Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Arterioscler Thromb Vasc Biol ; 39(10): 1967-1985, 2019 10.
Article in English | MEDLINE | ID: mdl-31434492

ABSTRACT

OBJECTIVE: Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS: The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.


Subject(s)
Atherosclerosis/metabolism , Hyperlipoproteinemia Type I/metabolism , Lipoprotein Lipase/metabolism , Macrophage Activation/genetics , Animals , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Humans , Hyperlipoproteinemia Type I/pathology , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Role , Sensitivity and Specificity , Triglycerides/metabolism
2.
Arterioscler Thromb Vasc Biol ; 38(9): 2207-2216, 2018 09.
Article in English | MEDLINE | ID: mdl-30354257

ABSTRACT

Objective- SGLT2 (sodium-glucose cotransporter 2) inhibition in humans leads to increased levels of LDL (low-density lipoprotein) cholesterol and decreased levels of plasma triglyceride. Recent studies, however, have shown this therapy to lower cardiovascular mortality. In this study, we aimed to determine how SGLT2 inhibition alters circulating lipoproteins. Approach and Results- We used a mouse model expressing human CETP (cholesteryl ester transfer protein) and human ApoB100 (apolipoprotein B100) to determine how SGLT2 inhibition alters plasma lipoprotein metabolism. The mice were fed a high-fat diet and then were made partially insulin deficient using streptozotocin. SGLT2 was inhibited using a specific antisense oligonucleotide or canagliflozin, a clinically available oral SGLT2 inhibitor. Inhibition of SGLT2 increased circulating levels of LDL cholesterol and reduced plasma triglyceride levels. SGLT2 inhibition was associated with increased LpL (lipoprotein lipase) activity in the postheparin plasma, decreased postprandial lipemia, and faster clearance of radiolabeled VLDL (very-LDL) from circulation. Additionally, SGLT2 inhibition delayed turnover of labeled LDL from circulation. Conclusions- Our studies in diabetic CETP-ApoB100 transgenic mice recapitulate many of the changes in circulating lipids found with SGLT2 inhibition therapy in humans and suggest that the increased LDL cholesterol found with this therapy is because of reduced clearance of LDL from the circulation and greater lipolysis of triglyceride-rich lipoproteins. Most prominent effects of SGLT2 inhibition in the current mouse model were seen with antisense oligonucleotides-mediated knockdown of SGLT2.


Subject(s)
Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Lipoproteins, LDL/blood , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Triglycerides/blood , Adipose Tissue/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Blood Glucose/metabolism , Down-Regulation , Fatty Acids, Nonesterified/blood , Gene Expression , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Myocardium/metabolism , RNA, Messenger/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
3.
J Lipid Res ; 58(6): 1132-1142, 2017 06.
Article in English | MEDLINE | ID: mdl-28404638

ABSTRACT

Lipid accumulation is a pathological feature of every type of kidney injury. Despite this striking histological feature, physiological accumulation of lipids in the kidney is poorly understood. We studied whether the accumulation of lipids in the fasted kidney are derived from lipoproteins or NEFAs. With overnight fasting, kidneys accumulated triglyceride, but had reduced levels of ceramide and glycosphingolipid species. Fasting led to a nearly 5-fold increase in kidney uptake of plasma [14C]oleic acid. Increasing circulating NEFAs using a ß adrenergic receptor agonist caused a 15-fold greater accumulation of lipid in the kidney, while mice with reduced NEFAs due to adipose tissue deficiency of adipose triglyceride lipase had reduced triglycerides. Cluster of differentiation (Cd)36 mRNA increased 2-fold, and angiopoietin-like 4 (Angptl4), an LPL inhibitor, increased 10-fold. Fasting-induced kidney lipid accumulation was not affected by inhibition of LPL with poloxamer 407 or by use of mice with induced genetic LPL deletion. Despite the increase in CD36 expression with fasting, genetic loss of CD36 did not alter fatty acid uptake or triglyceride accumulation. Our data demonstrate that fasting-induced triglyceride accumulation in the kidney correlates with the plasma concentrations of NEFAs, but is not due to uptake of lipoprotein lipids and does not involve the fatty acid transporter, CD36.


Subject(s)
Fasting/blood , Fasting/metabolism , Fatty Acids, Nonesterified/blood , Kidney/metabolism , Triglycerides/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction
4.
Arterioscler Thromb Vasc Biol ; 35(1): 102-10, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395613

ABSTRACT

OBJECTIVE: Diabetic hypertriglyceridemia is thought to be primarily driven by increased hepatic de novo lipogenesis. However, experiments in animal models indicated that insulin deficiency should decrease hepatic de novo lipogenesis and reduce plasma triglyceride levels. APPROACH AND RESULTS: To address the discrepancy between human data and genetically altered mouse models, we investigated whether insulin-deficient diabetic mice had triglyceride changes that resemble those in diabetic humans. Streptozotocin-induced insulin deficiency increased plasma triglyceride levels in mice. Contrary to the mouse models with impaired hepatic insulin receptor signaling, insulin deficiency did not reduce hepatic triglyceride secretion and de novo lipogenesis-related gene expression. Diabetic mice had a marked decrease in postprandial triglycerides clearance, which was associated with decreased lipoprotein lipase and peroxisome proliferator-activated receptor α mRNA levels in peripheral tissues and decreased lipoprotein lipase activity in skeletal muscle, heart, and brown adipose tissue. Diabetic heterozygous lipoprotein lipase knockout mice had markedly elevated fasting plasma triglyceride levels and prolonged postprandial triglycerides clearance. CONCLUSIONS: Insulin deficiency causes hypertriglyceridemia by decreasing peripheral lipolysis and not by an increase in hepatic triglycerides production and secretion.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hypertriglyceridemia/metabolism , Insulin/blood , Lipolysis , Liver/metabolism , Streptozocin , Triglycerides/blood , Adipose Tissue, Brown/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Hypertriglyceridemia/blood , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/genetics , Lipogenesis , Lipoprotein Lipase/deficiency , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Myocardium/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR delta/genetics , PPAR delta/metabolism , Postprandial Period , RNA, Messenger/metabolism , Signal Transduction , Time Factors
6.
Mol Metab ; 66: 101596, 2022 12.
Article in English | MEDLINE | ID: mdl-36100179

ABSTRACT

OBJECTIVE: Metabolomics as an approach to solve biological problems is exponentially increasing in use. Thus, this a pivotal time for the adoption of best practices. It is well known that disrupted tissue oxygen supply rapidly alters cellular energy charge. However, the speed and extent to which delayed mouse tissue freezing after dissection alters the broad metabolome is not well described. Furthermore, how tissue genotype may modulate such metabolomic drift and the degree to which traced 13C-isotopologue distributions may change have not been addressed. METHODS: By combined liquid chromatography (LC)- and gas chromatography (GC)-mass spectrometry (MS), we measured how levels of 255 mouse liver metabolites changed following 30-second, 1-minute, 3-minute, and 10-minute freezing delays. We then performed test-of-concept delay-to-freeze experiments evaluating broad metabolomic drift in mouse heart and skeletal muscle, differential metabolomic change between wildtype (WT) and mitochondrial pyruvate carrier (MPC) knockout mouse livers, and shifts in 13C-isotopologue abundances and enrichments traced from 13C-labled glucose into mouse liver. RESULTS: Our data demonstrate that delayed mouse tissue freezing after dissection leads to rapid hypoxia-driven remodeling of the broad metabolome, induction of both false-negative and false-positive between-genotype differences, and restructuring of 13C-isotopologue distributions. Notably, we show that increased purine nucleotide degradation products are an especially high dynamic range marker of delayed liver and heart freezing. CONCLUSIONS: Our findings provide a previously absent, systematic illustration of the extensive, multi-domain metabolomic changes occurring within the early minutes of delayed tissue freezing. They also provide a novel, detailed resource of mouse liver ex vivo, hypoxic metabolomic remodeling.


Subject(s)
Metabolome , Metabolomics , Animals , Mice , Metabolome/physiology , Metabolomics/methods , Hypoxia , Mice, Knockout , Genotype
7.
Antioxidants (Basel) ; 11(7)2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35883787

ABSTRACT

Xanthine oxidase (XO) contributes to oxidative stress and vascular disease. Hyperuricemia and gout are common in patients with chronic kidney disease (CKD), a population at increased risk of vascular disease. We evaluated effects of allopurinol on serum XO activity and metabolome of CKD patients who had participated in a randomized double-blind clinical trial of allopurinol vs. placebo. XO activity was measured in participants' serum. XO expression in venous endothelial cells was evaluated via immunofluorescence. Gas chromatography mass spectrometry (GC/MS) was utilized for metabolomics analysis. We found that in patients with stage 3 CKD and hyperuricemia, allopurinol lowered serum urate while increasing serum xanthine levels. Allopurinol, however, did not significantly suppress measured serum XO activity. Of note, baseline serum XO activity was low. Additionally, neither baseline serum XO activity nor XO protein expression were associated with measures of vascular dysfunction or with systemic or endothelial biomarkers of oxidative stress. Allopurinol affected several pathways, including pentose phosphate, pyrimidine, and tyrosine metabolism. Our findings suggest that circulating XO does not contribute to vascular disease in CKD patients. In addition to inhibition of XO activity, allopurinol was observed to impact other pathways; the implications of which require further study.

8.
JCI Insight ; 52019 08 08.
Article in English | MEDLINE | ID: mdl-31393858

ABSTRACT

Dual peroxisome proliferator-activated receptor (PPAR)α/γ agonists that were developed to target hyperlipidemia and hyperglycemia in type 2 diabetes patients, caused cardiac dysfunction or other adverse effects. We studied the mechanisms that underlie the cardiotoxic effects of a dual PPARα/γ agonist, tesaglitazar, in wild type and diabetic (leptin receptor deficient - db/db) mice. Mice treated with tesaglitazar-containing chow or high fat diet developed cardiac dysfunction despite lower plasma triglycerides and glucose levels. Expression of cardiac peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), which promotes mitochondrial biogenesis, had the most profound reduction among various fatty acid metabolism genes. Furthermore, we observed increased acetylation of PGC1α, which suggests PGC1α inhibition and lowered sirtuin 1 (SIRT1) expression. This change was associated with lower mitochondrial abundance. Combined pharmacological activation of PPARα and PPARγ in C57BL/6 mice reproduced the reduction of PGC1α expression and mitochondrial abundance. Resveratrol-mediated SIRT1 activation attenuated tesaglitazar-induced cardiac dysfunction and corrected myocardial mitochondrial respiration in C57BL/6 and diabetic mice but not in cardiomyocyte-specific Sirt1-/- mice. Our data shows that drugs, which activate both PPARα and PPARγ lead to cardiac dysfunction associated with PGC1α suppression and lower mitochondrial abundance likely due to competition between these two transcription factors.


Subject(s)
Heart Failure/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisomes/metabolism , Sirtuin 1/metabolism , Alkanesulfonates/adverse effects , Animals , Blood Glucose , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , PPAR alpha/agonists , PPAR gamma/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenylpropionates/adverse effects , Receptors, Leptin/metabolism , Sirtuin 1/genetics , Transcription Factors , Transcriptome
9.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2125-2137, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31029826

ABSTRACT

Cardiac metabolism affects systemic energetic balance. Previously, we showed that Krüppel-like factor (KLF)-5 regulates cardiomyocyte PPARα and fatty acid oxidation-related gene expression in diabetes. We surprisingly found that cardiomyocyte-specific KLF5 knockout mice (αMHC-KLF5-/-) have accelerated diet-induced obesity, associated with increased white adipose tissue (WAT). Alterations in cardiac expression of the mediator complex subunit 13 (Med13) modulates obesity. αMHC-KLF5-/- mice had reduced cardiac Med13 expression likely because KLF5 upregulates Med13 expression in cardiomyocytes. We then investigated potential mechanisms that mediate cross-talk between cardiomyocytes and WAT. High fat diet-fed αMHC-KLF5-/- mice had increased levels of cardiac and plasma FGF21, while food intake, activity, plasma leptin, and natriuretic peptides expression were unchanged. Consistent with studies reporting that FGF21 signaling in WAT decreases sumoylation-driven PPARγ inactivation, αMHC-KLF5-/- mice had less SUMO-PPARγ in WAT. Increased diet-induced obesity found in αMHC-KLF5-/- mice was absent in αMHC-[KLF5-/-;FGF21-/-] double knockout mice, as well as in αMHC-FGF21-/- mice that we generated. Thus, cardiomyocyte-derived FGF21 is a component of pro-adipogenic crosstalk between heart and WAT.


Subject(s)
Fibroblast Growth Factors/metabolism , Kruppel-Like Transcription Factors/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Weight , Diet, High-Fat , Female , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Leptin/blood , Male , Mediator Complex/genetics , Mediator Complex/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Obesity/etiology , Signal Transduction
10.
Cell Rep ; 28(10): 2608-2619.e6, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31484072

ABSTRACT

Hepatocellular carcinoma (HCC) is a devastating cancer increasingly caused by non-alcoholic fatty liver disease (NAFLD). Disrupting the liver Mitochondrial Pyruvate Carrier (MPC) in mice attenuates NAFLD. Thus, we considered whether liver MPC disruption also prevents HCC. Here, we use the N-nitrosodiethylamine plus carbon tetrachloride model of HCC development to test how liver-specific MPC knock out affects hepatocellular tumorigenesis. Our data show that liver MPC ablation markedly decreases tumorigenesis and that MPC-deficient tumors transcriptomically downregulate glutathione metabolism. We observe that MPC disruption and glutathione depletion in cultured hepatomas are synthetically lethal. Stable isotope tracing shows that hepatocyte MPC disruption reroutes glutamine from glutathione synthesis into the tricarboxylic acid (TCA) cycle. These results support a model where inducing metabolic competition for glutamine by MPC disruption impairs hepatocellular tumorigenesis by limiting glutathione synthesis. These findings raise the possibility that combining MPC disruption and glutathione stress may be therapeutically useful in HCC and additional cancers.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Citric Acid Cycle , Glutamine/metabolism , Glutathione/biosynthesis , Liver Neoplasms/metabolism , Mitochondria/metabolism , Pyruvic Acid/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Hepatocytes/metabolism , Humans , Liver Neoplasms/genetics , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Organ Specificity , Transcriptome/genetics
11.
Elife ; 82019 07 18.
Article in English | MEDLINE | ID: mdl-31305240

ABSTRACT

Metabolic cycles are a fundamental element of cellular and organismal function. Among the most critical in higher organisms is the Cori Cycle, the systemic cycling between lactate and glucose. Here, skeletal muscle-specific Mitochondrial Pyruvate Carrier (MPC) deletion in mice diverted pyruvate into circulating lactate. This switch disinhibited muscle fatty acid oxidation and drove Cori Cycling that contributed to increased energy expenditure. Loss of muscle MPC activity led to strikingly decreased adiposity with complete muscle mass and strength retention. Notably, despite decreasing muscle glucose oxidation, muscle MPC disruption increased muscle glucose uptake and whole-body insulin sensitivity. Furthermore, chronic and acute muscle MPC deletion accelerated fat mass loss on a normal diet after high fat diet-induced obesity. Our results illuminate the role of the skeletal muscle MPC as a whole-body carbon flux control point. They highlight the potential utility of modulating muscle pyruvate utilization to ameliorate obesity and type 2 diabetes.


Subject(s)
Glucose/metabolism , Metabolic Networks and Pathways , Mitochondria, Muscle/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Pyruvic Acid/metabolism , Thinness , Adiposity , Animals , Anion Transport Proteins/deficiency , Gene Deletion , Lactates/metabolism , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins/deficiency , Monocarboxylic Acid Transporters/deficiency , Muscle Strength
12.
J Clin Invest ; 128(10): 4329-4342, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30047927

ABSTRACT

Movement of circulating fatty acids (FAs) to parenchymal cells requires their transfer across the endothelial cell (EC) barrier. The multiligand receptor cluster of differentiation 36 (CD36) facilitates tissue FA uptake and is expressed in ECs and parenchymal cells such as myocytes and adipocytes. Whether tissue uptake of FAs is dependent on EC or parenchymal cell CD36, or both, is unknown. Using a cell-specific deletion approach, we show that EC, but not parenchymal cell, CD36 deletion increased fasting plasma FAs and postprandial triglycerides. EC-Cd36-KO mice had reduced uptake of radiolabeled long-chain FAs into heart, skeletal muscle, and brown adipose tissue; these uptake studies were replicated using [11C]palmitate PET scans. High-fat diet-fed EC-CD36-deficient mice had improved glucose tolerance and insulin sensitivity. Both EC and cardiomyocyte (CM) deletion of CD36 reduced heart lipid droplet accumulation after fasting, but CM deletion did not affect heart glucose or FA uptake. Expression in the heart of several genes modulating glucose metabolism and insulin action increased with EC-CD36 deletion but decreased with CM deletion. In conclusion, EC CD36 acts as a gatekeeper for parenchymal cell FA uptake, with important downstream effects on glucose utilization and insulin action.


Subject(s)
CD36 Antigens/metabolism , Endothelial Cells/metabolism , Fatty Acids/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Biological Transport, Active/genetics , CD36 Antigens/genetics , Endothelial Cells/pathology , Fatty Acids/genetics , Glucose/genetics , Glucose/metabolism , Humans , Insulin Resistance , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL