Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Psychoneuroendocrinology ; 102: 114-120, 2019 04.
Article in English | MEDLINE | ID: mdl-30544002

ABSTRACT

BACKGROUND: Posttraumatic stress disorder (PTSD) is associated with disturbed sleep and elevated levels of pro-inflammatory cytokines, including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Studies in animals and healthy humans have also shown that disrupted sleep elevates pro-inflammatory cytokines, including IL-6 and TNF-α. A better understanding of overnight cytokine levels and sleep might shed light on possible mechanisms for elevated inflammation in PTSD. Thus, we investigated overnight levels of IL-6 and TNF-α in individuals with and without PTSD while recording sleep polysomnography (PSG). METHOD: Serum samples were collected from otherwise healthy, medication-free participants with chronic PTSD (n = 44; 50% female; M age = 30.34 ± 8.11) and matched controls (n = 49; 53% female; M age = 30.53 ± 6.57) during laboratory PSG. Levels of IL-6 and TNF-α were measured at hours 0, 2, 4, 6, and 8 after typical sleep onset time using serial serum samples. Plasma IL-6 and TNF-α levels were quantified using enzyme-linked immunosorbent assays. RESULTS: Growth model analysis indicated a significantgroup by time interaction for IL-6 (t[247] = -2.92, p = .005) and a significant group by sex by time interaction for TNF-α (t[275] = 2.02, p = .04). PTSD positive men and women initially had higher IL-6 and TNF-α at sleep onset, but not at the end of their sleep cycle. Men with PTSD showed a peak of TNF-α at the end of the sleep cycle, whereas male control subjects demonstrated an inverted U-shaped profile. There were no significant differences in TNF-α levels overnight between women with and without PTSD. CONCLUSION: To our knowledge, this is the largest study to examine IL-6 overnight in a PTSD sample and the first study to examine overnight TNF-α in PTSD. Overnight IL-6 and TNF-α levels may be altered in individuals with PTSD compared to those without PTSD, and TNF-α trajectories also differed by sex. The current findings highlight the need to consider sex, sleep, time of day, and circadian variation when examining inflammation in PTSD. Additional research in broader study samples will be necessary to clarify associations between disrupted sleep, cytokines, and increased risk for disease in PTSD.


Subject(s)
Cytokines/analysis , Stress Disorders, Post-Traumatic/metabolism , Adult , Cytokines/blood , Female , Humans , Hydrocortisone/analysis , Hydrocortisone/blood , Inflammation/blood , Inflammation/metabolism , Interleukin-6/analysis , Interleukin-6/blood , Male , Middle Aged , Polysomnography , Sleep , Sleep Wake Disorders , Stress Disorders, Post-Traumatic/physiopathology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood
2.
Psychoneuroendocrinology ; 51: 557-66, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25465168

ABSTRACT

BACKGROUND: Inflammation may reduce hippocampal volume by blocking neurogenesis and promoting neurodegeneration. Posttraumatic stress disorder (PTSD) has been linked with both elevated inflammation and reduced hippocampal volume. However, few studies have examined associations between inflammatory markers and hippocampal volume, and none have examined these associations in the context of PTSD. METHODS: We measured levels of the inflammatory markers interleukin-6 (IL-6) and soluble receptor II for tumor necrosis factor (sTNF-RII) as well as hippocampal volume in 246 Gulf War veterans with and without current and past PTSD as assessed with the Clinician Administered PTSD Scale (CAPS). Enzyme-linked immunosorbent assays were used to measure inflammatory markers, and 1.5Tesla magnetic resonance imaging (MRI) and Freesurfer version 4.5 were used to quantify hippocampal volume. Hierarchical linear regression and analysis of covariance models were used to examine if hippocampal volume and PTSD status would be associated with elevated levels of IL-6 and sTNF-RII. RESULTS: Increased sTNF-RII, but not IL-6, was significantly associated with reduced hippocampal volume (ß=-0.14, p=0.01). The relationship between sTNF-RII and hippocampal volume was independent of potential confounds and covariates, including PTSD status. Although we observed no PTSD diagnosis-related differences in either IL-6 or sTNF-RII, higher PTSD severity was associated with significantly increased sTNF-RII (ß=0.24, p=0.04) and reduced IL-6 levels (ß=-0.24, p=0.04). CONCLUSIONS: Our results indicate that specific inflammatory proteins may be associated with brain structure and function as indexed by hippocampal volume and PTSD symptoms.


Subject(s)
Hippocampus/pathology , Inflammation/blood , Interleukin-6/blood , Receptors, Tumor Necrosis Factor, Type II/blood , Stress Disorders, Post-Traumatic/diagnosis , Veterans/psychology , Adult , Aged , Female , Gulf War , Humans , Inflammation/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Severity of Illness Index , Stress Disorders, Post-Traumatic/blood , Stress Disorders, Post-Traumatic/pathology
3.
J Lipid Res ; 55(12): 2501-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25320346

ABSTRACT

Many of the beneficial and adverse effects of niacin are mediated via a G protein receptor, G protein-coupled receptor 109A/hydroxycarboxylic acid 2 receptor (GPR109A/HCA2), which is highly expressed in adipose tissue and macrophages. Here we demonstrate that immune activation increases GPR109A/HCA2 expression. Lipopolysaccharide (LPS), TNF, and interleukin (IL) 1 increase GPR109A/HCA2 expression 3- to 5-fold in adipose tissue. LPS also increased GPR109A/HCA2 mRNA levels 5.6-fold in spleen, a tissue rich in macrophages. In peritoneal macrophages and RAW cells, LPS increased GPR109A/HCA2 mRNA levels 20- to 80-fold. Zymosan, lipoteichoic acid, and polyinosine-polycytidylic acid, other Toll-like receptor activators, and TNF and IL-1 also increased GPR109A/HCA2 in macrophages. Inhibition of the myeloid differentiation factor 88 or TIR-domain-containing adaptor protein inducing IFNß pathways both resulted in partial inhibition of LPS stimulation of GPR109A/HCA2, suggesting that LPS signals an increase in GPR109A/HCA2 expression by both pathways. Additionally, inhibition of NF-κB reduced the ability of LPS to increase GPR109A/HCA2 expression by ∼50% suggesting that both NF-κB and non-NF-κB pathways mediate the LPS effect. Finally, preventing the LPS-induced increase in GPR109A/HCA2 resulted in an increase in TG accumulation and the expression of enzymes that catalyze TG synthesis. These studies demonstrate that inflammation stimulates GPR109A/HCA2 and there are multiple intracellular signaling pathways that mediate this effect. The increase in GPR109A/HCA2 that accompanies macrophage activation inhibits the TG accumulation stimulated by macrophage activation.


Subject(s)
Disease Models, Animal , Intra-Abdominal Fat/metabolism , Macrophage Activation , Macrophages, Peritoneal/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Signal Transduction , Up-Regulation , 3T3-L1 Cells , Animals , Cell Line, Transformed , Cells, Cultured , Female , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/pathology , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/pathology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Lipid Metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mycoses/immunology , Mycoses/metabolism , Mycoses/pathology , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/pathology
4.
J Leukoc Biol ; 92(4): 829-39, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22753953

ABSTRACT

LPS treatment of macrophages induces TG accumulation, which is accentuated by TG-rich lipoproteins or FFA. We defined pathways altered during macrophage activation that contribute to TG accumulation. Glucose uptake increased with activation, accompanied by increased GLUT1. Oxidation of glucose markedly decreased, whereas incorporation of glucose-derived carbon into FA and sterols increased. Macrophage activation also increased uptake of FFA, associated with an increase in CD36. Oxidation of FA was markedly reduced, whereas the incorporation of FA into TGs increased, associated with increased GPAT3 and DGAT2. Additionally, macrophage activation decreased TG lipolysis; however, expression of ATGL or HSL was not altered. Macrophage activation altered gene expression similarly when incubated with exogenous FA or AcLDL. Whereas activation with ligands of TLR2 (zymosan), TLR3 (poly I:C), or TLR4 (LPS) induced alterations in macrophage gene expression, leading to TG accumulation, treatment of macrophages with cytokines had minimal effects. Thus, activation of TLRs leads to accumulation of TG in macrophages by multiple pathways that may have beneficial effects in host defense but could contribute to the accelerated atherosclerosis in chronic infections and inflammatory diseases.


Subject(s)
Macrophage Activation , Macrophages/metabolism , Triglycerides/metabolism , Animals , Cell Line , Fatty Acids/metabolism , Gene Expression/drug effects , Glucose/metabolism , Lipolysis , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mice , Myeloid Differentiation Factor 88/physiology , Toll-Like Receptors/physiology
5.
Biochem Biophys Res Commun ; 421(3): 612-5, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22538368

ABSTRACT

Angiopoietin like protein 4 (ANGPTL4) inhibits lipoprotein lipase (LPL) activity. Previous studies have shown that Toll-like Receptor (TLR) activation increases serum levels of ANGPTL4 and expression of ANGPTL4 in liver, heart, muscle, and adipose tissue in mice. ANGPTL4 is expressed in macrophages and is induced by inflammatory saturated fatty acids. The absence of ANGPTL4 leads to the increased uptake of pro-inflammatory saturated fatty acids by macrophages in the mesentery lymph nodes due to the failure of ANGPTL4 to inhibit LPL activity, resulting in peritonitis, intestinal fibrosis, weight loss, and death. Here we determined the effect of TLR activation on the expression of macrophage ANGPTL4. LPS treatment resulted in a 70% decrease in ANGPTL4 expression in mouse spleen, a tissue enriched in macrophages. In mouse peritoneal macrophages, LPS treatment also markedly decreased ANGPTL4 expression. In RAW cells, a macrophage cell line, LPS, zymosan, poly I:C, and imiquimod all inhibited ANGPTL4 expression. In contrast, neither TNF, IL-1, nor IL-6 altered ANGPTL4 expression. Finally, in cholesterol loaded macrophages, LPS treatment still decreased ANGPTL4 expression. Thus, while in most tissues ANGPTL4 expression is stimulated by inflammatory stimuli, in macrophages TLR activators inhibit ANGPTL4 expression, which could lead to a variety of down-stream effects important in host defense and wound repair.


Subject(s)
Angiopoietins/biosynthesis , Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , Toll-Like Receptors/metabolism , Aminoquinolines/pharmacology , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Female , Imiquimod , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology , Toll-Like Receptors/agonists , Zymosan/immunology , Zymosan/pharmacology
6.
Endocrinology ; 153(6): 2689-700, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22474187

ABSTRACT

The acute phase response (APR) produces marked alterations in lipid and carbohydrate metabolism including decreasing plasma ketone levels. Fibroblast growth factor 21 (FGF21) is a recently discovered hormone that regulates lipid and glucose metabolism and stimulates ketogenesis. Here we demonstrate that lipopolysaccharide (LPS), zymosan, and turpentine, which induce the APR, increase serum FGF21 levels 2-fold. Although LPS, zymosan, and turpentine decrease the hepatic expression of FGF21, they increase FGF21 expression in adipose tissue and muscle, suggesting that extrahepatic tissues account for the increase in serum FGF21. After LPS administration, the characteristic decrease in plasma ketone levels is accentuated in FGF21-/- mice, but this is not due to differences in expression of carnitine palmitoyltransferase 1α or hydroxymethyglutaryl-CoA synthase 2 in liver, because LPS induces similar decreases in the expression of these genes in FGF21-/- and control mice. However, in FGF21-/- mice, the ability of LPS to increase plasma free fatty acid levels is blunted. This failure to increase plasma free fatty acid could contribute to the accentuated decrease in plasma ketone levels because the transport of fatty acids from adipose tissue to liver provides the substrate for ketogenesis. Treatment with exogenous FGF21 reduced the number of animals that die and the rapidity of death after LPS administration in leptin-deficient ob/ob mice and to a lesser extent in control mice. FGF21 also protected from the toxic effects of cecal ligation and puncture-induced sepsis. Thus, FGF21 is a positive APR protein that protects animals from the toxic effects of LPS and sepsis.


Subject(s)
Acute-Phase Reaction/metabolism , Fibroblast Growth Factors/metabolism , Leptin/deficiency , Sepsis/metabolism , 3T3-L1 Cells , Acute-Phase Reaction/blood , Acute-Phase Reaction/etiology , Animals , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Nonesterified/blood , Female , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Kaplan-Meier Estimate , Ketones/blood , Leptin/genetics , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/deficiency , PPAR alpha/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/blood , Sepsis/physiopathology
7.
Innate Immun ; 18(2): 231-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21450790

ABSTRACT

Inhibition of adipocyte triglyceride biosynthesis is required for fatty acid mobilization during inflammation. Triglyceride biosynthesis requires glycerol 3-phosphate and phosphoenolpyruvate carboxykinase (PEPCK) plays a key role. We demonstrate that LPS, zymosan, and TNF-α decrease PEPCK in liver and fat. Turpentine decreases PEPCK in liver, but not in fat. The LPS-induced decrease in PEPCK does not occur in TLR4 deficient animals, indicating that this receptor is required. The LPS-induced decrease in hepatic PEPCK does not occur in TNF receptor/IL-1 receptor knockout mice, but occurs in fat, indicating that TNF-α/IL-1 is essential for the decrease in liver but not fat. In 3T3-L1 adipocytes TNF-α, IL-1, IL-6, and IFNγ inhibit PEPCK indicating that there are multiple pathways by which PEPCK is decreased in adipocytes. The binding of PPARγ and RXRα to the PPARγ response element in the PEPCK promoter is markedly decreased in adipose tissue nuclear extracts from LPS treated animals. Lipopolysaccharide and zymosan reduce PPARγ and RXRα expression in fat, suggesting that a decrease in PPARγ and RXRα accounts for the decrease in PEPCK. Thus, there are multiple cytokine pathways by which inflammation inhibits PEPCK expression in adipose tissue which could contribute to the increased mobilization of fatty acids during inflammation.


Subject(s)
Adipose Tissue/enzymology , Inflammation/enzymology , Liver/enzymology , Phosphoenolpyruvate Carboxylase/biosynthesis , 3T3-L1 Cells , Animals , Cytokines/biosynthesis , DNA, Complementary/biosynthesis , DNA, Complementary/isolation & purification , Electrophoretic Mobility Shift Assay , Fatty Acids, Nonesterified/metabolism , Female , Gluconeogenesis/drug effects , Inflammation/chemically induced , Lipolysis/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Retinoid X Receptor alpha/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Turpentine/pharmacology , Zymosan/pharmacology
8.
Inflamm Res ; 60(10): 991-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21751047

ABSTRACT

OBJECTIVE AND DESIGN: The aim of this study was to examine the expression of G protein-coupled receptor 81 (GPR81) in mouse adipose tissue in response to inflammatory stimuli. GPR81 is activated by lactate resulting in the inhibition of lipolysis. MATERIALS AND TREATMENT: Mice were injected with saline, lipopolysaccharide (LPS), zymosan, or turpentine, N = 5 per group. 3T3-L1 adipocytes were treated with tumor necrosis factor alpha, interleukin (IL)-l beta, IL-6, or interferon gamma. METHODS: GPR81 expression levels were measured by real-time PCR and statistical significance was determined by Student's t test. RESULTS: LPS resulted in a marked decrease in GPR81 mRNA level in mouse adipose tissue in C57BL/6 and OuJ mice, an effect that was not observed in HeJ mice, which have a mutation in TLR4. Zymosan and turpentine also decreased adipose tissue GPR81 expression. Cytokine treatment of 3T3-L1 adipocytes had no effect on GPR81 expression. GPR81 expression was decreased in ob/ob mice, an animal model of type 2 diabetes that is characterized by inflammation. CONCLUSION: Inflammation decreases the expression of GPR81 in adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Inflammation/metabolism , Receptors, G-Protein-Coupled/biosynthesis , 3T3-L1 Cells/cytology , Adipocytes/metabolism , Animals , Cytokines/metabolism , Female , Interferon-gamma/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipolysis , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/metabolism
9.
Rheumatology (Oxford) ; 50(8): 1458-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21441551

ABSTRACT

OBJECTIVE: Patients with RA have systemic inflammation and increased risk of cardiovascular (CV) events, including thrombosis. Levels of fibrinogen, a pro-thrombotic protein with predictive value for CV disease (CVD), are elevated during systemic inflammation. We compared circulating fibrinogen levels in patients with RA with healthy controls and evaluated the relationship with measures of disease activity. METHODS: Patients with RA and controls were recruited at the University of California, San Francisco (UCSF). Disease activity was evaluated using standard composite indices. Fibrinogen, ESR, serum CRP, acute-phase serum amyloid A and levels of selected cytokines were quantified. RESULTS: A total of 105 RA patients and 62 controls were studied. Among patients with RA, disease activity ranged from quiescent to highly active disease. Circulating fibrinogen levels were significantly higher in RA than in controls [median (interquartile range) 466 (391-575) vs 367 (309-419) mg/dl, respectively, P < 0.0001]. This difference remained highly statistically significant after adjustment for demographic variables and BMI. Although fibrinogen correlated significantly with clinical measures of disease activity, significantly elevated levels were observed at low levels of activity, even in RA patients with no detectable swollen or tender joints. In multivariable models, ~ 80% of the increased fibrinogen in RA was accounted for by increases in CRP and ESR. CONCLUSION: Circulating levels of fibrinogen are elevated in RA and correlated with markers of inflammation, but only modestly correlate with clinical assessments of disease activity. Even RA patients with excellent clinical disease control exhibit elevated levels compared with controls.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibrinogen/analysis , Adult , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/physiopathology , Biomarkers/blood , Cytokines/blood , Female , Health Status , Humans , Joints/pathology , Joints/physiopathology , Male , Middle Aged , Severity of Illness Index
10.
Innate Immun ; 17(2): 174-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20100709

ABSTRACT

Carbohydrate response element binding protein (ChREBP) is a recently discovered transcription factor whose levels and activity are increased by glucose leading to the activation of target genes, which include acetyl-CoA carboxylase, fatty acid synthase, and liver-type pyruvate kinase. Here, we demonstrate that lipopolysaccharide (LPS) treatment causes a marked decrease in ChREBP mRNA and protein levels in the liver of mice fed a normal chow diet or in mice fasted for 24 h and then re-fed a high carbohydrate diet. This decrease occurs rapidly and is a sensitive response (half-maximal dose 0.1 µg/mouse). The decrease in ChREBP is accompanied by a decrease in the expression of ChREBP target genes. Zymosan and turpentine treatment also decrease hepatic ChREBP levels and the expression of its target genes. Additionally, tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1ß) decrease liver ChREBP expression both in vivo and in Hep3B cells in culture. Finally, LPS decreased ChREBP expression in muscle and adipose tissue. These studies demonstrate that ChREBP is down-regulated during the acute phase response resulting in alterations in the expression of ChREBP regulated target genes. Thus, ChREBP joins a growing list of transcription factors that are regulated during the acute phase response.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Line , Dietary Carbohydrates/administration & dosage , Endotoxins/immunology , Endotoxins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Turpentine/metabolism , Zymosan/immunology , Zymosan/metabolism
11.
Biochem Biophys Res Commun ; 391(4): 1737-41, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20043872

ABSTRACT

The acute phase response is characterized by elevations in serum triglyceride levels due to both an increase in hepatic VLDL production and a delay in the clearance of triglyceride rich lipoproteins secondary to a decrease in lipoprotein lipase (LPL) activity. Recently there has been a marked increase in our understanding of factors that regulate LPL activity. GPIHBP1 facilitates the interaction of LPL and lipoproteins thereby allowing lipolysis to occur. Angiopoietin like proteins (ANGPTL) 3 and 4 inhibit LPL activity. In the present study, treatment of mice with LPS, an activator of TLR4 and a model of Gram-negative infections, did not alter the expression of GPIHBP1 in heart or adipose tissue. However, LPS decreased the expression of ANGPTL3 in liver and increased the expression of ANGPTL4 in heart, muscle, and adipose tissue. Serum ANGPTL4 protein levels were markedly increased at 8 and 16h following LPS treatment. Administration of zymosan, an activator of TLR2 and a model of fungal infections, also increased serum ANGPTL4 protein and mRNA levels in liver, heart, muscle, and adipose tissue. Finally, treatment of 3T3-L1 adipocytes with LPS or cytokines (TNF alpha, IL-1 beta, and interferon gamma) stimulated ANGPTL4 expression. These studies demonstrate that ANGPTL4 is a positive acute phase protein and the increase in ANGPTL4 could contribute to the hypertriglyceridemia that characteristically occurs during the acute phase response by inhibiting LPL activity.


Subject(s)
Acute-Phase Reaction/metabolism , Angiopoietins/biosynthesis , Hypertriglyceridemia/metabolism , 3T3-L1 Cells , Adipose Tissue/metabolism , Angiopoietin-Like Protein 4 , Animals , Female , Lipopolysaccharides , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardium/metabolism
12.
Atherosclerosis ; 209(1): 81-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19748622

ABSTRACT

Activation of macrophages by TLR agonists enhances foam cell formation, but the underlying mechanisms are not understood. We examined the effects of TLR agonists on ADRP/ADFP, a protein associated with forming lipid droplets, and Mal1 a fatty acid-binding protein, in two mouse macrophage cell lines and human monocytes. Low doses of LPS, a TLR4 agonist increased both mRNA and protein levels of ADRP/ADFP and Mal1 in RAW 264.7 macrophages. Following pretreatment with Intralipid, fatty acids, or acetyl-LDL to increase triglyceride or cholesterol ester storage, LPS treatment still increased ADRP/ADFP and Mal1 mRNA levels. LPS also induced ADRP/ADFP and Mal1 in J774 macrophages and ADRP/ADFP in human monocytes. Zymosan, a fungal product that activates TLR2, poly-I:C, a viral mimetic that activates TLR3, and imiquimod, a TLR7 agonist, also increased ADRP/ADFP. Zymosan, but not poly-I:C or imiquimod, induced Mal1. In contrast, neither gene was induced by TNFalpha, IL-1beta, IL-6, or interferon-gamma. Thus TLR agonists induce ADRP/ADFP and Mal1, which likely contributes to macrophage triglyceride and cholesterol ester storage leading to foam cell formation.


Subject(s)
Atherosclerosis/immunology , Fatty Acid-Binding Proteins/biosynthesis , Macrophages/immunology , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Toll-Like Receptors/agonists , Aminoquinolines/pharmacology , Animals , Cholesterol Esters/metabolism , Humans , Imiquimod , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages/drug effects , Mice , Perilipin-2 , Poly I-C/pharmacology , Toll-Like Receptors/immunology , Triglycerides/metabolism , Zymosan/pharmacology
13.
J Lipid Res ; 50(10): 2055-63, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19443862

ABSTRACT

Respiratory failure is a major cause of mortality during septic shock and is due in part to decreased ventilatory muscle contraction. Ventilatory muscles have high energy demands; fatty acid (FA) oxidation is an important source of ATP. FA oxidation is regulated by nuclear hormone receptors; studies have shown that the expression of these receptors is decreased in liver, heart, and kidney during sepsis. Here, we demonstrate that lipopolysaccharide (LPS) decreases FA oxidation and the expression of lipoprotein lipase (LPL), FA transport protein 1 (FATP-1), CD36, carnitine palmitoyltransferase beta, medium chain acyl-CoA dehydrogenase (MCAD), and acyl-CoA synthetase, key proteins required for FA uptake and oxidation, in the diaphragm. LPS also decreased mRNA levels of PPARalpha and beta/delta, RXRalpha, beta, and gamma, thyroid hormone receptor alpha and beta, and estrogen related receptor alpha (ERRalpha) and their coactivators PGC-1alpha, PGC-1beta, SRC1, SRC2, Lipin 1, and CBP. Zymosan resulted in similar changes in the diaphragm. Finally, in PPARalpha deficient mice, baseline CPT-1beta and FATP-1 levels were markedly decreased and were not further reduced by LPS suggesting that a decrease in the PPARalpha signaling pathway plays an important role in inducing some of these changes. The decrease in FA oxidation in the diaphragm may be detrimental, leading to decreased diaphragm contraction and an increased risk of respiratory failure during sepsis.


Subject(s)
Diaphragm/metabolism , Lipopolysaccharides/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Blotting, Western , Diaphragm/drug effects , Fatty Acids/metabolism , Female , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Polymerase Chain Reaction , Triglycerides/metabolism
14.
Am J Physiol Endocrinol Metab ; 295(6): E1502-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18940942

ABSTRACT

Infection and inflammation affect adipose triglyceride metabolism, resulting in increased plasma free fatty acid (FFA) and VLDL levels during the acute-phase response. Lipin-1, a multifunctional protein, plays a critical role in adipose differentiation, mitochondrial oxidation, and triglyceride synthesis. Here, we examined whether LPS [a Toll-like receptor (TLR)-4 activator], zymosan (a TLR-2 activator), and proinflammatory cytokines regulate lipin-1 in adipose tissue. LPS administration caused a marked decrease in the levels of lipin-1 mRNA and protein in adipose tissue. The decrease in lipin-1 mRNA levels occurred rapidly and lasted for at least 24 h. In contrast, lipin-2 and -3 mRNA levels did not change, suggesting specific repression of lipin-1. Zymosan similarly decreased lipin-1 mRNA without affecting lipin-2 or lipin-3 mRNA levels. To determine the pathways by which LPS repressed lipin-1, we examined the effect of proinflammatory cytokines on cultured adipocytes. In 3T3-L1 adipocytes, TNF-alpha, IL-1beta, and IFN-gamma, but not LPS or IL-6, caused a decrease in lipin-1 mRNA levels. Furthermore, TNF-alpha and IL-1beta administration also decreased mRNA levels of lipin-1 in adipose tissue in mice. Importantly, the LPS-induced decrease in lipin-1 mRNA levels was significantly but not totally blunted in TNF-alpha/IL-1 receptor-null mice compared with controls, suggesting key roles for TNF-alpha/IL-1beta and other cytokines in mediating LPS-induced repression of lipin-1. Together, our results demonstrate that expression of lipin-1, one of the essential triglyceride synthetic enzymes, was suppressed by LPS, zymosan, and proinflammatory cytokines in mouse adipose tissue and in cultured 3T3-L1 adipocytes, which could contribute to a decrease in the utilization of FFA to synthesize triglycerides in adipose tissue, thus promoting the release of FFA into the circulation.


Subject(s)
Adipocytes/drug effects , Adipose Tissue/drug effects , Cytokines/pharmacology , Lipopolysaccharides/pharmacology , Nuclear Proteins/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Female , Gene Expression Regulation/drug effects , Inflammation Mediators/pharmacology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Phosphatidate Phosphatase , RNA, Messenger/metabolism , Zymosan/pharmacology
15.
Biochem Biophys Res Commun ; 374(2): 341-4, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18638454

ABSTRACT

Inflammation induces marked changes in lipid and lipoprotein metabolism. Proprotein convertase subtilisin kexin 9 (PCSK9) plays an important role in regulating LDL receptor degradation. Here, we demonstrate that LPS decreases hepatic LDL receptor protein but at the same time hepatic LDL receptor mRNA levels are not decreased. We therefore explored the effect of LPS on PCSK9 expression. LPS results in a marked increase in hepatic PCSK9 mRNA levels (4h 2.5-fold increase; 38h 12.5-fold increase). The increase in PCSK9 is a sensitive response with 1microg LPS inducing a (1/2) maximal response. LPS also increased PCSK9 expression in the kidney. Finally, zymosan and turpentine, other treatments that induce inflammation, also stimulated hepatic expression of PCSK9. Thus, inflammation stimulates PCSK9 expression leading to increased LDL receptor degradation and decreasing LDL receptors thereby increasing serum LDL, which could have beneficial effects on host defense.


Subject(s)
Inflammation/enzymology , Receptors, LDL/metabolism , Serine Endopeptidases/biosynthesis , Animals , Female , Humans , Inflammation/chemically induced , Inflammation/immunology , Irritants/toxicity , Kidney/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Liver/metabolism , Mice , Mice, Inbred C57BL , Proprotein Convertase 9 , Proprotein Convertases , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, LDL/blood , Serine Endopeptidases/genetics , Turpentine/toxicity , Zymosan/toxicity
16.
J Lipid Res ; 49(10): 2179-87, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18574256

ABSTRACT

Inflammation produces marked changes in lipid metabolism, including increased serum fatty acids (FAs) and triglycerides (TGs), increased hepatic TG production and VLDL secretion, increased adipose tissue lipolysis, and decreased FA oxidation in liver and heart. Lipopolysaccharide (LPS) also increases TG and cholesteryl ester levels in kidneys. Here we confirm these findings and define potential mechanisms. LPS decreases renal FA oxidation by 40% and the expression of key proteins required for oxidation of FAs, including FA transport protein-2, fatty acyl-CoA synthase, carnitine palmitoyltransferase-1, medium-chain acyl-CoA dehydrogenase, and acyl-CoA oxidase. Similar decreases were observed in peroxisome proliferator-activated receptor alpha (PPARalpha)-deficient mice. LPS also caused a reduction in renal mRNA levels of PPARalpha (75% decrease), thyroid hormone receptor alpha (TRalpha) (92% decrease), and TRbeta (84% decrease), whereas PPARbeta/delta and gamma were not altered. Expression of PGC1 alpha and beta, coactivators required for PPARs and TR, was also decreased in kidneys of LPS-treated mice, as were mitochondrial genes regulated by PGC1 (Atp5g1, COX5a, Idh3a, and Ndufs8). Decreased renal FA oxidation could be a by-product of the systemic coordinated host response to increase FAs and TGs available for host defense and/or tissue repair. However, the kidney requires energy to support its transport functions, and the inability to generate energy via FA oxidation might contribute to the renal failure seen in severe sepsis.


Subject(s)
Fatty Acids/metabolism , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Fatty Acids/genetics , Female , Gene Expression Regulation/drug effects , Male , Mice , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptors/deficiency , Peroxisome Proliferator-Activated Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Thyroid Hormone/genetics , Triglycerides/blood
17.
Atherosclerosis ; 199(1): 19-26, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18054359

ABSTRACT

Inflammation can produce abnormalities that could increase the risk for atherosclerosis including alterations in lipid and lipoprotein metabolism. Apolipoprotein M is a recently described HDL-associated apoprotein expressed mainly in the liver and kidney with protective effects against atherosclerosis. In this study, we describe the regulation of apolipoprotein M during the acute phase response. Stimuli that produce systemic inflammation, LPS, zymosan, or turpentine, decrease apolipoprotein M mRNA levels in the liver and kidney. Treatment of Hep3B hepatoma cells with TNF or IL-1 also decreased apolipoprotein M mRNA levels. The decrease in apolipoprotein M mRNA leads to a decrease in apolipoprotein M secretion into the media in Hep3B cells and a decrease in mouse serum following LPS administration. Moreover, in humans with acute bacterial infections or chronic HIV infection, serum apolipoprotein M levels are decreased. Apolipoprotein M is a negative acute response protein that decreases during infection and inflammation. These results are consistent with the finding that infections and inflammatory disorders accompanied by systemic inflammation are associated with an increased risk of atherosclerosis.


Subject(s)
Acute-Phase Reaction/immunology , Apolipoproteins/immunology , Atherosclerosis/immunology , Sepsis/immunology , Vasculitis/immunology , Acute-Phase Reaction/metabolism , Animals , Apolipoproteins/blood , Apolipoproteins/genetics , Apolipoproteins M , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Carcinoma, Hepatocellular , Cell Line, Tumor , Female , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/physiopathology , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/physiopathology , Humans , Lipocalins , Lipopolysaccharides/pharmacology , Liver Neoplasms , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Sepsis/metabolism , Sepsis/physiopathology , Vasculitis/metabolism , Vasculitis/physiopathology
18.
Biochim Biophys Acta ; 1771(9): 1177-85, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17590392

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is a member of PLSCR gene family that has been implicated in multiple cellular processes including movement of phospholipids, gene regulation, immuno-activation, and cell proliferation/apoptosis. In the present study, we identified PLSCR1 as a positive intracellular acute phase protein that is upregulated by LPS in liver, heart, and adipose tissue, but not skeletal muscle. LPS administration resulted in a marked increase in PLSCR1 mRNA and protein levels in the liver. This stimulation occurred rapidly (within 2 h), and was very sensitive to LPS (half-maximal response at 0.1 microg/mouse). Moreover, two other APR-inducers, zymosan and turpentine, also produced significant increases in PLSCR1 mRNA and protein levels, indicating that PLSCR1 was stimulated in a number of models of the APR. To determine signaling pathways by which LPS stimulated PLSCR1, we examined the effect of proinflammatory cytokines in vitro and in vivo. TNFalpha, IL-1beta, and IL-6 all stimulated PLSCR1 in cultured Hep B3 hepatocytes, whereas only TNFalpha stimulated PLSCR1 in cultured 3T3-L1 adipocytes, suggesting cell type-specific effects of cytokines. Furthermore, the LPS-stimulated increase in liver PLSCR1 mRNA was greatly attenuated by 80% in TNFalpha and IL-1beta receptor null mice as compared to wild-type controls. In contrast, PLSCR1 levels in adipose tissue were induced to a similar extent in TNFalpha and IL-1beta receptor null mice and controls. These results indicate that maximal stimulation of PLSCR1 by LPS in liver required TNFalpha and/or IL-1beta, whereas the stimulation of PLSCR1 in adipose tissue is not dependent on TNFalpha and/or IL-1beta. These data provide evidence that PLSCR1 is a positive intracellular acute phase protein with a tissue-specific mechanism for up-regulation.


Subject(s)
Acute-Phase Reaction , Enzyme Induction , Isoenzymes , Phospholipid Transfer Proteins , 3T3-L1 Cells , Adipose Tissue/enzymology , Animals , Female , Genes, Immediate-Early , Humans , Interleukin-1beta/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lipopolysaccharides/immunology , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family , Muscle, Skeletal/enzymology , Myocardium/enzymology , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Solvents , Tumor Necrosis Factor-alpha/metabolism , Turpentine/metabolism , Zymosan/immunology
19.
Metabolism ; 56(2): 267-79, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17224343

ABSTRACT

During the acute phase response, cytokines induce marked alterations in lipid metabolism including an increase in serum triglyceride levels and a decrease in hepatic fatty acid oxidation, in bile acid synthesis, and in high-density lipoprotein levels. Here we demonstrate that tumor necrosis factor (TNF) and interleukin 1 (IL-1), but not IL-6, decrease the expression of retinoid X receptor alpha (RXRalpha), peroxisome proliferator-activated receptor alpha (PPARalpha), PPARgamma, liver X receptor alpha (LXRalpha), and coactivators PPARgamma coactivator 1alpha (PGC-1alpha), PGC-1beta, and steroid receptor coactivator 1 (SRC-1) in Hep3B human hepatoma cells. In addition, treatment of mice with TNF and IL-1 also decreased RXRalpha, PPARalpha, PPARgamma, LXRalpha, and PGC-1alpha messenger RNA (mRNA) levels in the liver. These decreases were accompanied by reduced binding of nuclear extracts to RXR, PPAR, and LXR response elements and decreased luciferase activity driven by PPAR and LXR response elements. In addition, the mRNA levels of proteins regulated by PPARalpha (carnitine palmitoyltransferase 1alpha) and LXR (sterol regulatory element binding protein) were decreased in Hep3B cells treated with TNF or IL-1. Finally, using constructs of the LXRalpha promoter or the PGC-1alpha promoter linked to luciferase, we were able to demonstrate that a decrease in transcription contributes to the reduction in mRNA levels of nuclear hormone receptors and coactivators. Thus, our results suggest that decreased expression of nuclear hormone receptors RXRalpha, PPARalpha, PPARgamma, and LXRalpha, as well as coactivators PGC-1alpha, PGC-1beta, and SRC-1 may contribute to the cytokine-induced alterations in hepatic lipid metabolism during the acute phase response.


Subject(s)
Interleukin-1/pharmacology , Liver/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factors/pharmacology , Acute-Phase Reaction/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Histone Acetyltransferases/metabolism , Liver/drug effects , Liver X Receptors , Mice , Mice, Inbred C57BL , Nuclear Receptor Coactivator 1 , Orphan Nuclear Receptors , PPAR alpha/metabolism , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptor alpha/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transfection
20.
J Lipid Res ; 47(10): 2179-90, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16847310

ABSTRACT

The acute-phase response (APR) leads to alterations in lipid metabolism and type II nuclear hormone receptors, which regulate lipid metabolism, are suppressed, in liver, heart, and kidney. Here, we examine the effect of the APR in adipose tissue. In mice, lipopolysaccharide produces a rapid, marked decrease in mRNA levels of nuclear hormone receptors [peroxisome proliferator-activated receptor gamma (PPARgamma), liver X receptor alpha (LXRalpha) and LXRbeta, thyroid receptor alpha (TRalpha) and TRbeta, and retinoid X receptor alpha (RXRalpha) and RXRbeta] and receptor coactivators [cAMP response element binding protein, steroid receptor coactivator 1 (SRC1) and SRC2, thyroid hormone receptor-associated protein, and peroxisome proliferator-activated receptor gamma co-activator 1alpha (PGC1alpha) and PGC1beta] along with decreased expression of target genes (adipocyte P2, phosphoenolpyruvate carboxykinase, glycerol-3-phosphate acyltransferase, ABCA1, apolipoprotein E, sterol-regulatory element binding protein-1c, glucose transport protein 4 (GLUT4), malic enzyme, and Spot14) involved in triglyceride (TG) and carbohydrate metabolism. We show that key TG synthetic enzymes, 1-acyl-sn-glycerol-3-phosphate acyltransferase-2, monoacylglycerol acyltransferase 1, and diacylglycerol acyltransferase 1, are PPARgamma-regulated genes and that they also decrease in the APR. In 3T3-L1 adipocytes, tumor necrosis factor-alpha (TNF-alpha) significantly decreases PPARgamma, LXRalpha and LXRbeta, RXRalpha and RXRbeta, SRC1 and SRC2, and PGC1alpha and PGC1beta mRNA levels, which are associated with a marked reduction in receptor-regulated genes. Moreover, TNF-alpha significantly reduces PPAR and LXR response element-driven transcription. Thus, the APR suppresses the expression of many nuclear hormone receptors and their coactivators in adipose tissue, which could be a mechanism to coordinately downregulate TG biosynthesis and thereby redirect lipids to other critical organs during the APR.


Subject(s)
Acute-Phase Reaction/metabolism , Adipose Tissue/metabolism , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Acute-Phase Reaction/genetics , Acyltransferases/metabolism , Adipocytes , Animals , Cells, Cultured , DNA-Binding Proteins , Female , Lipopolysaccharides/metabolism , Liver X Receptors , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors , PPAR gamma , RNA, Messenger/analysis , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Zymosan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...