Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
1.
Redox Biol ; 52: 102313, 2022 06.
Article in English | MEDLINE | ID: mdl-35447412

ABSTRACT

Lower circulating levels of glycine are consistently reported in association with cardiovascular disease (CVD), but the causative role and therapeutic potential of glycine in atherosclerosis, the underlying cause of most CVDs, remain to be established. Here, following the identification of reduced circulating glycine in patients with significant coronary artery disease (sCAD), we investigated a causative role of glycine in atherosclerosis by modulating glycine availability in atheroprone mice. We further evaluated the atheroprotective potential of DT-109, a recently identified glycine-based compound with dual lipid/glucose-lowering properties. Glycine deficiency enhanced, while glycine supplementation attenuated, atherosclerosis development in apolipoprotein E-deficient (Apoe-/-) mice. DT-109 treatment showed the most significant atheroprotective effects and lowered atherosclerosis in the whole aortic tree and aortic sinus concomitant with reduced superoxide. In Apoe-/- mice with established atherosclerosis, DT-109 treatment significantly reduced atherosclerosis and aortic superoxide independent of lipid-lowering effects. Targeted metabolomics and kinetics studies revealed that DT-109 induces glutathione formation in mononuclear cells. In bone marrow-derived macrophages (BMDMs), glycine and DT-109 attenuated superoxide formation induced by glycine deficiency. This was abolished in BMDMs from glutamate-cysteine ligase modifier subunit-deficient (Gclm-/-) mice in which glutathione biosynthesis is impaired. Metabolic flux and carbon tracing experiments revealed that glycine deficiency inhibits glutathione formation in BMDMs while glycine-based treatment induces de novo glutathione biosynthesis. Through a combination of studies in patients with CAD, in vivo studies using atherosclerotic mice and in vitro studies using macrophages, we demonstrated a causative role of glycine in atherosclerosis and identified glycine-based treatment as an approach to mitigate atherosclerosis through antioxidant effects mediated by induction of glutathione biosynthesis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Disease Models, Animal , Glutamate-Cysteine Ligase , Glutathione/metabolism , Glycine/pharmacology , Glycine/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/metabolism , Superoxides
2.
Cell Rep ; 36(4): 109420, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320345

ABSTRACT

Dysregulated glycine metabolism is emerging as a common denominator in cardiometabolic diseases, but its contribution to atherosclerosis remains unclear. In this study, we demonstrate impaired glycine-oxalate metabolism through alanine-glyoxylate aminotransferase (AGXT) in atherosclerosis. As found in patients with atherosclerosis, the glycine/oxalate ratio is decreased in atherosclerotic mice concomitant with suppression of AGXT. Agxt deletion in apolipoprotein E-deficient (Apoe-/-) mice decreases the glycine/oxalate ratio and increases atherosclerosis with induction of hepatic pro-atherogenic pathways, predominantly cytokine/chemokine signaling and dysregulated redox homeostasis. Consistently, circulating and aortic C-C motif chemokine ligand 5 (CCL5) and superoxide in lesional macrophages are increased. Similar findings are observed following dietary oxalate overload in Apoe-/- mice. In macrophages, oxalate induces mitochondrial dysfunction and superoxide accumulation, leading to increased CCL5. Conversely, AGXT overexpression in Apoe-/- mice increases the glycine/oxalate ratio and decreases aortic superoxide, CCL5, and atherosclerosis. Our findings uncover dysregulated oxalate metabolism via suppressed AGXT as a driver and therapeutic target in atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Molecular Targeted Therapy , Oxalates/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Bile Acids and Salts/metabolism , Cell Line , Chemokine CCL5/metabolism , Cholesterol/metabolism , Dependovirus/metabolism , Female , Glycine/metabolism , Homeostasis , Humans , Inflammation/pathology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Superoxides/metabolism , Transaminases/deficiency , Transaminases/metabolism
4.
Sci Transl Med ; 12(572)2020 12 02.
Article in English | MEDLINE | ID: mdl-33268508

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) including nonalcoholic steatohepatitis (NASH) has reached epidemic proportions with no pharmacological therapy approved. Lower circulating glycine is consistently reported in patients with NAFLD, but the causes for reduced glycine, its role as a causative factor, and its therapeutic potential remain unclear. We performed transcriptomics in livers from humans and mice with NAFLD and found suppression of glycine biosynthetic genes, primarily alanine-glyoxylate aminotransferase 1 (AGXT1). Genetic (Agxt1 -/- mice) and dietary approaches to limit glycine availability resulted in exacerbated diet-induced hyperlipidemia and steatohepatitis, with suppressed mitochondrial/peroxisomal fatty acid ß-oxidation (FAO) and enhanced inflammation as the underlying pathways. We explored glycine-based compounds with dual lipid/glucose-lowering properties as potential therapies for NAFLD and identified a tripeptide (Gly-Gly-L-Leu, DT-109) that improved body composition and lowered circulating glucose, lipids, transaminases, proinflammatory cytokines, and steatohepatitis in mice with established NASH induced by a high-fat, cholesterol, and fructose diet. We applied metagenomics, transcriptomics, and metabolomics to explore the underlying mechanisms. The bacterial genus Clostridium sensu stricto was markedly increased in mice with NASH and decreased after DT-109 treatment. DT-109 induced hepatic FAO pathways, lowered lipotoxicity, and stimulated de novo glutathione synthesis. In turn, inflammatory infiltration and hepatic fibrosis were attenuated via suppression of NF-κB target genes and TGFß/SMAD signaling. Unlike its effects on the gut microbiome, DT-109 stimulated FAO and glutathione synthesis independent of NASH. In conclusion, impaired glycine metabolism may play a causative role in NAFLD. Glycine-based treatment attenuates experimental NAFLD by stimulating hepatic FAO and glutathione synthesis, thus warranting clinical evaluation.


Subject(s)
Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat , Fatty Acids , Glutathione , Glycine , Humans , Liver , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy
5.
Microorganisms ; 8(4)2020 Mar 30.
Article in English | MEDLINE | ID: mdl-32235412

ABSTRACT

During the last few decades there has been a staggering rise in human consumption of soybean-oil (SO). The microbiome and specific taxa composing it are dramatically affected by diet; specifically, by high-fat diets. Increasing evidence indicates the association between dysbiosis and health or disease state, including cardiovascular diseases (CVD) and atherosclerosis pathogenesis in human and animal models. To investigate the effects of high SO intake, C57BL/6 mice were orally supplemented with SO-based emulsion (SOE) for one month, followed by analyses of atherosclerosis-related biomarkers and microbiota profiling by 16S rRNA gene sequencing of fecal DNA. SOE-supplementation caused compositional changes to 64 taxa, including enrichment in Bacteroidetes, Mucispirillum, Prevotella and Ruminococcus, and decreased Firmicutes. These changes were previously associated with atherosclerosis in numerous studies. Among the shifted taxa, 40 significantly correlated with at least one atherosclerosis-related biomarker (FDR < 0.05), while 13 taxa positively correlated with the average of all biomarkers. These microbial alterations also caused a microbial-derived metabolic-pathways shift, including enrichment in different amino-acid metabolic-pathways known to be implicated in CVD. In conclusion, our results demonstrate dysbiosis following SOE supplementation associated with atherosclerosis-related biomarkers. These findings point to the microbiome as a possible mediator to CVD, and it may be implemented into non-invasive diagnostic tools or as potential therapeutic strategies.

7.
J Cardiovasc Pharmacol Ther ; 25(2): 174-186, 2020 03.
Article in English | MEDLINE | ID: mdl-31648564

ABSTRACT

BACKGROUND: Synthetic forms of glucocorticoids (GCs; eg, prednisone, prednisolone) are anti-inflammatory drugs that are widely used in clinical practice. The role of GCs in cardiovascular diseases, including atherosclerosis, is highly controversial, and their impact on macrophage foam cell formation is still unknown. We investigated the effects of prednisone and prednisolone on macrophage oxidative stress and lipid metabolism. METHODS AND RESULTS: C57BL/6 mice were intraperitoneally injected with prednisone or prednisolone (5 mg/kg) for 4 weeks, followed by lipid metabolism analyses in the aorta and peritoneal macrophages. We also analyzed the effect of serum samples obtained from 9 healthy human volunteers before and after oral administration of prednisone (20 mg for 5 days) on J774A.1 macrophage atherogenicity. Finally, J774A.1 macrophages, human monocyte-derived macrophages, and fibroblasts were incubated with increasing concentrations (0-200 ng/mL) of prednisone or prednisolone, followed by determination of cellular oxidative status, and triglyceride and cholesterol metabolism. Prednisone and prednisolone treatment resulted in a significant reduction in triglyceride and cholesterol accumulation in macrophages, as observed in vivo, ex vivo, and in vitro. These effects were associated with GCs' inhibitory effect on triglyceride- and cholesterol-biosynthesis rates, through downregulation of diacylglycerol acyltransferase 1 and HMG-CoA reductase expression. Glucocorticoid-induced reduction of cellular lipid accumulation was mediated by the GC receptors on the macrophages, because the GC-receptor antagonist (RU486) abolished these effects. In fibroblasts, unlike macrophages, GCs showed no effects. CONCLUSION: Prednisone and prednisolone exhibit antiatherogenic activity by protecting macrophages from lipid accumulation and foam cell formation.


Subject(s)
Cholesterol/metabolism , Foam Cells/drug effects , Glucocorticoids/administration & dosage , Lipid Metabolism/drug effects , Macrophages, Peritoneal/drug effects , Prednisolone/administration & dosage , Prednisone/administration & dosage , Triglycerides/metabolism , Administration, Oral , Adolescent , Adult , Animals , Cell Line , Cholesterol/blood , Foam Cells/metabolism , Glucocorticoids/blood , Humans , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Prednisolone/blood , Prednisone/blood , Triglycerides/blood , Young Adult
8.
EBioMedicine ; 41: 62-72, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30772307

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) and resulting nonalcoholic steatohepatitis (NASH) are reaching global epidemic proportions. Lack of non-invasive diagnostic tools and effective therapies constitute two of the major hurdles for a bona fide treatment and a reversal of NASH progression and/or regression of the disease. Nitro-oleic acid (OA-NO2) has been proven effective in multiple experimental models of inflammation and fibrosis. Thus, the potential benefit of in vivo administration of OA-NO2 to treat advanced NAFLD was tested herein in a model of long-term NASH diet-induced liver damage. METHODS: Non-invasive imaging (e.g. photoacustic-ultrasound (PA-US)) was pursued to establish advanced experimental model of NASH in mice in which both steatosis and fibrosis were diagnosed prior experimental therapy with OA-NO2. Experimental controls included equimolar amounts of the non-nitrated oleic acid (OA). CLAMS and NMR-based analysis was used for energy metabolism. FINDINGS: CLAMS and NMR-based analysis demonstrates that OA-NO2 improves body composition and energy metabolism and inhibits hepatic triglyceride (TG) accumulation. Photoacoustic-ultrasound imaging revealed a robust inhibition of liver steatosis and fibrosis by OA-NO2. RNA-sequencing analysis uncovered inflammation and fibrosis as major pathways suppressed by OA-NO2 administration, as well as regulation of lipogenesis and lipolysis pathways, with a robust inhibition of SREBP1 proteolytic activation and subsequent lipogenesis gene expression by OA-NO2. These results were further supported by histological analysis and quantification of lipid accumulation, lobular inflammation (F4/80 staining) and fibrosis (collagen deposition, αSMA staining) as well as established parameters of liver damage (ALT). In vitro studies indicate that OA-NO2 inhibits TG biosynthesis and accumulation in hepatocytes and inhibits fibrogenesis in human stellate cells. INTERPRETATION: OA-NO2 improve steatohepatitis and fibrosis and may constitute an effective therapeutic approach against advanced NAFLD that warrants further clinical evaluation.


Subject(s)
Non-alcoholic Fatty Liver Disease/drug therapy , Oleic Acids/therapeutic use , Animals , Energy Metabolism , Lipogenesis , Liver/diagnostic imaging , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oleic Acids/administration & dosage , Oleic Acids/pharmacology , Proteolysis , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
9.
Eur J Nutr ; 58(2): 879-893, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29804185

ABSTRACT

PURPOSE: Obesity, which is characterized by triglyceride accumulation mainly in adipocytes but also in arterial wall cells such as macrophages, is a major risk factor for developing atherosclerosis. We aimed to identify the crosstalk related to lipid metabolism and oxidation status between adipocytes and macrophages. METHODS: We used a co-culture model system with J477A.1 cultured macrophages and 3T3L1 cultured adipocytes. For an in-vivo co-culture system, we used C57BL/6 mouse peritoneal macrophages and visceral or subcutaneous adipose tissue. RESULTS: Adipocytes significantly increased reactive oxygen species generation, up to twofold, and decreased cholesterol content by 22% in the co-cultured macrophages. Macrophages significantly increased triglyceride-biosynthesis rate by twofold and decreased triglyceride-degradation rate by 30%, resulting in increased triglyceride accumulation in the co-cultured adipocytes by up to 72%. In the in-vivo mouse model, visceral adipose tissue crosstalk with macrophages resulted in a significant pro-atherogenic phenotype with respect to cellular cholesterol metabolism. In contrast, the interaction between subcutaneous adipose tissue and macrophages mostly affected cellular triglyceride metabolism. There were no significant effects on mitochondrial respiration capacity in the macrophages. Upon oxidative-stress reduction in the co-cultured cells using the polyphenol-rich antioxidant, pomegranate juice, the expression of genes related to cellular lipid accumulation was significantly reduced. CONCLUSIONS: We reveal, for the first time, that paracrine interactions between adipocytes and macrophages result in oxidative stress and lipids metabolic alterations in both cells, toward increased atherogenicity which can be reversed by phenolic antioxidants.


Subject(s)
Adipocytes/metabolism , Atherosclerosis/metabolism , Lipid Metabolism/physiology , Macrophages/metabolism , Oxidative Stress/physiology , Adipose Tissue/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
10.
Lipids ; 53(11-12): 1031-1041, 2018 11.
Article in English | MEDLINE | ID: mdl-30560569

ABSTRACT

The polyphenol-rich pomegranate juice (PJ) and the high-density lipoprotein (HDL)-associated paraoxonase1 (PON1) are known as potent atheroprotective antioxidants, but their effects on other tissues related to cardiovascular disease (CVD) remain unknown. The current study aimed to investigate the effects of treating mice with PJ or recombinant PON1 (rePON1) on the oxidation and lipid status of CVD-related tissues: serum, aorta, heart, liver, kidney, visceral, and subcutaneous adipose tissues (VAT and SAT). Both PJ consumption and rePON1 injection decreased the serum levels of thiobarbituric acid-reactive substances (16% and 19%) and triacylglycerols (TAG, 24% and 27%), while only rePON1 increased the levels of thiol groups (35%) and decreased serum cholesterol (15%). Both PJ and rePON1 significantly decreased aortic cholesterol (38% and 32%) and TAG (62% and 58%) contents in association with downregulation of the key TAG biosynthetic enzyme diacylglycerol O-acyltransferase 1 (DGAT1, 71% and 65%), while only PJ decreased aortic lipid peroxides (47%). Substantial TAG-lowering effects of both PJ and rePON1 were observed also in the heart (31% and 42%), liver (34% and 42%), and kidney (42% and 57%). In both VAT and SAT, rePON1 decreased the levels of lipid peroxides (28% and 25%), while PJ decreased the TAG content (22% and 18%). Ex vivo incubation of SAT with serum derived from mice that consumed PJ or injected with rePON1 decreased SAT lipid peroxides (35% or 28%) and TAG mass (12% or 10%). These novel findings highlight potent TAG-lowering properties of exogenous (PJ) and endogenous (PON1) antioxidants in tissues associated with CVD.


Subject(s)
Antioxidants/pharmacology , Aryldialkylphosphatase/pharmacology , Cardiovascular Diseases/blood , Cardiovascular Diseases/metabolism , Lythraceae/chemistry , Plant Extracts/pharmacology , Triglycerides/blood , Animals , Cardiovascular Diseases/drug therapy , Cholesterol/blood , Lipid Peroxidation/drug effects , Male , Mice , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Subcutaneous Fat/drug effects
12.
Rambam Maimonides Med J ; 9(3)2018 Jul 30.
Article in English | MEDLINE | ID: mdl-29944113

ABSTRACT

The strong relationship between cardiovascular diseases (CVD), atherosclerosis, and endogenous or exogenous lipids has been recognized for decades, underestimating the contribution of other dietary components, such as amino acids, to the initiation of the underlying inflammatory disease. Recently, specific amino acids have been associated with incident cardiovascular disorders, suggesting their significant role in the pathogenesis of CVD. Special attention has been paid to the group of branched-chain amino acids (BCAA), leucine, isoleucine, and valine, since their plasma values are frequently found in high concentrations in individuals with CVD risk. Nevertheless, dietary BCAA, leucine in particular, have been associated with improved indicators of atherosclerosis. Therefore, their potential role in the process of atherogenesis and concomitant CVD development remains unclear. Macrophages play pivotal roles in the development of atherosclerosis. They can accumulate high amounts of circulating lipids, through a process known as macrophage foam cell formation, and initiate the atherogenesis process. We have recently screened for anti- or pro-atherogenic amino acids in the macrophage model system. Our study showed that glycine, cysteine, alanine, leucine, glutamate, and glutamine significantly affected macrophage atherogenicity mainly through modulation of the cellular triglyceride metabolism. The anti-atherogenic properties of glycine and leucine, and the pro-atherogenic effects of glutamine, were also confirmed in vivo. Further investigation is warranted to define the role of these amino acids in atherosclerosis and CVD, which may serve as a basis for the development of anti-atherogenic nutritional and therapeutic approaches.

14.
Biofactors ; 44(3): 245-262, 2018 May.
Article in English | MEDLINE | ID: mdl-29399895

ABSTRACT

Whereas atherogenicity of dietary lipids has been largely studied, relatively little is known about the possible contribution of dietary amino acids to macrophage foam-cell formation, a hallmark of early atherogenesis. Recently, we showed that leucine has antiatherogenic properties in the macrophage model system. In this study, an in-depth investigation of the role of leucine in macrophage lipid metabolism was conducted by supplementing humans, mice, or cultured macrophages with leucine. Macrophage incubation with serum obtained from healthy adults supplemented with leucine (5 g/d, 3 weeks) significantly decreased cellular cholesterol mass by inhibiting the rate of cholesterol biosynthesis and increasing cholesterol efflux from macrophages. Similarly, leucine supplementation to C57BL/6 mice (8 weeks) resulted in decreased cholesterol content in their harvested peritoneal macrophages (MPM) in relation with reduced cholesterol biosynthesis rate. Studies in J774A.1 murine macrophages revealed that leucine dose-dependently decreased cellular cholesterol and triglyceride mass. Macrophages treated with leucine (0.2 mM) showed attenuated uptake of very low-density lipoproteins and triglyceride biosynthesis rate, with a concurrent down-regulation of diacylglycerol acyltransferase-1, a key enzyme catalyzing triglyceride biosynthesis in macrophages. Similar effects were observed when macrophages were treated with α-ketoisocaproate, a key leucine metabolite. Finally, both in vivo and in vitro leucine supplementation significantly improved macrophage mitochondrial respiration and ATP production. The above studies, conducted in human, mice, and cultured macrophages, highlight a protective role for leucine attenuating macrophage foam-cell formation by mechanisms related to the metabolism of cholesterol, triglycerides, and energy production. © 2018 BioFactors, 44(3):245-262, 2018.


Subject(s)
Anticholesteremic Agents/pharmacology , Dietary Supplements , Foam Cells/drug effects , Keto Acids/pharmacology , Leucine/pharmacology , Macrophages/drug effects , Adenosine Triphosphate/agonists , Adenosine Triphosphate/biosynthesis , Adolescent , Adult , Animals , Cell Differentiation/drug effects , Cell Line , Cholesterol/biosynthesis , Cholesterol, VLDL/antagonists & inhibitors , Cholesterol, VLDL/biosynthesis , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/metabolism , Dose-Response Relationship, Drug , Foam Cells/cytology , Foam Cells/metabolism , Healthy Volunteers , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis
15.
Immunology ; 152(3): 484-493, 2017 11.
Article in English | MEDLINE | ID: mdl-28672048

ABSTRACT

Acquisition of a 'quiescence programme' by naive T cells is important to provide a stress-free environment and resistance to apoptosis while preserving their responsiveness to activating stimuli. Therefore, the survival and proper function of naive T cells depends on their ability to maintain quiescence. Recently we demonstrated that by preventing chronic unresolved endoplasmic reticulum (ER) stress, Schlafen2 (Slfn2) maintains a stress-free environment to conserve a pool of naive T cells ready to respond to a microbial invasion. These findings strongly suggest an intimate association between quiescence and stress signalling. However, the connection between ER stress conditions and loss of T-cell quiescence is unknown. Here we demonstrate that homeostasis of cholesterol and lipids, is disrupted in T cells and monocytes from Slfn2-mutant, elektra, mice with higher levels of lipid rafts and lipid droplets found in these cells. Moreover, elektra T cells had elevated levels of free cholesterol and cholesteryl ester due to increased de novo synthesis and higher levels of the enzyme HMG-CoA reductase. As cholesterol plays an important role in the transition of T cells from resting to active state, and ER regulates cholesterol and lipid synthesis, we suggest that regulation of cholesterol levels through the prevention of ER stress is an essential component of the mechanism by which Slfn2 regulates quiescence.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Cellular Senescence , Cholesterol/biosynthesis , Lymphocyte Activation , Mutation , T-Lymphocytes/metabolism , Animals , Cell Cycle Proteins/genetics , Cholesterol Esters/biosynthesis , Endoplasmic Reticulum Stress , Genotype , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipid Droplets/metabolism , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/immunology , Monocytes/metabolism , Phenotype , T-Lymphocytes/immunology , Up-Regulation
16.
J Nutr Biochem ; 45: 24-38, 2017 07.
Article in English | MEDLINE | ID: mdl-28431321

ABSTRACT

Atherosclerosis-related research has focused mainly on the effects of lipids on macrophage foam cell formation and atherogenesis, whereas the role of amino acids (AAs) was understudied. The current study aimed to identify anti- or pro-atherogenic AA in the macrophage model system and to elucidate the underlying metabolic and molecular mechanisms. J774A.1 cultured macrophages were treated with increasing concentrations of each 1 of the 20 AAs. Macrophage atherogenicity was assessed in terms of cellular toxicity, generation of reactive oxygen species (ROS) and cellular cholesterol or triglyceride content. At nontoxic concentrations (up to 1 mM), modest effects on ROS generation or cholesterol content were noted, but six specific AAs significantly affected macrophage triglyceride content. Glycine, cysteine, alanine and leucine significantly decreased macrophage triglyceride content (by 24%-38%), through attenuated uptake of triglyceride-rich very low-density lipoprotein (VLDL) by macrophages. In contrast, glutamate and glutamine caused a marked triglyceride accumulation in macrophages (by 107% and 129%, respectively), via a diacylglycerol acyltransferase-1 (DGAT1)-dependent increase in triglyceride biosynthesis rate with a concurrent maturation of the sterol regulatory element-binding protein-1 (SREBP1). Supplementation of apolipoprotein E-deficient (apoE-/-) mice with glycine for 40 days significantly decreased the triglyceride levels in serum and in peritoneal macrophages (MPMs) isolated from the mice (by 19%). In contrast, glutamine supplementation significantly increased MPM ROS generation and the accumulation of cholesterol and that of triglycerides (by 48%), via enhanced uptake of LDL and VLDL. Altogether, the present findings reveal some novel roles for specific AA in macrophage atherogenicity, mainly through modulation of cellular triglyceride metabolism.


Subject(s)
Amino Acids/metabolism , Atherosclerosis/metabolism , Macrophages/pathology , Triglycerides/metabolism , Amino Acids/blood , Amino Acids/pharmacology , Animals , Atherosclerosis/drug therapy , CD36 Antigens/metabolism , Cholesterol/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Lipoproteins, VLDL/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Knockout, ApoE , Receptors, LDL/metabolism , Scavenger Receptors, Class B/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
19.
Biofactors ; 43(1): 100-116, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27517171

ABSTRACT

During the last decades there has been a staggering rise in human consumption of soybean oil (SO) and its major polyunsaturated fatty acid linoleic acid (LA). The role of SO or LA in cardiovascular diseases is highly controversial, and their impact on macrophage foam cell formation, the hallmark of early atherogenesis, is unclear. To investigate the effects of high SO or LA intake on macrophage lipid metabolism and the related mechanisms of action, C57BL/6 mice were orally supplemented with increasing levels of SO-based emulsion or equivalent levels of purified LA for 1 month, followed by analyses of lipid accumulation and peroxidation in aortas, serum and in peritoneal macrophages (MPM) of the mice. Lipid peroxidation and triglyceride mass in aortas from SO or LA supplemented mice were dose-dependently and significantly increased. In MPM from SO or LA supplemented mice, lipid peroxides were significantly increased and a marked accumulation of cellular triglycerides was found in accordance with enhanced triglyceride biosynthesis rate and overexpression of diacylglycerol acyltransferase1 (DGAT1), the key enzyme in triglyceride biosynthesis. In cultured J774A.1 macrophages treated with SO or LA, triglyceride accumulated via increased oxidative stress and a p38 mitogen-activated protein kinase (MAPK)-mediated overexpression of DGAT1. Accordingly, anti-oxidants (pomegranate polyphenols), inhibition of p38 MAPK (by SB202190) or DGAT1 (by oleanolic acid), all significantly attenuated SO or LA-induced macrophage triglyceride accumulation. These findings reveal novel mechanisms by which supplementation with SO or LA stimulate macrophage foam cell formation, suggesting a pro-atherogenic role for overconsumption of SO or LA. © 2016 BioFactors, 43(1):100-116, 2017.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Foam Cells/physiology , Linoleic Acid/pharmacology , Macrophages, Peritoneal/physiology , Soybean Oil/pharmacology , Triglycerides/biosynthesis , Animals , Cell Line , Dietary Supplements , Drug Evaluation, Preclinical , Foam Cells/drug effects , Lipid Peroxidation , Macrophages, Peritoneal/drug effects , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Arch Toxicol ; 91(4): 1709-1725, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27696135

ABSTRACT

The unsaturated aldehyde acrolein is pro-atherogenic, and the polyphenol-rich pomegranate juice (PJ), known for its anti-oxidative/anti-atherogenic properties, inhibits macrophage foam cell formation, the hallmark feature of early atherosclerosis. This study aimed to investigate two unexplored areas of acrolein atherogenicity: macrophage lipid metabolism and the gut microbiota composition. The protective effects of PJ against acrolein atherogenicity were also evaluated. Atherosclerotic apolipoprotein E-deficient (apoE-/-) mice that were fed acrolein (3 mg/kg/day) for 1 month showed significant increases in serum and aortic cholesterol, triglycerides, and lipid peroxides. In peritoneal macrophages isolated from the mice and in J774A.1 cultured macrophages, acrolein exposure increased intracellular oxidative stress and stimulated cholesterol and triglyceride accumulation via enhanced rates of their biosynthesis and over-expression of key regulators of cellular lipid biosynthesis: sterol regulatory element-binding proteins (SREBPs), 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR), and diacylglycerol acyltransferase1 (DGAT1). Acrolein-fed mice demonstrated a major shift in the gut microbiota composition, including a significant phylum-level change in increased Firmicutes and decreased Bacteroidetes. At the family level, acrolein significantly increased the prevalence of Ruminococcaceae and Lachnospiraceae of which the Coprococcus genus was significantly and positively correlated with serum, aortic and macrophage lipid levels and peroxidation. The pro-atherogenic effects of acrolein on serum, aortas, macrophages, and the gut microbiota were substantially abolished by PJ. In conclusion, these findings provide novel mechanisms by which acrolein increases macrophage lipid accumulation and alters the gut microbiota composition in association with enhanced atherogenesis. Moreover, PJ was found as an effective strategy against acrolein atherogenicity.


Subject(s)
Acrolein/toxicity , Atherosclerosis/prevention & control , Lythraceae/chemistry , Macrophages/drug effects , Polyphenols/pharmacology , Animals , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Cell Line , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Lipid Metabolism/drug effects , Macrophages/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Knockout , Oxidative Stress/drug effects , Polyphenols/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...