Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell ; 151(1): 138-52, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021221

ABSTRACT

Inflammation and macrophage foam cells are characteristic features of atherosclerotic lesions, but the mechanisms linking cholesterol accumulation to inflammation and LXR-dependent response pathways are poorly understood. To investigate this relationship, we utilized lipidomic and transcriptomic methods to evaluate the effect of diet and LDL receptor genotype on macrophage foam cell formation within the peritoneal cavities of mice. Foam cell formation was associated with significant changes in hundreds of lipid species and unexpected suppression, rather than activation, of inflammatory gene expression. We provide evidence that regulated accumulation of desmosterol underlies many of the homeostatic responses, including activation of LXR target genes, inhibition of SREBP target genes, selective reprogramming of fatty acid metabolism, and suppression of inflammatory-response genes, observed in macrophage foam cells. These observations suggest that macrophage activation in atherosclerotic lesions results from extrinsic, proinflammatory signals generated within the artery wall that suppress homeostatic and anti-inflammatory functions of desmosterol.


Subject(s)
Atherosclerosis/immunology , Cholesterol/biosynthesis , Desmosterol/metabolism , Foam Cells/metabolism , Lipid Metabolism , Transcriptome , Animals , Atherosclerosis/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Fatty Acids/metabolism , Foam Cells/immunology , Gene Knockdown Techniques , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 111(35): 12746-51, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25139986

ABSTRACT

Initiation and resolution of inflammation are considered to be tightly connected processes. Lipoxins (LX) are proresolution lipid mediators that inhibit phlogistic neutrophil recruitment and promote wound-healing macrophage recruitment in humans via potent and specific signaling through the LXA4 receptor (ALX). One model of lipoxin biosynthesis involves sequential metabolism of arachidonic acid by two cell types expressing a combined transcellular metabolon. It is currently unclear how lipoxins are efficiently formed from precursors or if they are directly generated after receptor-mediated inflammatory commitment. Here, we provide evidence for a pathway by which lipoxins are generated in macrophages as a consequence of sequential activation of toll-like receptor 4 (TLR4), a receptor for endotoxin, and P2X7, a purinergic receptor for extracellular ATP. Initial activation of TLR4 results in accumulation of the cyclooxygenase-2-derived lipoxin precursor 15-hydroxyeicosatetraenoic acid (15-HETE) in esterified form within membrane phospholipids, which can be enhanced by aspirin (ASA) treatment. Subsequent activation of P2X7 results in efficient hydrolysis of 15-HETE from membrane phospholipids by group IVA cytosolic phospholipase A2, and its conversion to bioactive lipoxins by 5-lipoxygenase. Our results demonstrate how a single immune cell can store a proresolving lipid precursor and then release it for bioactive maturation and secretion, conceptually similar to the production and inflammasome-dependent maturation of the proinflammatory IL-1 family cytokines. These findings provide evidence for receptor-specific and combinatorial control of pro- and anti-inflammatory eicosanoid biosynthesis, and potential avenues to modulate inflammatory indices without inhibiting downstream eicosanoid pathways.


Subject(s)
Eicosanoids/metabolism , Group IV Phospholipases A2/metabolism , Inflammasomes/metabolism , Macrophages/enzymology , Signal Transduction/immunology , Animals , Aspirin/pharmacology , Celecoxib , Cell Line, Transformed , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/immunology , Group IV Phospholipases A2/immunology , Hydroxyeicosatetraenoic Acids/immunology , Hydroxyeicosatetraenoic Acids/metabolism , Inflammasomes/immunology , Interleukin-1/immunology , Interleukin-1/metabolism , Lipidoses/immunology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2X7/metabolism , Sulfonamides/pharmacology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
3.
J Biol Chem ; 288(50): 35812-23, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24189069

ABSTRACT

25-Hydroxycholesterol (25OHC) is an enzymatically derived oxidation product of cholesterol that modulates lipid metabolism and immunity. 25OHC is synthesized in response to interferons and exerts broad antiviral activity by as yet poorly characterized mechanisms. To gain further insights into the basis for antiviral activity, we evaluated time-dependent responses of the macrophage lipidome and transcriptome to 25OHC treatment. In addition to altering specific aspects of cholesterol and sphingolipid metabolism, we found that 25OHC activates integrated stress response (ISR) genes and reprograms protein translation. Effects of 25OHC on ISR gene expression were independent of liver X receptors and sterol-response element-binding proteins and instead primarily resulted from activation of the GCN2/eIF2α/ATF4 branch of the ISR pathway. These studies reveal that 25OHC activates the integrated stress response, which may contribute to its antiviral activity.


Subject(s)
Hydroxycholesterols/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Oxidative Stress/drug effects , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects , Animals , Bone Marrow Cells/cytology , Cholesterol Esters/metabolism , Gene Expression Profiling , Hydroxycholesterols/metabolism , Liver X Receptors , Macrophages/cytology , Macrophages/virology , Mice , Mice, Inbred C57BL , Muromegalovirus/physiology , Orphan Nuclear Receptors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingolipids/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors
4.
Nature ; 454(7200): 126-30, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18509338

ABSTRACT

With the recent recognition of non-coding RNAs (ncRNAs) flanking many genes, a central issue is to obtain a full understanding of their potential roles in regulated gene transcription programmes, possibly through different mechanisms. Here we show that an RNA-binding protein, TLS (for translocated in liposarcoma), serves as a key transcriptional regulatory sensor of DNA damage signals that, on the basis of its allosteric modulation by RNA, specifically binds to and inhibits CREB-binding protein (CBP) and p300 histone acetyltransferase activities on a repressed gene target, cyclin D1 (CCND1) in human cell lines. Recruitment of TLS to the CCND1 promoter to cause gene-specific repression is directed by single-stranded, low-copy-number ncRNA transcripts tethered to the 5' regulatory regions of CCND1 that are induced in response to DNA damage signals. Our data suggest that signal-induced ncRNAs localized to regulatory regions of transcription units can act cooperatively as selective ligands, recruiting and modulating the activities of distinct classes of RNA-binding co-regulators in response to specific signals, providing an unexpected ncRNA/RNA-binding protein-based strategy to integrate transcriptional programmes.


Subject(s)
Down-Regulation , RNA, Untranslated/metabolism , RNA-Binding Protein FUS/metabolism , Transcription, Genetic , Allosteric Regulation , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/metabolism , Cell Line , Consensus Sequence , Cyclin D1/genetics , DNA Damage , HeLa Cells , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Oligonucleotides/genetics , Promoter Regions, Genetic/genetics , RNA, Untranslated/genetics , RNA-Binding Protein FUS/genetics
5.
PLoS Genet ; 7(12): e1002401, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174696

ABSTRACT

Precise control of the innate immune response is required for resistance to microbial infections and maintenance of normal tissue homeostasis. Because this response involves coordinate regulation of hundreds of genes, it provides a powerful biological system to elucidate the molecular strategies that underlie signal- and time-dependent transitions of gene expression. Comprehensive genome-wide analysis of the epigenetic and transcription status of the TLR4-induced transcriptional program in macrophages suggests that Toll-like receptor 4 (TLR4)-dependent activation of nearly all immediate/early- (I/E) and late-response genes results from a sequential process in which signal-independent factors initially establish basal levels of gene expression that are then amplified by signal-dependent transcription factors. Promoters of I/E genes are distinguished from those of late genes by encoding a distinct set of signal-dependent transcription factor elements, including TATA boxes, which lead to preferential binding of TBP and basal enrichment for RNA polymerase II immediately downstream of transcriptional start sites. Global nuclear run-on (GRO) sequencing and total RNA sequencing further indicates that TLR4 signaling markedly increases the overall rates of both transcriptional initiation and the efficiency of transcriptional elongation of nearly all I/E genes, while RNA splicing is largely unaffected. Collectively, these findings reveal broadly utilized mechanisms underlying temporally distinct patterns of TLR4-dependent gene activation required for homeostasis and effective immune responses.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Immunity, Innate/genetics , Inflammation/genetics , Macrophages/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Epigenesis, Genetic/genetics , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Homeostasis , Humans , Immunity, Innate/immunology , Inflammation/immunology , Mice , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , TATA Box/genetics , Transcription Factors , Transcriptional Activation/genetics , Transcriptional Activation/immunology
6.
Am J Pathol ; 179(1): 189-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703401

ABSTRACT

CD45(+) and collagen I-positive (Col(+)) fibrocytes are implicated in fibrogenesis in skin, lungs, and kidneys. Fibrocyte migration in response to liver injury was investigated using bone marrow (BM) from chimeric mice expressing luciferase (Col-Luc→wt) or green fluorescent protein (Col-GFP→wt) under control of the α1(I) collagen promoter and enhancer, respectively. Monitored by luciferase expression, recruitment of fibrocytes was detected in CCl(4)-damaged liver and in spleen. Migration of CD45(+)Col(+) fibrocytes was regulated by chemokine receptors CCR2 and CCR1, as demonstrated, respectively, by 50% and 25% inhibition of fibrocyte migration in Col-Luc(CCR2-/-)→wt and Col-Luc(CCR1-/-)→wt mice. In addition to CCR2 and CCR1, egress of BM CD45(+)Col(+) cells was regulated by transforming growth factor-ß1 (TGF-ß1) and liposaccharide in vitro and in vivo, which suggests that release of TGF-ß1 and increased intestinal permeability have important roles in fibrocyte trafficking. In the injured liver, fibrocytes gave rise to (myo)fibroblasts. In addition, a BM population of CD45(+)Col(+) cells capable of differentiation into fibrocytes in culture was identified. Egress of CD45(+)Col(+) cells from BM was detected in the absence of injury or stress in aged mice but not in young mice. Development of liver fibrosis was also increased in aged mice and correlated with high numbers of liver fibrocytes. In conclusion, in response to liver injury, fibrocytes migrate from BM to the liver. Their migration is regulated by CCR2 and CCR1 but is compromised with age.


Subject(s)
Cell Differentiation , Cell Movement , Fibroblasts/cytology , Liver Cirrhosis/pathology , Liver/injuries , Liver/pathology , Animals , Biomarkers/metabolism , Blotting, Western , Bone Marrow/metabolism , Cell Adhesion , Cells, Cultured , Collagen/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoenzyme Techniques , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Liver/metabolism , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/cytology , Myofibroblasts/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Receptors, CCR1/physiology , Receptors, CCR2/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spleen/injuries , Spleen/metabolism , Spleen/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
7.
J Biol Chem ; 285(49): 38568-79, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20876532

ABSTRACT

Activation of RAW264.7 cells with a lipopolysaccharide specific for the TLR4 receptor, Kdo(2)-lipid A (KLA), causes a large increase in cellular sphingolipids, from 1.5 to 2.6 × 10(9) molecules per cell in 24 h, based on the sum of subspecies analyzed by "lipidomic" mass spectrometry. Thus, this study asked the following question. What is the cause of this increase and is there a cell function connected with it? The sphingolipids arise primarily from de novo biosynthesis based on [U-(13)C]palmitate labeling, inhibition by ISP1 (myriocin), and an apparent induction of many steps of the pathway (according to the distribution of metabolites and microarray analysis), with the exception of ceramide, which is also produced from pre-existing sources. Nonetheless, the activated RAW264.7 cells have a higher number of sphingolipids per cell because KLA inhibits cell division; thus, the cells are larger and contain increased numbers of membrane vacuoles termed autophagosomes, which were detected by the protein marker GFP-LC3. Indeed, de novo biosynthesis of sphingolipids performs an essential structural and/or signaling function in autophagy because autophagosome formation was eliminated by ISP1 in KLA-stimulated RAW264.7 cells (and mutation of serine palmitoyltransferase in CHO-LYB cells); furthermore, an anti-ceramide antibody co-localizes with autophagosomes in activated RAW264.7 cells versus the Golgi in unstimulated or ISP1-inhibited cells. These findings establish that KLA induces profound changes in sphingolipid metabolism and content in this macrophage-like cell line, apparently to produce sphingolipids that are necessary for formation of autophagosomes, which are thought to play important roles in the mechanisms of innate immunity.


Subject(s)
Autophagy/drug effects , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Sphingolipids/biosynthesis , Toll-Like Receptor 4/agonists , Animals , Autophagy/genetics , Autophagy/immunology , CHO Cells , Cell Division/drug effects , Cell Division/genetics , Cell Division/immunology , Cell Line , Cricetinae , Cricetulus , Golgi Apparatus/genetics , Golgi Apparatus/immunology , Golgi Apparatus/metabolism , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mutation , Phagosomes/immunology , Phagosomes/metabolism , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/immunology , Serine C-Palmitoyltransferase/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Sphingolipids/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
8.
J Biol Chem ; 285(51): 39976-85, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20923771

ABSTRACT

We report the lipidomic response of the murine macrophage RAW cell line to Kdo(2)-lipid A, the active component of an inflammatory lipopolysaccharide functioning as a selective TLR4 agonist and compactin, a statin inhibitor of cholesterol biosynthesis. Analyses of lipid molecular species by dynamic quantitative mass spectrometry and concomitant transcriptomic measurements define the lipidome and demonstrate immediate responses in fatty acid metabolism represented by increases in eicosanoid synthesis and delayed responses characterized by sphingolipid and sterol biosynthesis. Lipid remodeling of glycerolipids, glycerophospholipids, and prenols also take place, indicating that activation of the innate immune system by inflammatory mediators leads to alterations in a majority of mammalian lipid categories, including unanticipated effects of a statin drug. Our studies provide a systems-level view of lipid metabolism and reveal significant connections between lipid and cell signaling and biochemical pathways that contribute to innate immune responses and to pharmacological perturbations.


Subject(s)
Immunity, Innate , Inflammation Mediators/metabolism , Lipid Metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Animals , Cell Line , Immunity, Innate/drug effects , Immunity, Innate/physiology , Inflammation Mediators/immunology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Macrophages/immunology , Mice , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
9.
J Clin Invest ; 117(6): 1658-69, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17525798

ABSTRACT

PPAR gamma is required for fat cell development and is the molecular target of antidiabetic thiazolidinediones (TZDs), which exert insulin-sensitizing effects in adipose tissue, skeletal muscle, and liver. Unexpectedly, we found that inactivation of PPAR gamma in macrophages results in the development of significant glucose intolerance plus skeletal muscle and hepatic insulin resistance in lean mice fed a normal diet. This phenotype was associated with increased expression of inflammatory markers and impaired insulin signaling in adipose tissue, muscle, and liver. PPAR gamma-deficient macrophages secreted elevated levels of factors that impair insulin responsiveness in muscle cells in a manner that was enhanced by exposure to FFAs. Consistent with this, the relative degree of insulin resistance became more severe in mice lacking macrophage PPAR gamma following high-fat feeding, and these mice were only partially responsive to TZD treatment. These findings reveal an essential role of PPAR gamma in macrophages for the maintenance of whole-body insulin action and in mediating the antidiabetic actions of TZDs.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Liver/metabolism , Muscle, Skeletal/metabolism , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Base Sequence , DNA Primers/genetics , Gene Expression Profiling , Liver/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/drug effects , PPAR gamma/deficiency , PPAR gamma/genetics , Promoter Regions, Genetic
10.
Diabetes ; 51(6): 1907-12, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12031980

ABSTRACT

It is well described that excessive lipid metabolism can cause insulin resistance in both animals and humans, and this has been implicated as a causative factor in the development of insulin resistance and type 2 diabetes in humans. Recently, we have shown that intravenous lipid emulsion (liposyn) infusion during a 120-min euglycemic-hyperinsulinemic clamp led to significant reductions in insulin action and fatty acid translocase (FAT/CD36) skeletal muscle protein expression. After reviewing the literature, it became evident that essentially all past studies, including our own, were conducted in male animals. Therefore, to determine whether there were sex determinants of fat-induced insulin resistance, we assessed the impact of free fatty acid (FFA) elevation on insulin action in female rats. Here, we report that a fourfold elevation in plasma FFA concentration induced a 40% reduction in the insulin-stimulated glucose disposal rate, a 30% decline in insulin-stimulated skeletal muscle insulin substrate receptor-1 (IRS-1) phosphorylation, a 48% decrease in IRS-1-associated phosphatidylinositol (PI) 3-kinase activity, and a 50% reduction in muscle FAT/CD36 protein expression in male rats. In striking contrast, we found no effect of FFA elevation to cause insulin resistance, changes in IRS-1/PI 3-kinase, or FAT/CD36 protein levels in female animals. Our findings indicate that female animals are protected from lipid-induced reductions in insulin action.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Insulin Resistance , Animals , Blood Glucose/metabolism , Emulsions , Fat Emulsions, Intravenous/administration & dosage , Fatty Acids, Nonesterified/blood , Female , Glucose Clamp Technique , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Kinetics , Lecithins , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Rats , Rats, Wistar , Safflower Oil , Sex Characteristics , Soybean Oil
11.
J Leukoc Biol ; 90(3): 563-74, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21653236

ABSTRACT

Eicosanoid metabolism differs in profile and quantity between macrophages of different tissue origin and method of elicitation, as well as between primary and immortalized macrophages after activation with inflammatory stimuli. Using a lipidomic approach, we comprehensively analyzed the eicosanoids made by murine RPMs, TGEMs, BMDM, and the macrophage-like cell line RAW after stimulation with the TLR-4-specific agonist KLA. Direct correlation among total COX metabolites, COX side-products (11-HETE, 15-HETE), COX-2 mRNA, and protein at 8 h was found when comparing each cell type. Comprehensive qPCR analysis was used to compare relative transcript levels between the terminal prostanoid synthases themselves as well as between each cell type. Levels of PGE(2), PGD(2), and TxB(2) generally correlated with enzyme transcript expression of PGES, PGDS, and TBXS, providing evidence of comparable enzyme activities. PGIS transcript was expressed only in RPM and TGEM macrophages and at an exceptionally low level, despite high metabolite production compared with other synthases. Presence of PGIS in RPM and TGEM also lowered the production of PGE(2) versus PGD(2) by approximately tenfold relative to BMDM and RAW cells, which lacked this enzyme. Our results demonstrate that delayed PG production depends on the maximal level of COX-2 expression in different macrophages after TLR-4 stimulation. Also, the same enzymes in each cell largely dictate the profile of eicosanoids produced depending on the ratios of expression between them, with the exception of PGIS, which appears to have much greater synthetic capacity and competes selectively with mPGES-1.


Subject(s)
Eicosanoids/metabolism , Lipids/analysis , Macrophages/drug effects , Macrophages/metabolism , Toll-Like Receptor 4/metabolism , Animals , Blotting, Western , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Prostaglandin-E Synthases , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
12.
J Mol Med (Berl) ; 89(10): 997-1013, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21499735

ABSTRACT

Bone marrow (BM)-derived fibrocytes are a population of CD45(+) and collagen Type I-expressing cells that migrate to the spleen and to target injured organs, such as skin, lungs, kidneys, and liver. While CD45(+)Col(+) fibrocytes contribute to collagen deposition at the site of injury, the role of CD45(+)Col(+) cells in spleen has not been elucidated. Here, we demonstrate that hepatotoxic injury (CCl(4)), TGF-ß1, lipopolysaccharide, or infection with Listeria monocytogenes induce rapid recruitment of CD45(+)Col(+) fibrocyte-like cells to the spleen. These cells have a gene expression pattern that includes antimicrobial factors (myleoperoxidase, cathelicidin, and defensins) and MHC II at higher levels than found on quiescent or activated macrophages. The immune functions of these splenic CD45(+)Col(+) fibrocyte-like cells include entrapment of bacteria into extracellular DNA-based structures containing cathelicidin and presentation of antigens to naïve CD8(+) T cells to induce their proliferation. Stimulation of these splenic fibrocyte-like cells with granulocyte macrophage-colony stimulating factor or macrophage-colony stimulating factor induces downregulation of collagen expression and terminal differentiation into the dendritic cells or macrophage. Thus, splenic CD45(+)Col(+) cells are a population of rapidly mobilized BM-derived fibrocyte-like cells that respond to inflammation or infection to participate in innate and adaptive immune responses.


Subject(s)
Adaptive Immunity/immunology , Fibroblasts/pathology , Immunity, Innate/immunology , Spleen/immunology , Spleen/pathology , Acute Disease , Adaptive Immunity/drug effects , Animals , Antigen Presentation/immunology , Carbon Tetrachloride , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Extracellular Space/drug effects , Extracellular Space/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Expression Profiling , Immunity, Innate/drug effects , Immunity, Innate/genetics , Leukocyte Common Antigens/metabolism , Lipopolysaccharides/pharmacology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/pathology , Liver/immunology , Liver/microbiology , Liver/pathology , Liver/ultrastructure , Liver Cirrhosis , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Phagocytosis/drug effects , Phenotype , Spleen/microbiology , Spleen/ultrastructure , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transforming Growth Factor beta1/pharmacology
13.
J Biol Chem ; 282(48): 35279-92, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17916553

ABSTRACT

Obesity and type 2 diabetes are characterized by decreased insulin sensitivity, elevated concentrations of free fatty acids (FFAs), and increased macrophage infiltration in adipose tissue (AT). Here, we show that FFAs can cause activation of RAW264.7 cells primarily via the JNK signaling cascade and that TLR2 and TLR4 are upstream of JNK and help transduce FFA proinflammatory signals. We also demonstrate that F4/80(+)CD11b(+)CD11c(+) bone marrow-derived dendritic cells (BMDCs) have heightened proinflammatory activity compared with F4/80(+)CD11b(+)CD11c(-) bone marrow-derived macrophages and that the proinflammatory activity and JNK phosphorylation of BMDCs, but not bone marrow-derived macrophages, was further increased by FFA treatment. F4/80(+)CD11b(+)CD11c(+) cells were found in AT, and the proportion and number of these cells in AT is increased in ob/ob mice and by feeding wild type mice a high fat diet for 1 and 12 weeks. AT F4/80(+)CD11b(+)CD11c(+) cells express increased inflammatory markers compared with F4/80(+)CD11b(+)CD11c(-) cells, and FFA treatment increased inflammatory responses in these cells. In addition, we found that CD11c expression is increased in skeletal muscle of high fat diet-fed mice and that conditioned medium from FFA-treated wild type BMDCs, but not TLR2/4 DKO BMDCs, can induce insulin resistance in L6 myotubes. Together our results show that FFAs can activate CD11c(+) myeloid proinflammatory cells via TLR2/4 and JNK signaling pathways, thereby promoting inflammation and subsequent cellular insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids, Nonesterified/metabolism , MAP Kinase Kinase 4/metabolism , Macrophages/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Bone Marrow Cells/metabolism , CD11b Antigen/biosynthesis , CD11c Antigen/biosynthesis , Inflammation , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Models, Biological , Signal Transduction
14.
J Lipid Res ; 47(5): 1097-111, 2006 May.
Article in English | MEDLINE | ID: mdl-16479018

ABSTRACT

The LIPID MAPS Consortium (www.lipidmaps.org) is developing comprehensive procedures for identifying all lipids of the macrophage, following activation by endotoxin. The goal is to quantify temporal and spatial changes in lipids that occur with cellular metabolism and to develop bioinformatic approaches that establish dynamic lipid networks. To achieve these aims, an endotoxin of the highest possible analytical specification is crucial. We now report a large-scale preparation of 3-deoxy-D-manno-octulosonic acid (Kdo)(2)-Lipid A, a nearly homogeneous Re lipopolysaccharide (LPS) sub-structure with endotoxin activity equal to LPS. Kdo(2)-Lipid A was extracted from 2 kg cell paste of a heptose-deficient Escherichia coli mutant. It was purified by chromatography on silica, DEAE-cellulose, and C18 reverse-phase resin. Structure and purity were evaluated by electrospray ionization/mass spectrometry, liquid chromatography/mass spectrometry and (1)H-NMR. Its bioactivity was compared with LPS in RAW 264.7 cells and bone marrow macrophages from wild-type and toll-like receptor 4 (TLR-4)-deficient mice. Cytokine and eicosanoid production, in conjunction with gene expression profiling, were employed as readouts. Kdo(2)-Lipid A is comparable to LPS by these criteria. Its activity is reduced by >10(3) in cells from TLR-4-deficient mice. The purity of Kdo(2)-Lipid A should facilitate structural analysis of complexes with receptors like TLR-4/MD2.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Toll-Like Receptor 4/physiology , Animals , Chromatography, High Pressure Liquid/methods , Escherichia coli/metabolism , Lipopolysaccharides/isolation & purification , Mice , Nuclear Magnetic Resonance, Biomolecular , Prostaglandin D2/metabolism , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL