ABSTRACT
Growing empirical evidence suggests that nutrition and bacterial metabolites might impact the systemic immune response in the context of disease and autoimmunity. We report that long-chain fatty acids (LCFAs) enhanced differentiation and proliferation of T helper 1 (Th1) and/or Th17 cells and impaired their intestinal sequestration via p38-MAPK pathway. Alternatively, dietary short-chain FAs (SCFAs) expanded gut T regulatory (Treg) cells by suppression of the JNK1 and p38 pathway. We used experimental autoimmune encephalomyelitis (EAE) as a model of T cell-mediated autoimmunity to show that LCFAs consistently decreased SCFAs in the gut and exacerbated disease by expanding pathogenic Th1 and/or Th17 cell populations in the small intestine. Treatment with SCFAs ameliorated EAE and reduced axonal damage via long-lasting imprinting on lamina-propria-derived Treg cells. These data demonstrate a direct dietary impact on intestinal-specific, and subsequently central nervous system-specific, Th cell responses in autoimmunity, and thus might have therapeutic implications for autoimmune diseases such as multiple sclerosis.
Subject(s)
Autoimmunity/drug effects , Central Nervous System/immunology , Dietary Fats/pharmacology , Duodenum/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Fatty Acids/pharmacology , Lymphopoiesis/drug effects , T-Lymphocyte Subsets/drug effects , Animals , Dietary Fats/toxicity , Duodenum/metabolism , Duodenum/microbiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acids/chemistry , Fatty Acids/toxicity , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Gene Expression Regulation/immunology , Lauric Acids/toxicity , Liver X Receptors , MAP Kinase Signaling System , Mice , Molecular Weight , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/genetics , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , TranscriptomeABSTRACT
BACKGROUND: Dietary frying oil may have endocrine-disrupting effects, as a feminization effect was observed in cohorts of C57BL/6J male mice fetuses from dams consuming oxidized frying oil (OFO) during pregnancy. OBJECTIVE: The aim of present study was to test the hypothesis that OFO is an anti-androgen. METHODS: In experiment 1, male progeny of Sprague Dawley female rats fed fresh oil or an OFO diet (10 g fat/100 g, from fresh or 24-h-fried soybean oil; [control diet (C) and OFO groups, respectively] from midgestation through lactation were studied. Pups were weaned at 3 wk of age and then consumed their mothers' diet until 9 wk of age. In addition, a group of dams and pups that consumed a high-fat diet (HF; 10 g fried and 20 g fresh soybean oil/100 g) was included to counteract body-weight loss associated with OFO ingestion. Indices of male reproductive development and testosterone homeostasis were measured. In experiment 2, male rats were allocated to C and OFO groups (treated as above) and indices of male fertility compared at 9-10 wk of age. RESULTS: In experiment 1, final body weights of the HF group were lower (17%) than the C group but higher (14%) than the OFO group (P < 0.0001 for each). In addition to abnormalities in seminiferous tubules, HF and OFO groups did not differ from one another, but, compared with the C group, had delayed preputial separation (4.9 d) and reductions in serum testosterone concentrations (17-74%), anogenital distance (8-20%), weights of androgen-dependent tissues (8-30%), testicular testosterone and cholesterol concentrations (30-40%), and mRNA levels of genes involved in steroidogenesis and cholesterol homeostasis (30-70%). In experiment 2, OFO-exposed males had 20% lower sperm motility (P < 0.05); however, when mated to normal females, pregnancy rates and litter sizes did not differ between OFO and C groups. CONCLUSIONS: The anti-androgenic effect of OFO in Sprague Dawley rats was attributed to decreased testicular concentrations of cholesterol (testosterone precursor) and not body-weight loss.
Subject(s)
Cholesterol/metabolism , Homeostasis/drug effects , Soybean Oil/toxicity , Testis/drug effects , Testosterone/metabolism , Animal Nutritional Physiological Phenomena , Animals , Cooking , Dietary Fats/administration & dosage , Dietary Fats/toxicity , Female , Male , Oxidation-Reduction , Pregnancy , Prenatal Exposure Delayed Effects , Prenatal Nutritional Physiological Phenomena , Rats , Rats, Sprague-Dawley , Testis/metabolismABSTRACT
Liver fatty acid binding protein (L-Fabp) modulates lipid trafficking in enterocytes, hepatocytes, and hepatic stellate cells (HSCs). We examined hepatocyte vs. HSC L-Fabp deletion in hepatic metabolic adaptation and fibrotic injury. Floxed L-Fabp mice were bred to different transgenic Cre mice or injected with adeno-associated virus type 8 (AAV8) Cre and fed diets to promote steatosis and fibrosis or were subjected to either bile duct ligation or CCl4 injury. Albumin-Cre-mediated L-Fabp deletion revealed recombination in hepatocytes and HSCs; these findings were confirmed with 2 other floxed alleles. Glial fibrillary acid protein-Cre and platelet-derived growth factor receptor ß-Cre-mediated L-Fabp deletion demonstrated recombination only in HSCs. Mice with albumin promoter-driven Cre recombinase (Alb-Cre)-mediated or AAV8-mediated L-Fabp deletion were protected against food withdrawal-induced steatosis. Mice with Alb-Cre-mediated L-Fabp deletion were protected against high saturated fat-induced steatosis and fibrosis, phenocopying germline L-Fabp-/- mice. Mice with HSC-specific L-Fabp deletion exhibited retinyl ester depletion yet demonstrated no alterations in fibrosis. On the other hand, fibrogenic resolution after CCl4 administration was impaired in mice with Alb-Cre-mediated L-Fabp deletion. These findings suggest cell type-specific roles for L-Fabp in mitigating hepatic steatosis and in modulating fibrogenic injury and reversal.-Newberry, E. P., Xie, Y., Lodeiro, C., Solis, R., Moritz, W., Kennedy, S., Barron, L., Onufer, E., Alpini, G., Zhou, T., Blaner, W. S., Chen, A., Davidson, N. O. Hepatocyte and stellate cell deletion of liver fatty acid binding protein reveal distinct roles in fibrogenic injury.
Subject(s)
Carbon Tetrachloride Poisoning/metabolism , Fatty Acid-Binding Proteins/physiology , Fatty Liver/metabolism , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Albumins/genetics , Animals , Bile Ducts , Carbon Tetrachloride Poisoning/pathology , Crosses, Genetic , Dependovirus/genetics , Dietary Fats/toxicity , Fatty Acid-Binding Proteins/deficiency , Fatty Acids/toxicity , Fatty Liver/etiology , Fatty Liver/pathology , Female , Fibrosis , Food Deprivation , Gene Deletion , Genes, Synthetic , Hepatic Stellate Cells/pathology , Hepatocytes/pathology , Integrases , Ligation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Promoter Regions, GeneticABSTRACT
Objective- Vascular smooth muscle cell (VSMC) transformation to an osteochondrogenic phenotype is an initial step toward arterial calcification, which is highly correlated with cardiovascular disease-related morbidity and mortality. TLR2 (Toll-like receptor 2) plays a pathogenic role in the development of vascular diseases, but its regulation in calcification of arteries and VSMCs remains unclear. We postulate that TLR2-mediated inflammation participates in mediating atherosclerotic arterial calcification and VSMC calcification. Approach and Results- We found that ApoE-/- Tlr2-/- genotype in mice suppressed high-fat diet-induced atherosclerotic plaques formation during initiation but progressively lost its preventative capacity, compared with ApoE-/- mice. However, TLR2 deficiency prohibited high-fat diet-induced advanced atherosclerotic calcification, chondrogenic metaplasia, and OPG (osteoprotegerin) downregulation in the calcified lesions. Incubation of VSMCs in a calcifying medium revealed that TLR2 agonists significantly increased VSMC calcification and chondrogenic differentiation. Furthermore, TLR2 deficiency suppressed TLR2 agonist-mediated VSMC chondrogenic differentiation and consequent calcification, which were triggered via the concerted actions of IL (interleukin)-6-mediated RANKL (receptor activator of nuclear factor κB ligand) induction and OPG suppression. Inhibition experiments with pharmacological inhibitors demonstrated that IL-6-mediated RANKL induction is signaled by p38 and ERK1/2 (extracellular signal-regulated kinase 1/2) pathways, whereas the OPG is suppressed via NF-κB (nuclear factor κB) dependent signaling mediated by ERK1/2. Conclusions- We concluded that on ligand binding, TLR2 activates p38 and ERK1/2 signaling to selectively modulate the upregulation of IL-6-mediated RANKL and downregulation of OPG. These signaling pathways act in concert to induce chondrogenic transdifferentiation of VSMCs, which in turn leads to vascular calcification during the pathogenesis of atherosclerosis.
Subject(s)
Atherosclerosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Chondrogenesis/physiology , Interleukin-6/physiology , MAP Kinase Signaling System , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , Toll-Like Receptor 2/physiology , Animals , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Calcinosis/genetics , Cells, Cultured , Cholesterol, Dietary/toxicity , Diet, High-Fat/adverse effects , Dietary Fats/toxicity , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Random AllocationABSTRACT
INTRODUCTION AND AIM: Patients with NASH have increased risk for sepsis or cardiovascular disease after Liver transplantation. An important role of Toll-like receptor (TLR) 4 in the pathogenesis of nonalcoholic steatohepatitis (NASH) was demonstrated. Here, we study the role of miR-182-5p in TLR4 expression and high-fat-diet (HFD)-induced NASH in vitro and in vivo Material and methods. Following transfection with a miR-182-5p mimic, the effect of miR-182-5p on TLR4 in RAW264.7 and HepG2 cells was investigated. Following administration of the miR-182-5p mimic into the livers of HFD-induced NASH mice, we determined the in vivo expression of TLR4, TNFa, and IL-6 and assessed the histologic features of the livers. Results Following lipopolysaccharide (LPS) treatment of RAW264.7 cells, real-time RT-PCR and western blot results indicated decreases levels of TLR4 mRNA and protein in the miR-182-5p group as compared with levels observed in controls, with similar trends were observed in TNFa and IL-6 protein levels. Following oleic acid (OA) treatment of HepG2 cells, TLR4, TNFa, and IL-6 levels were significantly decreased in the miR-182-5p group as compared with levels observed in controls. Following miR-182-5p administration, TLR4 mRNA and protein levels decreased along with those of TNFa and IL-6 proteins, and the liver weight/body weight ratio of treated mice was less than that observed in controls. Furthermore, hematoxylin and eosin staining showed that the miR-182-5p-treated group exhibited low adiposecell cross-sectional areas, and Oil Red O staining showed decreases in the size of lipid droplets in the miR-182-5p-treated group. CONCLUSIONS: miR-182-5p ameliorated HFD-induced NASH by suppressing TLR4.
Subject(s)
Liver/pathology , MicroRNAs/pharmacology , Non-alcoholic Fatty Liver Disease/genetics , Animals , Blotting, Western , Cells, Cultured , Dietary Fats/toxicity , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , RNA/genetics , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/geneticsABSTRACT
Background: Palmitic acid (PA) is the main saturated fatty acid naturally occurring in animal fats and vegetable oils. In recent decades, palm oil, an alternative lipid source containing high amounts of PA, has been widely used to replace fish oil in aquafeed. Objective: We investigated the hepatotoxicity of PA in zebrafish and the underlying mechanism. Methods: One-month-old zebrafish fed a high-fat diet (HFD) containing 16% soybean oil and 3 PA-incorporated HFDs [4%, 8%, and 12% PA (12PA)] for 2 wk (experiment 1) and 4 wk (experiment 2) were used to evaluate PA-induced liver damage and endoplasmic reticulum (ER) stress. Germ-free (GF) zebrafish fed low-fat, high-fat, or 12PA diets for 5 d were used to study the direct effects of PA on liver damage (experiment 3). GF zebrafish colonized with HFD or 12PA microbiota for 48 h were used to elucidate the indirect effects of PA-altered microbiota on liver damage (experiment 4). Last, GF zebrafish colonized with HFD or 12PA microbiota were used to evaluate the effects of different microbiotas on PA absorption (experiment 5). Results: In experiment 1, the proportion of PA in the liver linearly increased as its percentage in dietary lipid increased (r2 = 0.83, P < 0.05). In experiment 2, the expression of glucose-regulated protein 78 (Grp78) and C/EBP-homologous protein (Chop) was higher in the 12PA group than in the HFD group (2.2- and 2.7-fold, respectively; P < 0.05). The activity of caspase-12 was increased by 61.1% in the 12PA group compared with the HFD group (P < 0.05). In experiment 3, caspase-12 activity was higher in the 12PA group than in the HFD group (P < 0.05). In experiment 4, GF zebrafish colonized with PA-altered microbiota had higher caspase-12 activity (P < 0.05) than those colonized by HFD microbiota. In experiment 5, PA-altered microbiota promoted PA absorption (P < 0.05) and aggravated ER stress and liver damage in the context of high-PA feeding. Conclusions: The PA-altered microbiota indirectly induced ER stress and liver damage in zebrafish. Moreover, the PA microbiota promoted the absorption of PA, leading to enhanced PA overflow into the liver and aggravated hepatotoxicity of PA in zebrafish.
Subject(s)
Chemical and Drug Induced Liver Injury/microbiology , Dietary Fats/toxicity , Endoplasmic Reticulum Stress , Gastrointestinal Microbiome , Liver/drug effects , Palmitic Acid/toxicity , Animal Feed , Animals , Caspase 12/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Liver/metabolism , Palm Oil/chemistry , Palm Oil/toxicity , ZebrafishABSTRACT
Heart failure is associated with generalized insulin resistance. Moreover, insulin-resistant states such as type 2 diabetes mellitus and obesity increases the risk of heart failure even after adjusting for traditional risk factors. Insulin resistance or type 2 diabetes mellitus alters the systemic and neurohumoral milieu, leading to changes in metabolism and signaling pathways in the heart that may contribute to myocardial dysfunction. In addition, changes in insulin signaling within cardiomyocytes develop in the failing heart. The changes range from activation of proximal insulin signaling pathways that may contribute to adverse left ventricular remodeling and mitochondrial dysfunction to repression of distal elements of insulin signaling pathways such as forkhead box O transcriptional signaling or glucose transport, which may also impair cardiac metabolism, structure, and function. This article will review the complexities of insulin signaling within the myocardium and ways in which these pathways are altered in heart failure or in conditions associated with generalized insulin resistance. The implications of these changes for therapeutic approaches to treating or preventing heart failure will be discussed.
Subject(s)
Heart Failure/physiopathology , Insulin/physiology , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Dietary Fats/toxicity , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Fatty Acids/metabolism , Forecasting , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Heart Failure/etiology , Heart Failure/prevention & control , Heart Failure/therapy , Humans , Hyperinsulinism/physiopathology , Insulin Resistance/physiology , Mitochondria, Heart/physiology , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Obesity/complications , Obesity/physiopathology , Protein Transport , Receptor, Insulin/physiology , Risk Factors , Signal Transduction/physiology , Ventricular Remodeling/physiologyABSTRACT
RETRACTED: Obesity is a significant risk factor for acute respiratory distress syndrome. The mechanisms underlying this association are unknown. We recently showed that diet-induced obese mice exhibit pulmonary vascular endothelial dysfunction, which is associated with enhanced susceptibility to LPS-induced acute lung injury. Here, we demonstrate that lung endothelial dysfunction in diet-induced obese mice coincides with increased endoplasmic reticulum (ER) stress. Specifically, we observed enhanced expression of the major sensors of misfolded proteins, including protein kinase R-like ER kinase, inositol-requiring enzyme α, and activating transcription factor 6, in whole lung and in primary lung endothelial cells isolated from diet-induced obese mice. Furthermore, we found that primary lung endothelial cells exposed to serum from obese mice, or to saturated fatty acids that mimic obese serum, resulted in enhanced expression of markers of ER stress and the induction of other biological responses that typify the lung endothelium of diet-induced obese mice, including an increase in expression of endothelial adhesion molecules and a decrease in expression of endothelial cell-cell junctional proteins. Similar changes were observed in lung endothelial cells and in whole-lung tissue after exposure to tunicamycin, a compound that causes ER stress by blocking N-linked glycosylation, indicating that ER stress causes endothelial dysfunction in the lung. Treatment with 4-phenylbutyric acid, a chemical protein chaperone that reduces ER stress, restored vascular endothelial cell expression of adhesion molecules and protected against LPS-induced acute lung injury in diet-induced obese mice. Our work indicates that fatty acids in obese serum induce ER stress in the pulmonary endothelium, leading to pulmonary endothelial cell dysfunction. Our work suggests that reducing protein load in the ER of pulmonary endothelial cells might protect against acute respiratory distress syndrome in obese individuals.
Subject(s)
Acute Lung Injury/physiopathology , Endoplasmic Reticulum Stress/physiology , Endothelial Cells/metabolism , Fatty Acids, Nonesterified/blood , Fatty Acids/blood , Lung/pathology , Obesity/physiopathology , Unfolded Protein Response/physiology , Activating Transcription Factor 6/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Animals , Cell Adhesion Molecules/metabolism , Dietary Fats/therapeutic use , Dietary Fats/toxicity , Disease Susceptibility , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Fatty Acids/pharmacology , Fatty Acids, Monounsaturated/therapeutic use , Lipopolysaccharides/toxicity , Lung/metabolism , Male , Mice , Mice, Inbred AKR , Obesity/blood , Obesity/complications , Phenylbutyrates/pharmacology , Protein Serine-Threonine Kinases/metabolism , Serum , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolismABSTRACT
Epidemiological studies reveal associations between obesity/metabolic syndrome and mood disorders. We assessed behavioural changes in rats fed diets enriched in fat and fructose in different proportions and correlated the observed alterations with biochemical changes induced by the diets. Three groups of rats were used as follows: control (C) animals fed regular rat chow, rats fed high-fat diet (HF) and rats fed high-fat and high-fructose diet (HFHF). HF and HFHF animals were also given a 10% fructose solution as drinking water. Behavioural and biochemical parameters were determined. Anxiety was measured by the open-field and the social interaction test. Depression-like behaviour was evaluated by the forced swimming test. The object recognition test was utilized to assess effects on memory. Diet-exposed animals displayed signs of anxiety in the open-field (HF rats had reduced central time; HFHF rats had reduced number of central entries) and in the social interaction test (decreased time of interaction in HF group). In the forced swimming test, the immobility time was prolonged in the HFHF group. While different measures of anxiety scores correlated with visceral adiposity and dyslipidemia, results from both social interaction and forced swimming tests were significantly associated with lipid peroxidation, which in turn also correlated with the metabolic parameters. The experimental diets did not affect the object recognition memory. Both experimental diets induced metabolic derangements in rats and provoked similar anxiety- and depression-like behaviours. Lipid peroxidation seems to play a role in translating diet-induced metabolic alterations into behavioural disorders.
Subject(s)
Anxiety/etiology , Depression/etiology , Diet, High-Fat/adverse effects , Fructose/toxicity , Lipid Peroxidation/drug effects , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Depression/metabolism , Dietary Fats/toxicity , Insulin Resistance , Lipid Peroxidation/physiology , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Rats, Wistar , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Social BehaviorABSTRACT
BACKGROUND: As the prevalence of nonalcoholic fatty liver disease (NAFLD), including steatosis and nonalcoholic steatohepatitis, is increasing, novel dietary approaches are required for the prevention and treatment of NAFLD. OBJECTIVE: We evaluated the potential of mung bean protein isolate (MuPI) to prevent NAFLD progression. METHODS: In Expts. 1 and 2, the hepatic triglyceride (TG) concentration was compared between 8-wk-old male mice fed a high-fat diet (61% of energy from fat) containing casein, MuPI, and soy protein isolate and an MuPI-constituent amino acid mixture as a source of amino acids (18% of energy) for 4 wk. In Expt. 3, hepatic fatty acid synthase (Fasn) expression was evaluated in 8-wk-old male Fasn-promoter-reporter mice fed a casein- or MuPI-containing high-fat diet for 20 wk. In Expt. 4, hepatic fibrosis was examined in 8-wk-old male mice fed an atherogenic diet (61% of energy from fat, containing 1.3 g cholesterol/100 g diet) containing casein or MuPI (18% of energy) as a protein source for 20 wk. RESULTS: In the high fat-diet mice, the hepatic TG concentration in the MuPI group decreased by 66% and 47% in Expt. 1 compared with the casein group (P < 0.001) and the soy protein isolate group (P = 0.001), respectively, and decreased by 56% in Expt. 2 compared with the casein group (P = 0.011). However, there was no difference between the MuPI-constituent amino acid mixture and casein groups in Expt. 2. In Expt. 3, Fasn-promoter-reporter activity and hepatic TG concentration were lower in the MuPI group than in those fed casein (P < 0.05). In Expt. 4, in mice fed an atherogenic diet, hepatic fibrosis was not induced in the MuPI group, whereas it developed overtly in the casein group. CONCLUSION: MuPI potently reduced hepatic lipid accumulation in mice and may be a potential foodstuff to prevent NAFLD onset and progression.
Subject(s)
Dietary Proteins/administration & dosage , Fatty Liver/prevention & control , Inflammation/prevention & control , Liver Cirrhosis/prevention & control , Vigna/chemistry , Animals , Dietary Fats/toxicity , Dietary Proteins/analysis , Fatty Acid Synthase, Type I/metabolism , Fatty Liver/chemically induced , Gene Expression Regulation , Inflammation/metabolism , Liver Cirrhosis/metabolism , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, TransgenicABSTRACT
RATIONALE: Chemokine-controlled arterial leukocyte recruitment is a crucial process in atherosclerosis. Formyl peptide receptor 2 (FPR2) is a chemoattractant receptor that recognizes proinflammatory and proresolving ligands. The contribution of FPR2 and its proresolving ligand annexin A1 to atherosclerotic lesion formation is largely undefined. OBJECTIVE: Because of the ambivalence of FPR2 ligands, we here investigate the role of FPR2 and its resolving ligand annexin A1 in atherogenesis. METHODS AND RESULTS: Deletion of FPR2 or its ligand annexin A1 enhances atherosclerotic lesion formation, arterial myeloid cell adhesion, and recruitment. Mechanistically, we identify annexin A1 as an endogenous inhibitor of integrin activation evoked by the chemokines CCL5, CCL2, and CXCL1. Specifically, the annexin A1 fragment Ac2-26 counteracts conformational activation and clustering of integrins on myeloid cells evoked by CCL5, CCL2, and CXCL1 through inhibiting activation of the small GTPase Rap1. In vivo administration of Ac2-26 largely diminishes arterial recruitment of myeloid cells in a FPR2-dependent fashion. This effect is also observed in the presence of selective antagonists to CCR5, CCR2, or CXCR2, whereas Ac2-26 was without effect when all 3 chemokine receptors were antagonized simultaneously. Finally, repeated treatment with Ac2-26 reduces atherosclerotic lesion sizes and lesional macrophage accumulation. CONCLUSIONS: Instructing the annexin A1-FPR2 axis harbors a novel approach to target arterial leukocyte recruitment. With the ability of Ac2-26 to counteract integrin activation exerted by various chemokines, delivery of Ac2-26 may be superior in inhibition of arterial leukocyte recruitment when compared with blocking individual chemokine receptors.
Subject(s)
Annexin A1/physiology , Aortic Diseases/etiology , Atherosclerosis/etiology , Animals , Annexin A1/deficiency , Annexin A1/genetics , Annexin A1/pharmacology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Chemokine CCL2/physiology , Chemokine CCL5/physiology , Chemokine CXCL1/physiology , Chemotaxis/drug effects , Dietary Fats/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/physiology , Peptides/pharmacology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR5/physiology , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/physiology , Receptors, Interleukin-8B/antagonists & inhibitors , rap1 GTP-Binding Proteins/physiologyABSTRACT
RATIONALE: Hyperlipidemia and type 2 diabetes mellitus (T2DM) severely impair adaptive vascular growth responses in ischemic muscles. This is largely attributed to dysregulated gene expression, although details of the changes are unknown. OBJECTIVE: To define the role of promoter methylation in adaptive vascular growth in hyperlipidemia (LDLR(-/-)ApoB(100/100)) and T2DM (IGF-II/LDLR(-/-)ApoB(100/100)) mouse models of hindlimb ischemia. METHODS AND RESULTS: Unilateral hindlimb ischemia was induced by ligating femoral artery. Perfusion was assessed using ultrasound, and capillary and arteriole parameters were assessed using immunohistochemistry. Genome-wide methylated DNA sequencing was performed with DNA isolated from ischemic muscle, tissue macrophages (MÏs), and endothelial cells. Compared with the controls, hyperlipidemia and T2DM mice showed impaired perfusion recovery, which was associated with impaired angiogenesis and arteriogenesis. Genome-wide proximal promoter DNA methylation analysis suggested differential patterns of methylation in MÏ genes in ischemic muscles. Classically activated M1-MÏ gene promoters, including Cfb, Serping1, and Tnfsf15, were significantly hypomethylated, whereas alternatively activated M2-MÏ gene promoters, including Nrp1, Cxcr4, Plxnd1, Arg1, Cdk18, and Fes, were significantly hypermethylated in MÏs isolated from hyperlipidemia and T2DM ischemic muscles compared with controls. These results combined with mRNA expression and immunohistochemistry showed the predominance of proinflammatory M1-MÏs, compared with anti-inflammatory and proangiogenic M2-MÏs in hyperlipidemia and T2DM ischemic muscles. CONCLUSIONS: We found significant promoter hypomethylation of genes typical for proinflammatory M1-MÏs and hypermethylation of anti-inflammatory, proangiogenic M2-MÏ genes in hyperlipidemia and T2DM ischemic muscles. Epigenetic alterations modify MÏ phenotype toward proinflammatory M1 as opposed to anti-inflammatory, proangiogenic, and tissue repair M2 phenotype, which may contribute to the impaired adaptive vascular growth under these pathological conditions.
Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Gene Expression Regulation/genetics , Hindlimb/blood supply , Hyperlipidemias/genetics , Ischemia/pathology , Macrophages/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/genetics , Promoter Regions, Genetic , Reperfusion Injury/genetics , Adaptation, Physiological , Animals , Apolipoprotein B-100/genetics , Arterioles/pathology , Capillaries/pathology , Dietary Fats/toxicity , Disease Models, Animal , Endothelial Cells/metabolism , Genome-Wide Association Study , Hindlimb/diagnostic imaging , Inflammation , Ischemia/diagnostic imaging , Ischemia/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Receptors, LDL/deficiency , Reperfusion , Reperfusion Injury/pathology , UltrasonographyABSTRACT
RATIONALE: Proinflammatory adaptive immune responses are recognized as major drivers of atherosclerotic lesion formation. Although CD8(+) T cells have recently been proposed as a proatherogenic cell subset, their full scope of actions remains to be elucidated. OBJECTIVE: We here addressed the contribution of CD8(+) T cells to monocyte trafficking in atherosclerosis. METHOD AND RESULTS: We observed that CD8(+) T cells express proinflammatory cytokines (interferon-γ, tumor necrosis factor-α, and interleukin-12) within atherosclerotic lesions and spleens of high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Antibody-mediated CD8(+) T-cell depletion in high-fat diet-fed Ldlr(-/-) mice decreased atherosclerotic plaque formation, associated with decreased macrophage accumulation within lesions. Despite a reduction in vascular chemokine (CC-motif) ligand 2 and chemokine (CXC-motif) ligand 1 expression, CD8(+) T-cell depletion did not directly affect monocyte recruitment to inflamed vessels. However, CD8(+) T-cell depletion decreased chemokine (CC-motif) ligand serum concentrations and circulating Ly6C(high) monocyte counts. We further evidenced that CD8(+) T-cell depletion decreased levels of mature monocytes and myeloid granulocyte-monocyte progenitors in the bone marrow and spleen of hypercholesterolemic mice, effects that were partially reproduced by interferon-γ neutralization, showing a role for interferon-γ. CONCLUSIONS: These data suggest that CD8(+) T cells promote atherosclerosis by controlling monopoiesis and circulating monocyte levels, which ultimately contributes to plaque macrophage burden without affecting direct monocyte recruitment, identifying this cell subset as a critical regulator of proatherogenic innate immune cell responses in atherosclerosis.
Subject(s)
Antigens, Ly/analysis , Atherosclerosis/immunology , CD8-Positive T-Lymphocytes/immunology , Monocytes/immunology , Myelopoiesis/immunology , Animals , Antilymphocyte Serum/therapeutic use , Atherosclerosis/etiology , Bone Marrow/metabolism , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/metabolism , Carotid Stenosis/immunology , Carotid Stenosis/pathology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Diet, Atherogenic/adverse effects , Dietary Fats/toxicity , Endarterectomy, Carotid , Gene Expression Regulation/immunology , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/etiology , Interferon-gamma/physiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/geneticsABSTRACT
A novel medium- and long-chain triacylglycerols (MLCT), with 30% (w/w) medium-chain fatty acids (MCFA) was evaluated for its safety as a dietary fat in mice and rats. The subacute oral toxicity study showed that the maximum tolerated dose exceeded 54.33 g/kg body weight (kg bw)/day. In the 90-day feeding study, no dose-related adverse effects were observed in rats administered diets formulated with different levels of MLCT (2.0, 4.0, and 8.0 g/kg bw/day) as compared to the rapeseed oil control diet. Further safety assessment in pregnant rats did not reveal any significant difference relative to the control at a treatment level up to 8.0 g MLCT/kg bw/day. The results from this study indicated the safe use of MLCT with high contents of MCFA in food products for improving human health.
Subject(s)
Dietary Fats/toxicity , Fatty Acids/toxicity , Triglycerides/toxicity , Animals , Dietary Fats/administration & dosage , Fatty Acids/chemistry , Mice , Rapeseed Oil/toxicity , Rats , Triglycerides/chemistryABSTRACT
The pyruvate dehydrogenase complex (PDH) has been hypothesized to link lipid exposure to skeletal muscle insulin resistance through a glucose-fatty acid cycle in which increased fatty acid oxidation increases acetyl-CoA concentrations, thereby inactivating PDH and decreasing glucose oxidation. However, whether fatty acids induce insulin resistance by decreasing PDH flux remains unknown. To genetically examine this hypothesis we assessed relative rates of pyruvate dehydrogenase flux/mitochondrial oxidative flux and insulin-stimulated rates of muscle glucose metabolism in awake mice lacking pyruvate dehydrogenase kinase 2 and 4 [double knockout (DKO)], which results in constitutively activated PDH. Surprisingly, increased glucose oxidation in DKO muscle was accompanied by reduced insulin-stimulated muscle glucose uptake. Preferential myocellular glucose utilization in DKO mice decreased fatty acid oxidation, resulting in increased reesterification of acyl-CoAs into diacylglycerol and triacylglycerol, with subsequent activation of PKC-θ and inhibition of insulin signaling in muscle. In contrast, other putative mediators of muscle insulin resistance, including muscle acylcarnitines, ceramides, reactive oxygen species production, and oxidative stress markers, were not increased. These findings demonstrate that modulation of oxidative substrate selection to increase muscle glucose utilization surprisingly results in muscle insulin resistance, offering genetic evidence against the glucose-fatty acid cycle hypothesis of muscle insulin resistance.
Subject(s)
Insulin Resistance/physiology , Protein Serine-Threonine Kinases/deficiency , Pyruvate Dehydrogenase Complex/metabolism , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Citric Acid Cycle , Dietary Fats/pharmacology , Dietary Fats/toxicity , Enzyme Activation , Fatty Acids/metabolism , Glucose/metabolism , Glycogen/metabolism , Hyperinsulinism/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle, Skeletal/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C-theta , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/genetics , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Substrate SpecificityABSTRACT
RATIONALE: Sialylation by α2,3-sialyltransferases has been shown to be a crucial glycosylation step in the generation of functional selectin ligands. Recent evidence suggests that sialylation also affects the binding of chemokines to their corresponding receptor. OBJECTIVE: Because the chemokine receptors for Ccl5 and Ccl2 are important in atherogenic recruitment of neutrophils and monocytes, we here investigated the role of α2,3-sialyltransferase IV (ST3Gal-IV) in Ccl5- and Ccl2-mediated myeloid cell arrest and further studied its relevance in a mouse model of atherosclerosis. METHODS AND RESULTS: St3Gal4-deficient myeloid cells showed a reduced binding of Ccl5 and an impaired Ccl5-triggered integrin activation. Correspondingly, Ccl5-induced arrest on tumor necrosis factor-α-stimulated endothelium was almost completely abrogated, as observed in flow chamber adhesion assays and during ex vivo perfusion or intravital microscopy of carotid arteries. Moreover, Ccl5-triggered neutrophil and monocyte extravasation into the peritoneal cavity was severely reduced in St3Gal4(-/-) mice. In contrast, St3Gal4 deficiency did not significantly affect Ccl2 binding and only marginally decreased Ccl2-induced flow arrest of myeloid cells. In agreement with the crucial role of leukocyte accumulation in atherogenesis, and the importance of Ccl5 chemokine receptors mediating myeloid cell recruitment to atherosclerotic vessels, St3Gal4 deficiency drastically reduced the size, stage, and inflammatory cell content of atherosclerotic lesions in Apoe(-/-) mice on high-fat diet. CONCLUSIONS: In summary, these findings identify ST3Gal-IV as a promising target to reduce inflammatory leukocyte recruitment and arrest.
Subject(s)
Atherosclerosis/enzymology , Chemokine CCL5/physiology , Leukocyte Rolling/physiology , Myeloid Cells/pathology , Sialyltransferases/deficiency , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Chemokine CCL2/metabolism , Dietary Fats/toxicity , Female , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylneuraminic Acid/metabolism , Neuraminidase/pharmacology , Protein Processing, Post-Translational , Sialyltransferases/genetics , Sialyltransferases/physiology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , beta-Galactoside alpha-2,3-SialyltransferaseABSTRACT
This article gives a brief overview of the available scientific data on the toxicity of lipid oxidation products. The description is logically divided into two parts: the toxicity of primary oxidation products (lipid peroxides) and toxicity of secondary oxidation products (carbonyl compounds). Mechanisms of their toxic effect, indicating the most sensitive target organ, as well as the metabolic pathways and main products of their metabolism and half-lethal doses of the main products of oxidation are characterized. It is noted that the most toxic products among them are secondary products of lipid oxidation, in particular, the acrolein with a half-lethal dose of 7-46 mg per kg body weight. It is concluded that the accent in the control of lipid and lipid containing food safety must be shifted to identify specific, the most toxic, secondary lipid oxidation products.
Subject(s)
Dietary Fats/toxicity , Food Safety , Lipid Metabolism/drug effects , Lipids/toxicity , Animals , Humans , Oxidation-ReductionABSTRACT
OBJECTIVE: Shear stress-induced pseudopod retraction is an anti-inflammatory measure that minimizes neutrophil activity and is regulated by membrane cholesterol. We tested the hypothesis that a hypercholesterolemic impairment of shear mechanotransduction alters the neutrophil flow behavior leading to microvascular dysfunction. APPROACH AND RESULTS: We examined the shear effects on the flow behavior of human leukocytes. When subjected to shearing during cone-plate viscometry, leukocyte suspensions exhibited parallel time-dependent reductions in viscosity and pseudopod activity. Shear-induced reductions in suspension viscosity were attenuated by membrane cholesterol enrichment. We also showed that enhanced pseudopod activity of leukocyte suspensions in 10% hematocrit significantly (P<0.05) raised the flow resistance of microvascular mimics. These results implicate an impaired neutrophil pseudopod retraction response to shear in hypercholesterolemic microvascular dysfunction. We confirmed this using near-infrared diffuse correlation spectroscopy to assess skeletal muscle blood flow regulation in the hindlimbs of mice subjected to reactive hyperemia. Using a custom protocol for the mouse, we extrapolated an adjusted peak flow and time to adjusted peak flow to quantify the early phase of the blood flow recovery response during reactive hyperemia when shear mechanobiology likely has a maximal impact. Compared with mice on normal diet, hypercholesterolemic mice exhibited significantly (P<0.05) reduced adjusted peak flow and prolonged time to adjusted peak flow which correlated (r=0.4 and r=-0.3, respectively) with neutrophil shear responsiveness and were abrogated by neutropenia. CONCLUSIONS: These results provide the first evidence that the neutrophils contribute to tissue blood flow autoregulation. Moreover, a deficit in the neutrophil responsiveness to shear may be a feature of hypercholesterolemia-related microvascular dysfunction.
Subject(s)
Hemorheology , Hypercholesterolemia/blood , Leukocytes/physiology , Microcirculation , Animals , Blood Viscosity , Cholesterol/blood , Cholesterol/physiology , Dietary Fats/toxicity , Hindlimb/blood supply , Humans , Hyperemia/blood , Leukocytes/drug effects , Male , Mechanotransduction, Cellular/physiology , Membrane Lipids/blood , Membrane Lipids/physiology , Mice , Mice, Knockout , Microcirculation/physiology , Muscle, Skeletal/blood supply , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutropenia/blood , Neutrophils/physiology , Pseudopodia/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Regional Blood Flow , Shear Strength/physiology , Spectroscopy, Near-Infrared , SuspensionsABSTRACT
OBJECTIVE: Atherosclerosis is driven by inflammatory reactions that are shared with the innate immune system. Toll-like receptor-9 (TLR9) is an intracellular pattern recognition receptor of the innate immune system that is currently under clinical investigation as a therapeutic target in inflammatory diseases. Here, we investigated whether TLR9 has a role in the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Newly generated double-knockout ApoE(-/-):TLR9(-/-) mice and control ApoE(-/-) mice were fed a high-fat diet from 8 weeks and effects on lesion size, cellular composition, inflammatory status, and plasma lipids were assessed after 8, 12, 15, and 20 weeks. All 4 time points demonstrated exacerbated atherosclerotic lesion severity in ApoE(-/-):TLR9(-/-) mice, with a corresponding increase in lipid deposition and accumulation of macrophages, dendritic cells, and CD4(+) T cells. Although ApoE(-/-):TLR9(-/-) mice exhibited an increase in plasma very low-density lipoprotein/low-density-lipoprotein cholesterol, the very low-density lipoprotein/low-density lipoprotein:high-density lipoprotein ratio was unaltered because of a parallel increase in plasma high-density lipoprotein cholesterol. As a potential mechanism accounting for plaque progression in ApoE(-/-):TLR9(-/-) mice, CD4(+) T-cell accumulation was further investigated and depletion of these cells in ApoE(-/-):TLR9(-/-) mice significantly reduced lesion severity. As a final translational approach, administration of a TLR9 agonist (type B CpG oligodeoxynucleotide 1668) to ApoE(-/-) mice resulted in a reduction of lesion severity. CONCLUSIONS: Genetic deletion of the innate immune receptor TLR9 exacerbated atherosclerosis in ApoE(-/-) mice fed a high-fat diet. CD4(+) T cells were identified as potential mediators of this effect. A type B CpG oligodeoxynucleotide TLR9 agonist reduced lesion severity, thus identifying a novel therapeutic approach in atherosclerosis.
Subject(s)
Atherosclerosis/prevention & control , Toll-Like Receptor 9/physiology , Animals , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Dietary Fats/toxicity , Disease Progression , Lipoproteins, HDL/blood , Lipoproteins, VLDL/blood , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/therapeutic use , Random Allocation , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunologyABSTRACT
OBJECTIVE: Unresolved inflammatory response of macrophages plays a pivotal role in the pathogenesis of atherosclerosis. Previously we showed that ribosomal protein L13a-dependent translational silencing suppresses the synthesis of a cohort of inflammatory proteins in monocytes and macrophages. We also found that genetic abrogation of L13a expression in macrophages significantly compromised the resolution of inflammation in a mouse model of lipopolysaccharide-induced endotoxemia. However, its function in the pathogenesis of atherosclerosis is not known. Here, we examine whether L13a in macrophage has a protective role against high-fat diet-induced atherosclerosis. APPROACH AND RESULTS: We bred the macrophage-specific L13a knockout mice L13a Flox(+/+) Cre(+/+) onto apolipoprotein E-deficient background and generated the experimental double knockout mice L13a Flox(+/+) Cre(+/+) apolipoprotein E deficient (apoE(-/-)). L13a Flox(+/+) Cre(-/-) mice on apolipoprotein E-deficient background were used as controls. Control and knockout mice were subjected to high-fat diet for 10 weeks. Evaluation of aortic sinus sections and entire aorta by en face showed significantly higher atherosclerosis in the knockout mice. Severity of atherosclerosis in knockout mice was accompanied by thinning of the smooth muscle cell layer in the media, larger macrophage area in the intimal plaque region and higher plasma levels of inflammatory cytokines. In addition, macrophages isolated from knockout mice had higher polyribosomal abundance of several target mRNAs, thus showing defect in translation control. CONCLUSIONS: Our data demonstrate that loss of L13a in macrophages increases susceptibility to atherosclerosis in apolipoprotein E-deficient mice, revealing an important role of L13a-dependent translational control as an endogenous protection mechanism against atherosclerosis.