Dgat1+/- and Dgat1+/+ C57Bl/6 mice fed on either standard chow or a high fat diet. Results Body fat content was lower in the Dgat1-/- than the Dgat1+/+ mice in both experiments; lean body mass was higher in male Dgat1-/- than Dgat1+/+ mice fed on the high fat diet. Energy intake and expenditure were higher in male Dgat1-/- than Dgat1+/+ mice; these differences were less marked or absent in females. The body fat content of female Dgat1+/- mice was intermediate between that of Dgat1-/- and Dgat1+/+ mice, whereas male Dgat1+/- mice were similar to or fatter than Dgat1+/+ mice. Glucose tolerance was improved and plasma insulin reduced in Dgat1-/- mice fed on the high fat diet, but not on the chow diet. Both male and female Dgat1+/- mice had similar glucose tolerance to Dgat1+/+ mice. Conclusion These results suggest that although ablation of DGAT1 improves glucose tolerance by preventing obesity in mice fed on a high fat diet, it does not improve glucose tolerance in mice fed on a low fat diet.
Collapse
Affiliation(s)
- Steven JY Wang
- Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, UK
| | - Claire Cornick
- Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, UK
| | | | | | - Jonathan RS Arch
- Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, UK
| |
Collapse
|
34
|
Streeper RS, Koliwad SK, Villanueva CJ, Farese RV. Effects of DGAT1 deficiency on energy and glucose metabolism are independent of adiponectin. Am J Physiol Endocrinol Metab 2006; 291:E388-94. [PMID: 16595853 PMCID: PMC1552042 DOI: 10.1152/ajpendo.00621.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mice lacking acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the terminal step in triacylglycerol synthesis, have enhanced insulin sensitivity and are protected from obesity, a result of increased energy expenditure. In these mice, factors derived from white adipose tissue (WAT) contribute to the systemic changes in metabolism. One such factor, adiponectin, increases fatty acid oxidation and enhances insulin sensitivity. To test the hypothesis that adiponectin is required for the altered energy and glucose metabolism in DGAT1-deficient mice, we generated adiponectin-deficient mice and introduced adiponectin deficiency into DGAT1-deficient mice by genetic crosses. Although adiponectin-deficient mice fed a high-fat diet were heavier, exhibited worse glucose tolerance, and had more hepatic triacylglycerol accumulation than wild-type controls, mice lacking both DGAT1 and adiponectin, like DGAT1-deficient mice, were protected from diet-induced obesity, glucose intolerance, and hepatic steatosis. These findings indicate that adiponectin is required for normal energy, glucose, and lipid metabolism but that the metabolic changes induced by DGAT1-deficient WAT are independent of adiponectin and are likely due to other WAT-derived factors. Our findings also suggest that the pharmacological inhibition of DGAT1 may be useful for treating human obesity and insulin resistance associated with low circulating adiponectin levels.
Collapse
Affiliation(s)
- Ryan S. Streeper
- Gladstone Institute of Cardiovascular Disease
- Cardiovascular Research Institute and
| | - Suneil K. Koliwad
- Gladstone Institute of Cardiovascular Disease
- Cardiovascular Research Institute and
- Departments of Medicine and
| | | | - Robert V. Farese
- Gladstone Institute of Cardiovascular Disease
- Cardiovascular Research Institute and
- Departments of Medicine and
- Biochemistry and Biophysics, and
- The Diabetes Center, University of California, San Francisco, California
- Address for reprint requests and other correspondence: Robert V. Farese, Jr., Gladstone Institute of Cardiovascular Disease, 1650 Owens St., San Francisco, CA 94158 (e-mail: )
| |
Collapse
|
35
|
Abstract
The prevalence of obesity is increasing rapidly in most parts of the world and effective therapeutic drugs are urgently needed. The discovery of leptin in 1994 initiated a new understanding of adipose tissue function, and adipose tissue is now known to not only store and release fatty acids, but also to produce a wealth of factors that have an impact on the regulation of body weight and blood glucose homeostasis. Also, adipocytes express proteins that engage signalling pathways playing important roles in fuel substrate and energy metabolism. These proteins constitute a diverse array of adipose target candidates for the development of drugs to treat obesity. Some of these potential targets have been validated and are now in drug development stages, providing hope that the current obesity epidemic can be addressed by effective drug treatments in the near future.
Collapse
|
36
|
Kusunoki J, Kanatani A, Moller DE. Modulation of fatty acid metabolism as a potential approach to the treatment of obesity and the metabolic syndrome. Endocrine 2006; 29:91-100. [PMID: 16622296 DOI: 10.1385/endo:29:1:91] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2005] [Revised: 11/30/1999] [Accepted: 10/20/2005] [Indexed: 12/21/2022]
Abstract
Increased de novo lipogenesis and reduced fatty acid oxidation are probable contributors to adipose accretion in obesity. Moreover, these perturbations have a role in leading to non-alcoholic steatohepatitis, dyslipidemia, and insulin resistance--via "lipotoxicity"-related mechanisms. Research in this area has prompted an effort to evaluate several discrete enzymes in these pathways as targets for future therapeutic intervention. Acetyl-CoA carboxylase 1 (ACC1) and ACC2 regulate fatty acid synthesis and indirectly control fatty acid oxidation via a key product, malonyl CoA. Based on mouse genetic and preclinical pharmacologic evidence, inhibition of ACC1 and/or ACC2 may be a useful approach to treat obesity and metabolic syndrome. Similarly, available data suggest that inhibition of other enzymes in this pathway, including fatty acid synthase, stearoyl CoA desaturase, and diacylglycerol acytransferase 1, will have beneficial effects. AMP-activated protein kinase is a master regulator of nutrient metabolism, which controls several aspects of lipid metabolism. Activation of AMPK in selected tissues is also a potential therapeutic approach. Inhibition of hormone-sensitive lipase is another possible approach. The rationale for modulating the activity of these enzymes and their relative merits (and downsides) as possible therapeutic targets are further discussed.
Collapse
Affiliation(s)
- Jun Kusunoki
- Department of Metabolic Disorders, Banyu-Tsukuba Research Institute, Tsukuba, Japan
| | | | | |
Collapse
|
37
|
Chen HC. Enhancing energy and glucose metabolism by disrupting triglyceride synthesis: Lessons from mice lacking DGAT1. Nutr Metab (Lond) 2006; 3:10. [PMID: 16448557 PMCID: PMC1382234 DOI: 10.1186/1743-7075-3-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Accepted: 01/31/2006] [Indexed: 01/09/2023] Open
Abstract
Although the ability to make triglycerides is essential for normal physiology, excess accumulation of triglycerides results in obesity and is associated with insulin resistance. Inhibition of triglyceride synthesis, therefore, may represent a feasible strategy for the treatment of obesity and type 2 diabetes. Acyl CoA:diacylglycerol acyltransferase 1 (DGAT1) is one of two DGAT enzymes that catalyze the final reaction in the known pathways of mammalian triglyceride synthesis. Mice lacking DGAT1 have increased energy expenditure and insulin sensitivity and are protected against diet-induced obesity and glucose intolerance. These metabolic effects of DGAT1 deficiency result in part from the altered secretion of adipocyte-derived factors. Studies of DGAT1-deficient mice have helped to provide insights into the mechanisms by which cellular lipid metabolism modulates systemic carbohydrate and insulin metabolism, and a better understanding of how DGAT1 deficiency enhances energy expenditure and insulin sensitivity may identify additional targets or strategies for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Hubert C Chen
- Department of Medicine, University of California, San Francisco, CA 94103, USA.
| |
Collapse
|
38
|
Farese RV, Sajan MP, Standaert ML. Atypical protein kinase C in insulin action and insulin resistance. Biochem Soc Trans 2005; 33:350-3. [PMID: 15787604 DOI: 10.1042/bst0330350] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It now seems clear that aPKC (atypical protein kinase C) isoforms are required for insulin-stimulated glucose transport in muscle and adipocytes. Moreover, there are marked defects in the activation of aPKCs under a variety of insulin-resistant conditions in humans, monkeys and rodents. In humans, defects in aPKC in muscle are seen in Type II diabetes and its precursors, obesity, the obesity-associated polycystic ovary syndrome and impaired glucose tolerance. These defects in muscle aPKC activation are due to both impaired activation of insulin receptor substrate-1-dependent PI3K (phosphoinositide 3-kinase) and the direct activation of aPKCs by the lipid product of PI3K, PI-3,4,5-(PO4)3. Although it is still uncertain which underlying defect comes first, the resultant defect in aPKC activation in muscle most certainly contributes significantly to the development of skeletal muscle insulin resistance. Of further note, unlike the seemingly ubiquitous presence of defective aPKC activation in skeletal muscle in insulin-resistant states, the activation of aPKC is normal or increased in livers of Type II diabetic and obese rodents. The maintenance of aPKC activation in the liver may explain how insulin-dependent lipid synthesis is maintained in these states, as aPKCs function mainly in the activation of enzymes important for lipid synthesis. Thus increased activation of liver aPKC in hyperinsulinaemic states may contribute significantly to the development of hyperlipidaemia in insulin-resistant states.
Collapse
Affiliation(s)
- R V Farese
- Research Service, James A. Haley Veterans Administration Medical Center, and Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
39
|
Chen HC, Farese RV. Inhibition of triglyceride synthesis as a treatment strategy for obesity: lessons from DGAT1-deficient mice. Arterioscler Thromb Vasc Biol 2004; 25:482-6. [PMID: 15569818 DOI: 10.1161/01.atv.0000151874.81059.ad] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Because the ability to make triglycerides is essential for the accumulation of adipose tissue, inhibition of triglyceride synthesis may ameliorate obesity and its related medical consequences. Acyl coenzyme A (CoA):diacylglycerol acyltransferase 1 (DGAT1) is 1 of 2 DGAT enzymes that catalyze the final reaction in the known pathways of mammalian triglyceride synthesis. Mice lacking DGAT1 are resistant to obesity and have increased sensitivity to insulin and leptin. DGAT1-deficient mice are also resistant to diet-induced hepatic steatosis. The effects of DGAT1 deficiency on energy and glucose metabolism result in part from the altered secretion of adipocyte-derived factors. Although complete DGAT1 deficiency causes alopecia and impairs development of the mammary gland, these abnormalities are not observed in mice with partial DGAT1 deficiency. These findings suggest that pharmacological inhibition of DGAT1 may be a feasible therapeutic strategy for human obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Hubert C Chen
- Department of Medical Sciences, Amgen Inc, Thousand Oaks, USA
| | | |
Collapse
|
40
|
Suzuki R, Tobe K, Aoyama M, Sakamoto K, Ohsugi M, Kamei N, Nemoto S, Inoue A, Ito Y, Uchida S, Hara K, Yamauchi T, Kubota N, Terauchi Y, Kadowaki T. Expression of DGAT2 in white adipose tissue is regulated by central leptin action. J Biol Chem 2004; 280:3331-7. [PMID: 15550388 DOI: 10.1074/jbc.m410955200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes catalyze the final step in mammalian triglyceride synthesis, and their functions are considered to be involved in the mechanisms of obesity, insulin resistance, and leptin resistance. Insulin receptor substrate-2 (IRS-2)-deficient mice exhibit obesity-associated with hypertrophic adipocytes and leptin resistance. Screening for transcripts of genes involved in fatty acid and triglyceride synthesis to investigate the mechanism of the hypertrophic change in the adipocytes showed that expression of DGAT2 mRNA was up-regulated in the white adipose tissue (WAT) of Irs2-/- mice, whereas that of DGAT1 was down-regulated. This reciprocal expression of DGAT1 and DGAT2 was also observed in WAT of leptin-deficient ob/ob mice. A high fat diet also resulted in increased DGAT2 and reduced DGAT1 in the WAT of C57BL/6 mice. Induction of adipocyte hypertrophy in vitro up-regulated both DGAT1 and DGAT2 expression in 3T3-L1 cells, suggesting that adipocyte non-autonomous mechanism in vivo is required for the reciprocal changes in expression of DGAT1 and DGAT2. In fact, intracerebroventricular infusion of leptin reduced DGAT2 expression in WAT of Irs2-/- mice and ob/ob mice, independently of DGAT1 expression. We propose the hypothesis that leptin regulates adipocyte size by altering expression patterns of DGAT via central nervous system to determine the levels of triglyceride synthesis.
Collapse
Affiliation(s)
- Ryo Suzuki
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|