Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 10(8): 1128-1133, 2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31413796

ABSTRACT

Diacylglycerol O-acyltransferase 1 (DGAT1) inhibitor Pradigastat (1) was shown to be effective at decreasing postprandial triglyceride levels in a patient population with familial chylomicronemia syndrome (FCS). Although pradigastat does not cause photosensitization in humans at the high clinical dose of 40 mg, a positive signal was observed in preclinical models of phototoxicity. Herein, we describe a preclinical phototoxicity mitigation strategy for diarylamine containing molecules utilizing the introduction of an amide or suitable heterocyclic function. This strategy led to the development of two second-generation compounds with low risk of phototoxicity, disparate exposure profiles, and comparable efficacy to 1 in a rodent lipid bolus model for post-prandial plasma triglycerides.

2.
Bioorg Med Chem Lett ; 26(4): 1245-8, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26804232

ABSTRACT

Diamide compounds were identified as potent DGAT1 inhibitors in vitro, but their poor molecular properties resulted in low oral bioavailability, both systemically and to DGAT1 in the enterocytes of the small intestine, resulting in a lack of efficacy in vivo. Replacing an N-alkyl group on the diamide with an N-aryl group was found to be an effective strategy to confer oral bioavailability and oral efficacy in this lipophilic diamide class of inhibitors.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diamide/chemistry , Enzyme Inhibitors/chemistry , Animals , Cell Line, Tumor , Diacylglycerol O-Acyltransferase/metabolism , Diamide/chemical synthesis , Diamide/pharmacokinetics , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Half-Life , Humans , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
Aging (Albany NY) ; 4(1): 13-27, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22291164

ABSTRACT

Calorie restriction results in leanness, which is linked to metabolic conditions that favor longevity. We show here that deficiency of the triglyceride synthesis enzyme acyl CoA:diacylglycerol acyltransferase 1 (DGAT1), which promotes leanness, also extends longevity without limiting food intake. Female DGAT1-deficient mice were protected from age-related increases in body fat, tissue triglycerides, and inflammation in white adipose tissue. This protection was accompanied by increased mean and maximal life spans of ~25% and ~10%, respectively. Middle-agedDgat1-/- mice exhibited several features associated with longevity, including decreased levels of circulating insulin growth factor 1 (IGF1) and reduced fecundity. Thus, deletion of DGAT1 in mice provides a model of leanness and extended lifespan that is independent of calorie restriction.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Longevity/genetics , Longevity/physiology , Adipose Tissue/physiology , Aging/metabolism , Animals , Body Composition , Bone Density/genetics , Bone Density/physiology , Caloric Restriction , Diacylglycerol O-Acyltransferase/deficiency , Diacylglycerol O-Acyltransferase/genetics , Energy Metabolism/genetics , Energy Metabolism/physiology , Female , Fertility , Gene Expression Profiling , Gene Expression Regulation , Genotype , Litter Size , Mice , Mice, Knockout , Thinness/enzymology , Thinness/metabolism
4.
J Lipid Res ; 52(4): 657-67, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21317108

ABSTRACT

The total contribution of the acyl CoA:diacylglycerol acyltransferase (DGAT) enzymes, DGAT1 and DGAT2, to mammalian triacylglycerol (TG) synthesis has not been determined. Similarly, whether DGAT enzymes are required for lipid droplet (LD) formation is unknown. In this study, we examined the requirement for DGAT enzymes in TG synthesis and LDs in differentiated adipocytes with genetic deletions of DGAT1 and DGAT2. Adipocytes with a single deletion of either enzyme were capable of TG synthesis and LD formation. In contrast, adipocytes with deletions of both DGATs were severely lacking in TG and did not have LDs, indicating that DGAT1 and DGAT2 account for nearly all TG synthesis in adipocytes and appear to be required for LD formation during adipogenesis. DGAT enzymes were not absolutely required for LD formation in mammalian cells, however; macrophages deficient in both DGAT enzymes were able to form LDs when incubated with cholesterol-rich lipoproteins. Although adipocytes lacking both DGATs had no TG or LDs, they were fully differentiated by multiple criteria. Our findings show that DGAT1 and DGAT2 account for the vast majority of TG synthesis in mice, and DGAT function is required for LDs in adipocytes, but not in all cell types.


Subject(s)
Adipocytes/enzymology , Adipocytes/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Triglycerides/biosynthesis , Animals , Blotting, Western , Diacylglycerol O-Acyltransferase/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Macrophages/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Triglycerides/genetics
5.
J Biol Chem ; 285(42): 31995-2002, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20685656

ABSTRACT

SIRT4, a member of the sirtuin family, has been implicated in the regulation of insulin secretion by modulation of glutamate dehydrogenase. However, the role of this enzyme in the regulation of metabolism in other tissues is unknown. In this study we investigated whether depletion of SIRT4 would enhance liver and muscle metabolic functions. To do this SIRT4 was knocked down using an adenoviral shRNA in mouse primary hepatocytes and myotubes. We observed a significant increase in gene expression of mitochondrial and fatty acid metabolism enzymes in hepatocytes with reduced SIRT4 levels. SIRT4 knockdown also increased SIRT1 mRNA and protein levels both in vitro and in vivo. In agreement with the increased fatty acid oxidation (FAO) gene expression, we showed a significant increase in FAO in SIRT4 knockdown primary hepatocytes compared with control, and this effect was dependent on SIRT1. In primary myotubes, knockdown of SIRT4 resulted in increased FAO, cellular respiration, and pAMPK levels. When SIRT4 was knocked down in vivo by tail vein injection of a shRNA adenovirus, we observed a significant increase in hepatic mitochondrial and FAO gene expression consistent with the findings in primary hepatocytes. Taken together these findings demonstrate that SIRT4 inhibition increases fat oxidative capacity in liver and mitochondrial function in muscle, which might provide therapeutic benefits for diseases associated with ectopic lipid storage such as type 2 diabetes.


Subject(s)
Fatty Acids/metabolism , Genes, Mitochondrial , Hepatocytes/physiology , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/physiology , Myoblasts/physiology , Sirtuins/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Gene Knockdown Techniques , Hepatocytes/cytology , Mice , Mitochondrial Proteins/genetics , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Oxidation-Reduction , Oxygen Consumption , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuins/genetics
6.
J Clin Invest ; 120(3): 756-67, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124729

ABSTRACT

Diet-induced obesity (DIO) leads to inflammatory activation of macrophages in white adipose tissue (WAT) and subsequently to insulin resistance. PPARgamma agonists are antidiabetic agents known to suppress inflammatory macrophage activation and to induce expression of the triacylglycerol (TG) synthesis enzyme acyl CoA: diacylglycerol acyltransferase 1 (DGAT1) in WAT and in adipocytes. Here, we investigated in mice the relationship between macrophage lipid storage capacity and DIO-associated inflammatory macrophage activation. Mice overexpressing DGAT1 in both macrophages and adipocytes (referred to herein as aP2-Dgat1 mice) were more prone to DIO but were protected against inflammatory macrophage activation, macrophage accumulation in WAT, systemic inflammation, and insulin resistance. To assess the contribution of macrophage DGAT1 expression to this phenotype, we transplanted wild-type mice with aP2-Dgat1 BM. These mice developed DIO similar to that of control mice but retained the protection from WAT inflammation and insulin resistance seen in aP2-Dgat1 mice. In isolated macrophages, Dgat1 mRNA levels correlated directly with TG storage capacity and inversely with inflammatory activation by saturated fatty acids (FAs). Moreover, PPARgamma agonists increased macrophage Dgat1 mRNA levels, and the protective effects of these agonists against FA-induced inflammatory macrophage activation were absent in macrophages isolated from Dgat1-null mice. Thus, increasing DGAT1 expression in murine macrophages increases their capacity for TG storage, protects against FA-induced inflammatory activation, and is sufficient to reduce the inflammatory and metabolic consequences of DIO.


Subject(s)
Diacylglycerol O-Acyltransferase/biosynthesis , Diet/adverse effects , Insulin Resistance , Macrophages/enzymology , Obesity/enzymology , Triglycerides/metabolism , Adipocytes/enzymology , Adipose Tissue, White/enzymology , Animals , Diacylglycerol O-Acyltransferase/genetics , Fatty Acids , Gene Expression Regulation, Enzymologic/genetics , Inflammation/genetics , Inflammation/mortality , Mice , Mice, Knockout , Obesity/etiology , Obesity/genetics , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Triglycerides/genetics
7.
J Biol Chem ; 284(7): 4292-9, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19028692

ABSTRACT

Retinoic acid (RA) is a potent signaling molecule that is essential for many biological processes, and its levels are tightly regulated by mechanisms that are only partially understood. The synthesis of RA from its precursor retinol (vitamin A) is an important regulatory mechanism. Therefore, the esterification of retinol with fatty acyl moieties to generate retinyl esters, the main storage form of retinol, may also regulate RA levels. Here we show that the neutral lipid synthesis enzyme acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) functions as the major acyl-CoA:retinol acyltransferase (ARAT) in murine skin. When dietary retinol is abundant, DGAT1 deficiency results in elevated levels of RA in skin and cyclical hair loss; both are prevented by dietary retinol deprivation. Further, DGAT1-deficient skin exhibits enhanced sensitivity to topically administered retinol. Deletion of the enzyme specifically in the epidermis causes alopecia, indicating that the regulation of RA homeostasis by DGAT1 is autonomous in the epidermis. These findings show that DGAT1 functions as an ARAT in the skin, where it acts to maintain retinoid homeostasis and prevent retinoid toxicity. Our findings may have implications for human skin or hair disorders treated with agents that modulate RA signaling.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Epidermis/enzymology , Homeostasis/physiology , Retinol O-Fatty-Acyltransferase/metabolism , Tretinoin/metabolism , Alopecia/enzymology , Alopecia/genetics , Animals , Diacylglycerol O-Acyltransferase/genetics , Female , Homeostasis/drug effects , Male , Mice , Mice, Knockout , Retinoids/genetics , Retinoids/metabolism , Retinol O-Fatty-Acyltransferase/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tretinoin/pharmacology
8.
Mol Cell Biol ; 27(24): 8807-14, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923681

ABSTRACT

Homologs of the Saccharomyces cerevisiae Sir2 protein, sirtuins, promote longevity in many organisms. Studies of the sirtuin SIRT3 have so far been limited to cell culture systems. Here, we investigate the localization and function of SIRT3 in vivo. We show that endogenous mouse SIRT3 is a soluble mitochondrial protein. To address the function and relevance of SIRT3 in the regulation of energy metabolism, we generated and phenotypically characterized SIRT3 knockout mice. SIRT3-deficient animals exhibit striking mitochondrial protein hyperacetylation, suggesting that SIRT3 is a major mitochondrial deacetylase. In contrast, no mitochondrial hyperacetylation was detectable in mice lacking the two other mitochondrial sirtuins, SIRT4 and SIRT5. Surprisingly, despite this biochemical phenotype, SIRT3-deficient mice are metabolically unremarkable under basal conditions and show normal adaptive thermogenesis, a process previously suggested to involve SIRT3. Overall, our results extend the recent finding of lysine acetylation of mitochondrial proteins and demonstrate that SIRT3 has evolved to control reversible lysine acetylation in this organelle.


Subject(s)
Histone Deacetylases/metabolism , Lysine/metabolism , Mammals/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Sequence Homology, Amino Acid , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuins/metabolism , Acetylation , Adipose Tissue, Brown/cytology , Animals , Feeding Behavior , Food Deprivation , Gene Targeting , Glutamate Dehydrogenase/metabolism , Mice , Mitochondria, Liver/enzymology , Mitochondrial Proteins/deficiency , Sirtuin 2 , Sirtuin 3 , Sirtuins/deficiency , Solubility , Thermogenesis
9.
Proc Natl Acad Sci U S A ; 103(42): 15675-80, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-17030820

ABSTRACT

A pluripotent cell line, C3H10T1/2, is induced to undergo adipogenesis by a mixture of factors that includes a glucocorticoid such as dexamethasone. We found that expression of myostatin (MSTN), a TGF-beta family member extensively studied in muscle, was induced by dexamethasone under those differentiation conditions. Moreover, MSTN could substitute for dexamethasone in the adipogenesis mixture. However, the adipocytes induced by MSTN in both cell culture and transgenic mice were small and expressed markers characteristic of immature adipocytes. These adipocytes exhibited cell-autonomous increases in insulin sensitivity and glucose oxidation. In mice, these effects produced elevated systemic insulin sensitivity and resistance to diet-induced obesity. Modulation of the final stages of adipogenesis may provide a novel approach to understanding and treating metabolic disease.


Subject(s)
Adipocytes/metabolism , Transforming Growth Factor beta/metabolism , Adipocytes/cytology , Adipogenesis/physiology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adipose Tissue/physiology , Animals , Cell Differentiation/physiology , Cell Line , Dexamethasone/metabolism , Diet , Glucose/metabolism , Insulin/metabolism , Male , Mice , Mice, Transgenic , Myostatin , Obesity/metabolism , Oxidation-Reduction
10.
Am J Physiol Endocrinol Metab ; 291(2): E388-94, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16595853

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.


Subject(s)
Adiponectin/metabolism , Adipose Tissue/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Eating/physiology , Energy Metabolism/physiology , Glucose/metabolism , Oxygen Consumption/physiology , Animals , Mice , Mice, Knockout
11.
J Biol Chem ; 278(14): 11782-93, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12556524

ABSTRACT

Glucose-6-phosphatase catalyzes the terminal step in the gluconeogenic and glycogenolytic pathways. In HepG2 cells, the maximum repression of basal glucose-6-phosphatase catalytic subunit (G6Pase) gene transcription by insulin requires two distinct promoter regions, designated A (located between -231 and -199) and B (located between -198 and -159), that together form an insulin response unit. Region A binds hepatocyte nuclear factor-1, which acts as an accessory factor to enhance the effect of insulin, mediated through region B, on G6Pase gene transcription. We have previously shown that region B binds the transcriptional activator FKHR (FOXO1a) in vitro. Chromatin immunoprecipitation assays demonstrate that FKHR also binds the G6Pase promoter in situ and that insulin inhibits this binding. Region B contains three insulin response sequences (IRSs), designated IRS 1, 2, and 3, that share the core sequence T(G/A)TTTT. However, detailed analyses reveal that these three G6Pase IRSs are functionally distinct. Thus, FKHR binds IRS 1 with high affinity and IRS 2 with low affinity but it does not bind IRS 3. Moreover, in the context of the G6Pase promoter, IRS 1 and 2, but not IRS 3, are required for the insulin response. Surprisingly, IRS 3, as well as IRS 1 and IRS 2, can each confer an inhibitory effect of insulin on the expression of a heterologous fusion gene, indicating that, in this context, a transcription factor other than FKHR, or its orthologs, can also mediate an insulin response through the T(G/A)TTTT motif.


Subject(s)
Catalytic Domain/genetics , Glucose-6-Phosphatase/genetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Nerve Tissue Proteins , Promoter Regions, Genetic/genetics , Animals , Base Sequence , Carcinoma, Hepatocellular , Chloramphenicol O-Acetyltransferase/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Mice , Rats , Recombinant Fusion Proteins/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tumor Cells, Cultured
12.
J Biol Chem ; 277(31): 27935-44, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12032154

ABSTRACT

Insulin stimulates malic enzyme (ME)-chloramphenicol acetyltransferase (CAT) and collagenase-1-CAT fusion gene expression in H4IIE cells through identical activator protein-1 (AP-1) motifs. In contrast, insulin and phorbol esters only stimulate collagenase-1-CAT and not ME-CAT fusion gene expression in HeLa cells. The experiments in this article were designed to explore the molecular basis for this differential cell type- and gene-specific regulation. The results highlight the influence of three variables, namely promoter context, AP-1 flanking sequence, and accessory elements that modulate insulin and phorbol ester signaling through the AP-1 motif. Thus, fusion gene transfection and proteolytic clipping gel retardation assays suggest that the AP-1 flanking sequence affects the conformation of AP-1 binding to the collagenase-1 and ME AP-1 motifs such that it selectively binds the latter in a fully activated state. However, this influence of ME AP-1 flanking sequence is dependent on promoter context. Thus, the ME AP-1 motif will mediate both an insulin and phorbol ester response in HeLa cells when introduced into either the collagenase-1 promoter or a specific heterologous promoter. But even in the context of the collagenase-1 promoter, the effects of both insulin and phorbol esters, mediated through the ME AP-1 motif are dependent on accessory factors.


Subject(s)
Collagenases/genetics , Insulin/physiology , Malate Dehydrogenase/genetics , Phorbol Esters/pharmacology , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , DNA Primers , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Humans , Mutagenesis , Oligodeoxyribonucleotides , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Transfection , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...