Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Immunity ; 41(1): 14-20, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25035950

ABSTRACT

Description of macrophage activation is currently contentious and confusing. Like the biblical Tower of Babel, macrophage activation encompasses a panoply of descriptors used in different ways. The lack of consensus on how to define macrophage activation in experiments in vitro and in vivo impedes progress in multiple ways, including the fact that many researchers still consider there to be only two types of activated macrophages, often termed M1 and M2. Here, we describe a set of standards encompassing three principles-the source of macrophages, definition of the activators, and a consensus collection of markers to describe macrophage activation-with the goal of unifying experimental standards for diverse experimental scenarios. Collectively, we propose a common framework for macrophage-activation nomenclature.


Subject(s)
Macrophage Activation/immunology , Macrophages/immunology , Terminology as Topic , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Guidelines as Topic , Humans , Macrophage Colony-Stimulating Factor/immunology , Mice , Research
2.
J Immunol ; 200(10): 3407-3419, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29626089

ABSTRACT

Obesity is associated with elevated levels of free fatty acids (FAs) and proinflammatory CD11c+ macrophages. However, whether and how free FAs contribute to CD11c+ macrophage differentiation and proinflammatory functions remain unclear. Here we report that dietary saturated FAs, but not unsaturated FAs, promoted the differentiation and function of CD11c+ macrophages. Specifically, we demonstrated that stearic acid (SA) significantly induced CD11c expression in monocytes through activation of the nuclear retinoid acid receptor. More importantly, cytosolic expression of epidermal FA binding protein (E-FABP) in monocytes/macrophages was shown to be critical to the mediation of the SA-induced effect. Depletion of E-FABP not only inhibited SA-induced CD11c upregulation in macrophages in vitro but also abrogated high-saturated-fat diet-induced skin lesions in obese mouse models in vivo. Altogether, our data demonstrate a novel mechanism by which saturated FAs promote obesity-associated inflammation through inducing E-FABP/retinoid acid receptor-mediated differentiation of CD11c+ macrophages.


Subject(s)
CD11c Antigen/metabolism , Fatty Acid-Binding Proteins/metabolism , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Stearic Acids/pharmacology , Animals , Fatty Acids/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Monocytes/drug effects , Monocytes/metabolism , Obesity/metabolism , Up-Regulation/drug effects
3.
J Immunol ; 198(2): 798-807, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27920274

ABSTRACT

Macrophages play a critical role in obesity-associated chronic inflammation and disorders. However, the molecular mechanisms underlying the response of macrophages to elevated fatty acids (FAs) and their contribution to metabolic inflammation in obesity remain to be fully elucidated. In this article, we report a new mechanism by which dietary FAs, in particular, saturated FAs (sFAs), are able to directly trigger macrophage cell death. We demonstrated that excess sFAs, but not unsaturated FAs, induced the production of cytotoxic ceramides (Cers) in macrophage cell lines. Most importantly, expression of adipose FA binding protein (A-FABP) in macrophages facilitated metabolism of excess sFAs for Cer synthesis. Inhibition or deficiency of A-FABP in macrophage cell lines decreased sFA-induced Cer production, thereby resulting in reduced cell death. Furthermore, we validated the role of A-FABP in promoting sFA-induced macrophage cell death with primary bone marrow-derived macrophages and high-fat diet-induced obese mice. Altogether, our data reveal that excess dietary sFAs may serve as direct triggers in induction of Cer production and macrophage cell death through elevated expression of A-FABP, thus establishing A-FABP as a new molecular sensor in triggering macrophage-associated sterile inflammation in obesity.


Subject(s)
Ceramides/biosynthesis , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/adverse effects , Macrophages/pathology , Animals , Blotting, Western , Cell Death , Diet, High-Fat , Flow Cytometry , Gene Knockdown Techniques , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Obesity/metabolism , Real-Time Polymerase Chain Reaction
4.
Mol Ther ; 25(7): 1641-1654, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28274798

ABSTRACT

The intestinal immune system is continuously exposed to massive amounts of nanoparticles derived from food. Whether nanoparticles from plants we eat daily have a role in maintaining intestinal immune homeostasis is poorly defined. Here, we present evidence supporting our hypothesis that edible nanoparticles regulate intestinal immune homeostasis by targeting dendritic cells (DCs). Using three mouse colitis models, our data show that orally given nanoparticles isolated from broccoli extracts protect mice against colitis. Broccoli-derived nanoparticle (BDN)-mediated activation of adenosine monophosphate-activated protein kinase (AMPK) in DCs plays a role in not only prevention of DC activation but also induction of tolerant DCs. Adoptively transferring DCs pre-pulsed with total BDN lipids, but not sulforaphane (SFN)-depleted BDN lipids, prevented DSS-induced colitis in C57BL/6 (B6) mice, supporting the role of BDN SFN in the induction of DC tolerance. Adoptively transferring AMPK+/+, but not AMPK-/-, DCs pre-pulsed with SFN prevented DSS-induced colitis in B6 mice, further supporting the DC AMPK role in SFN-mediated prevention of DSS-induced colitis. This finding could open new preventive or therapeutic avenues to address intestinal-related inflammatory diseases via activating AMPK.


Subject(s)
AMP-Activated Protein Kinases/genetics , Anti-Inflammatory Agents/pharmacology , Brassica/chemistry , Colitis, Ulcerative/prevention & control , Dendritic Cells/drug effects , Nanoparticles/chemistry , AMP-Activated Protein Kinases/metabolism , Administration, Oral , Adoptive Transfer , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Disease Models, Animal , Enzyme Activation/drug effects , Gene Expression , Humans , Immune Tolerance , Isothiocyanates/chemistry , Lipids/isolation & purification , Lipids/pharmacology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Plant Extracts/chemistry , Sodium Dodecyl Sulfate , Sulfoxides
5.
J Immunol ; 194(2): 584-94, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25512602

ABSTRACT

AMP-activated protein kinase (AMPK) is a conserved serine/threonine kinase with a critical function in the regulation of metabolic pathways in eukaryotic cells. Recently, AMPK has been shown to play an additional role as a regulator of inflammatory activity in leukocytes. Treatment of macrophages with chemical AMPK activators, or forced expression of a constitutively active form of AMPK, results in polarization to an anti-inflammatory phenotype. In addition, we reported previously that stimulation of macrophages with anti-inflammatory cytokines such as IL-10, IL-4, and TGF-ß results in rapid activation of AMPK, suggesting that AMPK contributes to the suppressive function of these cytokines. In this study, we investigated the role of AMPK in IL-10-induced gene expression and anti-inflammatory function. IL-10-stimulated wild-type macrophages displayed rapid activation of PI3K and its downstream targets Akt and mammalian target of rapamycin complex (mTORC1), an effect that was not seen in macrophages generated from AMPKα1-deficient mice. AMPK activation was not impacted by treatment with either the PI3K inhibitor LY294002 or the JAK inhibitor CP-690550, suggesting that IL-10-mediated activation of AMPK is independent of PI3K and JAK activity. IL-10 induced phosphorylation of both Tyr(705) and Ser(727) residues of STAT3 in an AMPKα1-dependent manner, and these phosphorylation events were blocked by inhibition of Ca(2+)/calmodulin-dependent protein kinase kinase ß, an upstream activator of AMPK, and by the mTORC1 inhibitor rapamycin, respectively. The impaired STAT3 phosphorylation in response to IL-10 observed in AMPKα1-deficient macrophages was accompanied by reduced suppressor of cytokine signaling 3 expression and an inadequacy of IL-10 to suppress LPS-induced proinflammatory cytokine production. Overall, our data demonstrate that AMPKα1 is required for IL-10 activation of the PI3K/Akt/mTORC1 and STAT3-mediated anti-inflammatory pathways regulating macrophage functional polarization.


Subject(s)
AMP-Activated Protein Kinases/immunology , Interleukin-10/immunology , Macrophages/immunology , Signal Transduction/immunology , AMP-Activated Protein Kinases/genetics , Animals , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-4/genetics , Interleukin-4/immunology , Lipopolysaccharides/toxicity , Macrophages/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
6.
BMC Immunol ; 16: 28, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25962726

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease in which dysregulated immune cells attack myelin in the central nervous system (CNS), leading to irreversible neuronal degeneration. Our previous studies have demonstrated that epidermal fatty acid binding protein (E-FABP), widely expressed in immune cells, in particular in dendritic cells (DCs) and T lymphocytes, fuels the overactive immune responses in the mouse model of experimental autoimmune encephalomyelitis (EAE). METHODS: In the present study, we conducted an intensive computational docking analysis to identify novel E-FABP inhibitors for regulation of immune cell functions and for treatment of EAE. RESULTS: We demonstrate that compound [2-(4-acetylphenoxy)-9,10-dimethoxy-6,7-dihydropyrimido[6,1-a]isoquinolin-4-one; designated as EI-03] bound to the lipid binding pocket of E-FABP and enhanced the expression of peroxisome proliferator-activating receptor (PPAR) γ. Further in vitro experiments showed that EI-03 regulated DC functions by inhibition of TNFα production while promoting IL-10 secretion. Moreover, EI-03 treatment counterregulated T cell balance by decreasing effector T cell differentiation (e.g. Th17, Th1) while increasing regulatory T cell development. Most importantly, mice treated with this newly identified compound exhibited reduced clinical symptoms of EAE in mouse models. CONCLUSIONS: Taken together, we have identified a new compound which displays a potential therapeutic benefit for treatment of MS by targeting E-FABP.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Fatty Acid-Binding Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Animals , Cell Differentiation , Cytokines/biosynthesis , Dendritic Cells/metabolism , Drug Evaluation, Preclinical , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism
7.
J Immunol ; 191(3): 1164-74, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23797672

ABSTRACT

The role of JAK-3 in TLR-mediated innate immune responses is poorly understood, although the suppressive function of JAK3 inhibition in adaptive immune response has been well studied. In this study, we found that JAK3 inhibition enhanced TLR-mediated immune responses by differentially regulating pro- and anti- inflammatory cytokine production in innate immune cells. Specifically, JAK3 inhibition by pharmacological inhibitors or specific small interfering RNA or JAK3 gene knockout resulted in an increase in TLR-mediated production of proinflammatory cytokines while concurrently decreasing the production of IL-10. Inhibition of JAK3 suppressed phosphorylation of PI3K downstream effectors including Akt, mammalian target of rapamycin complex 1, glycogen synthase kinase 3ß (GSK3ß), and CREB. Constitutive activation of Akt or inhibition of GSK3ß abrogated the capability of JAK3 inhibition to enhance proinflammatory cytokines and suppress IL-10 production. In contrast, inhibition of PI3K enhanced this regulatory ability of JAK3 in LPS-stimulated monocytes. At the transcriptional level, JAK3 knockout lead to the increased phosphorylation of STATs that could be attenuated by neutralization of de novo inflammatory cytokines. JAK3 inhibition exhibited a GSK3 activity-dependent ability to enhance phosphorylation levels and DNA binding of NF-κB p65. Moreover, JAK3 inhibition correlated with an increased CD4(+) T cell response. Additionally, higher neutrophil infiltration, IL-17 expression, and intestinal epithelium erosion were observed in JAK3 knockout mice. These findings demonstrate the negative regulatory function of JAK3 and elucidate the signaling pathway by which JAK3 differentially regulates TLR-mediated inflammatory cytokine production in innate immune cells.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Inflammation/immunology , Janus Kinase 3/metabolism , Toll-Like Receptors/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CREB-Binding Protein/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Immunity, Innate/immunology , Inflammation/genetics , Interleukin-10/biosynthesis , Interleukin-17/biosynthesis , Intestinal Mucosa/immunology , Intestines/immunology , Janus Kinase 3/genetics , Lipopolysaccharides , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Monocytes/drug effects , Monocytes/immunology , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Neutrophils/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism
8.
Biochim Biophys Acta ; 1831(7): 1199-207, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24046860

ABSTRACT

Obesity results in increased macrophage recruitment to adipose tissue that promotes a chronic low-grade inflammatory state linked to increased fatty acid efflux from adipocytes. Activated macrophages produce a variety of pro-inflammatory lipids such as leukotriene C4 (LTC4) and 5-, 12-, and 15-hydroxyeicosatetraenoic acid (HETE) suggesting the hypothesis that fatty acids may stimulate eicosanoid synthesis. To assess if eicosanoid production increases with obesity, adipose tissue of leptin deficient ob/ob mice was analyzed. In ob/ob mice, LTC4 and 12-HETE levels increased in the visceral (but not subcutaneous) adipose depot while the 5-HETE levels decreased and 15-HETE abundance was unchanged. Since macrophages produce the majority of inflammatory molecules in adipose tissue, treatment of RAW264.7 or primary peritoneal macrophages with free fatty acids led to increased secretion of LTC4 and 5-HETE, but not 12- or 15-HETE. Fatty acid binding proteins (FABPs) facilitate the intracellular trafficking of fatty acids and other hydrophobic ligands and in vitro stabilize the LTC4 precursor leukotriene A4 (LTA4) from non-enzymatic hydrolysis. Consistent with a role for FABPs in LTC4 synthesis, treatment of macrophages with HTS01037, a specific FABP inhibitor, resulted in a marked decrease in both basal and fatty acid-stimulated LTC4 secretion but no change in 5-HETE production or 5-lipoxygenase expression. These results indicate that the products of adipocyte lipolysis may stimulate the 5-lipoxygenase pathway leading to FABP-dependent production of LTC4 and contribute to the insulin resistant state.


Subject(s)
Adipose Tissue/immunology , Fatty Acid-Binding Proteins/immunology , Fatty Acids/immunology , Leukotriene C4/immunology , Macrophages/immunology , Obesity/immunology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/analysis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/immunology , Adipose Tissue/pathology , Animals , Cell Line , Cells, Cultured , Fatty Acids/analysis , Female , Hydroxyeicosatetraenoic Acids/analysis , Hydroxyeicosatetraenoic Acids/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology
9.
Biochim Biophys Acta ; 1831(7): 1199-207, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23583845

ABSTRACT

Obesity results in increased macrophage recruitment to adipose tissue that promotes a chronic low-grade inflammatory state linked to increased fatty acid efflux from adipocytes. Activated macrophages produce a variety of pro-inflammatory lipids such as leukotriene C4 (LTC4) and 5-, 12-, and 15-hydroxyeicosatetraenoic acid (HETE) suggesting the hypothesis that fatty acids may stimulate eicosanoid synthesis. To assess if eicosanoid production increases with obesity, adipose tissue of leptin deficient ob/ob mice was analyzed. In ob/ob mice, LTC4 and 12-HETE levels increased in the visceral (but not subcutaneous) adipose depot while the 5-HETE levels decreased and 15-HETE abundance was unchanged. Since macrophages produce the majority of inflammatory molecules in adipose tissue, treatment of RAW264.7 or primary peritoneal macrophages with free fatty acids led to increased secretion of LTC4 and 5-HETE, but not 12- or 15-HETE. Fatty acid binding proteins (FABPs) facilitate the intracellular trafficking of fatty acids and other hydrophobic ligands and in vitro stabilize the LTC4 precursor leukotriene A4 (LTA4) from non-enzymatic hydrolysis. Consistent with a role for FABPs in LTC4 synthesis, treatment of macrophages with HTS01037, a specific FABP inhibitor, resulted in a marked decrease in both basal and fatty acid-stimulated LTC4 secretion but no change in 5-HETE production or 5-lipoxygenase expression. These results indicate that the products of adipocyte lipolysis may stimulate the 5-lipoxygenase pathway leading to FABP-dependent production of LTC4 and contribute to the insulin resistant state.

10.
J Immunol ; 186(7): 3858-65, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339365

ABSTRACT

We previously showed that monophosphoryl lipid A (MLA) activates TLR4 in dendritic cells (DCs) in a Toll/IL-1R domain-containing adaptor inducing IFN-ß (TRIF)-biased manner: MLA produced from Salmonella minnesota Re595 induced signaling events and expression of gene products that were primarily TRIF dependent, whereas MyD88-dependent signaling was impaired. Moreover, when tested in TRIF-intact/MyD88-deficient DCs, synthetic MLA of the Escherichia coli chemotype (sMLA) showed the same activity as its diphosphoryl, inflammatory counterpart (synthetic diphosphoryl lipid A), indicating that TRIF-mediated signaling is fully induced by sMLA. Unexpectedly, we found that the transcript level of one proinflammatory cytokine was increased in sMLA-treated cells by MyD88 deficiency to the higher level induced by synthetic diphosphoryl lipid A, which suggested MyD88 may paradoxically help restrain proinflammatory signaling by TRIF-biased sMLA. In this article, we demonstrate that sMLA induces MyD88 recruitment to TLR4 and activates the anti-inflammatory lipid phosphatase SHIP1 in an MyD88-dependent manner. At the same time, MyD88-dependent signaling activity at the level of IL-1R-associated kinase 1 is markedly reduced. Increased SHIP1 activity is associated with reductions in sMLA-induced IκB kinase α/ß and IFN regulatory factor 3 activation and with restrained expression of their downstream targets, endothelin-1 and IFN-ß, respectively. Results of this study identify a pattern that is desirable in the context of vaccine adjuvant design: TRIF-biased sMLA can stimulate partial MyD88 activity, with MyD88-dependent SHIP1 helping to reduce proinflammatory signaling in DCs.


Subject(s)
Adjuvants, Immunologic/physiology , Dendritic Cells/immunology , Inflammation Mediators/physiology , Lipid A/analogs & derivatives , Myeloid Differentiation Factor 88/physiology , Phosphoric Monoester Hydrolases/physiology , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , Bone Marrow Cells/pathology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Escherichia coli/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inositol Polyphosphate 5-Phosphatases , Lipid A/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Salmonella/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/physiology
11.
Semin Immunol ; 21(5): 257-64, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19540774

ABSTRACT

Macrophages reside in all tissues as resident populations and as immigrants recruited in response to tissue injury, inflammation or pathogen invasion. Under normal conditions, macrophages contribute to tissue homeostasis and provide innate immune surveillance. Both macrophages and their progenitors, bone marrow-derived monocytes, constitutively express the tumor necrosis factor receptor superfamily member, CD40, and are capable of a robust response to CD40 ligation resulting in the induction or enhancement of expression of genes with a predominantly pro-inflammatory function. CD40 signaling in macrophages in the context of host responses to pathogens plays a crucial role in host defense. However, macrophage responses to CD40 ligation in the context of autoimmune and cardiovascular disease contribute to disease pathogenesis. In this review, we discuss the role of CD40 in both protective and destructive processes, including the signaling pathways engaged and the factors capable of modulating CD40 signal transduction.


Subject(s)
CD40 Antigens/immunology , Inflammation/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Humans
12.
J Biol Chem ; 286(52): 44295-305, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22045807

ABSTRACT

Activation of the PI3K pathway plays a pivotal role in regulating the inflammatory response. The loss of mTORC2 has been shown to abrogate the activation of Akt, a critical downstream component of PI3K signaling. However, the biological importance of mTORC2 in innate immunity is currently unknown. Here we demonstrate that rictor, a key component of mTORC2, plays a critical role in controlling the innate inflammatory response via its ability to regulate FoxO1. Upon LPS stimulation, both rictor-deficient mouse embryonic fibroblasts (MEFs) and rictor knockdown dendritic cells exhibited a hyperinflammatory phenotype. The hyperinflammatory phenotype was due to a defective Akt signaling axis, because both rictor-deficient MEFs and rictor knockdown dendritic cells exhibited attenuated Akt phosphorylation and kinase activity. Analysis of downstream Akt targets revealed that phosphorylation of FoxO1 was impaired in rictor-deficient cells, resulting in elevated nuclear FoxO1 levels and diminished nuclear export of FoxO1 upon LPS stimulation. Knockdown of FoxO1 attenuated the hyperinflammatory phenotype exhibited by rictor-deficient MEFs. Moreover, FoxO1 deletion in dendritic cells attenuated the capacity of LPS to induce inflammatory cytokine expression. These findings identify a novel signaling pathway by which mTORC2 regulates the TLR-mediated inflammatory response through its ability to regulate FoxO1.


Subject(s)
Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Trans-Activators/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
13.
Cell Metab ; 3(4): 247-56, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581002

ABSTRACT

Inflammatory processes play an important role in the pathogenesis of vascular diseases, and insulin-resistant diabetes mellitus type 2 represents an important risk factor for the development of atherosclerosis. To directly address the role of insulin resistance in myeloid lineage cells in the development of atherosclerosis, we have created mice with myeloid lineage-specific inactivation of the insulin receptor gene. On an ApoE-deficient background, MphIRKO mice developed smaller atherosclerotic lesions. There was a dramatic decrease in LPS-stimulated IL-6 and IL-1beta expression in the presence of macrophage autonomous insulin resistance. Consistently, while insulin-resistant IRS-2-deficient mice on an ApoE-deficient background display aggravated atherosclerosis, fetal liver cell transplantation of IRS-2(-/-) ApoE(-/-) cells ameliorated atherosclerosis in Apo-E-deficient mice. Thus, systemic versus myeloid cell-restricted insulin resistance has opposing effects on the development of atherosclerosis, providing direct evidence that myeloid lineage autonomous insulin signaling provides proinflammatory signals predisposing to the development of atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Insulin Resistance/physiology , Insulin/physiology , Myeloid Cells/physiology , Adoptive Transfer , Animals , Apolipoproteins E/physiology , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Blotting, Western , Cell Line , Cell Lineage , Immunoprecipitation , Interleukin-1/physiology , Interleukin-6/physiology , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Insulin/genetics , Receptor, Insulin/physiology , Signal Transduction
14.
J Lipid Res ; 52(12): 2209-2225, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21957201

ABSTRACT

The generation of oxidized phospholipids in lipoproteins has been linked to vascular inflammation in atherosclerotic lesions. Products of phospholipid oxidation increase endothelial activation; however, their effects on macrophages are poorly understood, and it is unclear whether these effects are regulated by the biochemical pathways that metabolize oxidized phospholipids. We found that incubation of 1-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) with THP-1-derived macrophages upregulated the expression of cytokine genes, including granulocyte/macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor (TNF)-α, monocyte chemotactic protein 1 (MCP-1), interleukin (IL)-1ß, IL-6, and IL-8. In these cells, reagent POVPC was either hydrolyzed to lyso-phosphatidylcholine (lyso-PC) or reduced to 1-palmitoyl-2-(5-hydroxy-valeroyl)-sn-glycero-3-phosphocholine (PHVPC). Treatment with the phospholipase A(2) (PLA(2)) inhibitor, pefabloc, decreased POVPC hydrolysis and increased PHVPC accumulation. Pefabloc also increased the induction of cytokine genes in POVPC-treated cells. In contrast, PHVPC accumulation and cytokine production were decreased upon treatment with the aldose reductase (AR) inhibitor, tolrestat. In comparison with POVPC, lyso-PC led to 2- to 3-fold greater and PHVPC 10- to 100-fold greater induction of cytokine genes. POVPC-induced cytokine gene induction was prevented in bone-marrow derived macrophages from AR-null mice. These results indicate that although hydrolysis is the major pathway of metabolism, reduction further increases the proinflammatory responses to POVPC. Thus, vascular inflammation in atherosclerotic lesions is likely to be regulated by metabolism of phospholipid aldehydes in macrophages.


Subject(s)
Inflammation/metabolism , Phospholipid Ethers/metabolism , Phospholipid Ethers/pharmacology , Aldehyde Reductase/metabolism , Animals , Cell Line , Cytokines/genetics , Humans , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Oxidation-Reduction , Up-Regulation/drug effects
15.
Alcohol Clin Exp Res ; 35(8): 1435-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21463338

ABSTRACT

BACKGROUND: Alcohol abuse has long-term deleterious effects on the immune system, and results in a depletion and loss of function of CD4(+) T lymphocytes, which regulate both innate and adaptive immunity. T-lymphocyte activation via T-cell receptor (TCR) involves the lipid raft colocalization and aggregation of proteins into the immunological signalosome, which triggers a signaling cascade resulting in the production of interleukin-2 (IL-2). IL-2 regulates the proliferation and clonal expansion of activated T cells and is essential for an effective immune response. The present work examines the mechanisms underlying ethanol-induced dysfunction of CD4(+) T lymphocytes based on the hypothesis that ethanol downregulates lipid raft-mediated TCR signal transduction and resultant IL-2 production. METHODS: Primary or cultured human T lymphocytes were exposed to ethanol for 24 hours prior to stimulation with anti-CD3/anti-CD28 antibodies or phytohemagglutinin. Effects of ethanol exposure on TCR-signaling (including activation of Lck, ZAP70, LAT, and PLCγ1) and IL-2 gene expression were examined. RESULTS: Exposure of both primary and cultured human CD4(+) T lymphocytes to physiologically relevant concentrations of ethanol leads to down-regulation of IL-2 mRNA and protein via inhibition of DNA-binding activity of NFAT, the essential transcription factor for IL-2. Ethanol decreases tyrosine phosphorylation and activation of upstream signaling proteins PLCγ1, LAT, ZAP70, and Lck. These effects are prevented by inhibition of metabolism of ethanol. Sucrose density gradient fractionation and confocal microscopy revealed that ethanol inhibited essential upstream lipid raft-mediated TCR-dependent signaling events, namely colocalization of Lck, ZAP70, LAT, and PLCγ1 with plasma membrane lipid rafts. CONCLUSIONS: Overall, our data demonstrate that ethanol inhibits lipid raft-mediated TCR-signaling in CD4(+) T lymphocytes, resulting in suppression of IL-2 production. These findings may represent a novel mechanism underlying alcohol abuse-associated immune suppression and may be particularly relevant in diseases such as HIV/AIDS and hepatitis C virus infection where alcohol abuse is a known comorbidity.


Subject(s)
Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Immune Tolerance/drug effects , Interleukin-2/metabolism , Membrane Microdomains/drug effects , Receptors, Antigen, T-Cell/metabolism , CD4-Positive T-Lymphocytes/drug effects , Central Nervous System Depressants/immunology , Ethanol/immunology , Humans , Immunoprecipitation , Interleukin-2/analysis , Interleukin-2/immunology , Jurkat Cells , Membrane Microdomains/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocyte Subsets/drug effects
16.
J Immunol ; 182(12): 7625-33, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494286

ABSTRACT

Epidermal fatty acid-binding protein, E-FABP, a lipid chaperone, has been shown to regulate the inflammatory function of macrophages and dendritic cells. Herein, we demonstrate that T cell expression of E-FABP promotes Th17 differentiation, while counterregulating development of FoxP3(+) regulatory T cells (Tregs). In response to immunization with myelin oligodendrocyte glycoprotein peptide (MOG(35-55)), E-FABP-deficient mice generated reduced levels of Th17 cells and elevated levels of Tregs, as compared with wild-type mice. Likewise, naive CD4(+) T cells isolated from E-FABP-deficient mice showed reduced expression of IL-17 and enhanced expression of FoxP3, in vitro, when subjected to Th17 or Treg polarizing conditions, respectively. It has been demonstrated previously that IL-21, induced by IL-6, stimulates the expression of the nuclear receptors retinoic acid-related orphan receptor (ROR)gammat and RORalpha, which in turn induce expression of IL-17. We found that the impaired Th17 differentiation by E-FABP-deficient CD4(+) T cells was associated with lower levels of IL-21 expression in response to IL-6, as well as reduced expression of RORgammat and RORalpha. However, E-FABP-deficient CD4(+) T cells expressed significantly higher levels of the nuclear receptor peroxisome proliferator-activating receptor (PPAR)gamma than did wild-type CD4(+) T cells, and treatment with the PPARgamma antagonist GW9662 restored expression of IL-21, RORgammat, RORalpha, and IL-17 by E-FABP-deficient T cells to wild-type levels. The negative influence of E-FABP deficiency on IL-17 expression was attributed to PPARgamma-mediated suppression of IL-6-induced STAT3 activity. Thus, taken together, our data indicate that expression of E-FABP by CD4(+) T cells contributes to the control of IL-6 stimulation of the IL-21/ROR/IL-17 pathway and to the Th17/Treg counterbalance.


Subject(s)
Cell Differentiation/immunology , Fatty Acid-Binding Proteins/metabolism , Interleukin-17/immunology , Neoplasm Proteins/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , PPAR gamma/metabolism , Tretinoin/pharmacology
17.
Nat Med ; 7(6): 699-705, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11385507

ABSTRACT

The adipocyte fatty-acid-binding protein, aP2, has an important role in regulating systemic insulin resistance and lipid metabolism. Here we demonstrate that aP2 is also expressed in macrophages, has a significant role in their biological responses and contributes to the development of atherosclerosis. Apolipoprotein E (ApoE)-deficient mice also deficient for aP2 showed protection from atherosclerosis in the absence of significant differences in serum lipids or insulin sensitivity. aP2-deficient macrophages showed alterations in inflammatory cytokine production and a reduced ability to accumulate cholesterol esters when exposed to modified lipoproteins. Apoe-/- mice with Ap2+/+ adipocytes and Ap2-/- macrophages generated by bone-marrow transplantation showed a comparable reduction in atherosclerotic lesions to those with total aP2 deficiency, indicating an independent role for macrophage aP2 in atherogenesis. Through its distinct actions in adipocytes and macrophages, aP2 provides a link between features of the metabolic syndrome and could be a new therapeutic target for the prevention of atherosclerosis.


Subject(s)
Adipocytes/physiology , Apolipoproteins E/metabolism , Arteriosclerosis/physiopathology , Carrier Proteins/metabolism , Macrophages/physiology , Neoplasm Proteins , Nerve Tissue Proteins , Tumor Suppressor Proteins , Animals , Aorta/cytology , Arteriosclerosis/etiology , Arteriosclerosis/prevention & control , Bone Marrow Transplantation , Cell Line , Cholesterol Esters/metabolism , Diet , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Female , Foam Cells/physiology , Glucose/metabolism , Humans , Insulin/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Mol Cancer ; 9: 85, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20412587

ABSTRACT

BACKGROUND: Most prostate cancer (PCa)-related deaths are due to metastasis, which is mediated in part by chemokine receptor and corresponding ligand interaction. We have previously shown that PCa tissue and cell lines express high levels of the chemokine receptor CXCR5, than compared to their normal counterparts, and interaction of CXCR5 with its specific ligand (CXCL13) promoted PCa cell invasion, migration, and differential matrix metalloproteinase (MMP) expression. This study dissects some of the molecular mechanisms following CXCL13-CXCR5 interaction that mediate PCa cell migration and invasion. RESULTS: Using Western blot analysis, kinase-specific cell-based ELISAs, and migration and invasion assays, we show that PCa cell lines differentially express phosphoinositide-3 kinase (PI3K) catalytic subunit isoforms and dedicator of cytokinesis 2 (DOCK2). Specifically, we show that PC3 and normal prostatic epithelial (RWPE-1), but not LNCaP cell lines expressed DOCK2, while RWPE, PC3, and LNCaP cell lines expressed PI3K-p110alpha and -p110beta. Moreover, PC3 selectively expressed PI3K-p110gamma, but LNCaP and RWPE cell lines expressed PI3Kp110delta. CXCL13 caused CXCR5-dependent activation of the PI3Kp85alpha in LNCaP cells, and p85alpha as well as -p101 in PC3 cells. CXCL13-CXCR5 interaction regulated LNCaP and PC3 cell migration and invasion through extracellular signal-regulated kinase 1/2 (ERK1/2) activation that was primarily dependent on the PI3Kp110 isoform(s), Src, and focal adhesion kinase (FAK), but not DOCK2. CONCLUSIONS: While additional studies will be needed to determine the PI3K-independent (i.e., DOCK2-mediated) and -dependent events that dictate PCa cell responsiveness to CXCL13, these data provide evidence of the existence of cell type- and stimulus-specific signaling events that support migration and invasion of PCa cells.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , src-Family Kinases/metabolism , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Chemokine CXCL13/metabolism , Chemokine CXCL13/pharmacology , Chemokine CXCL5/metabolism , Enzyme Activation/physiology , Enzyme-Linked Immunosorbent Assay , GTPase-Activating Proteins , Humans , Male , Prostatic Neoplasms/pathology , Signal Transduction/physiology
19.
Eur J Immunol ; 39(8): 2126-35, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19609975

ABSTRACT

This study reveals that the IL-15 rapidly released into serum upon IL-12 injection into tumor-bearing mice is critical for the subsequent leukocytic infiltration of the tumor and tumor-bearing tissue. The increase in serum IL-15 occurs within 2 h after IL-12 injection concomitantly with a decrease in cytoplasmic IL-15 in tumor-associated Mphi (TAM). Injection of anti-IL-15 one hour prior to IL-12 abrogates subsequent leukocytic infiltration into the tumor and prevents the IL-12-induced reduction of primary tumor mass and the clearance of metastases. Administration of anti-IL-15 18 h after IL-12 did not have a detectable impact on IL-12-induced leukocytic infiltration of the tumor. Deletion of NK cells had no impact on the IL-12-induced change in the functional phenotype of TAM or on the subsequent initiation of leukocytic infiltration of the tumor. In concert with our previous studies demonstrating that IL-12 reduces tumor-supportive activities of TAM, the current study supports the hypothesis that functional re-programming of TAM not only undermines Mphi support for tumor growth but also contributes to a critical step in the initiation of anti-tumor immune responses. In this context, the functional plasticity and pro-immunogenic potential of TAM may constitute a significant and unappreciated target in existing cytokine therapies.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Interleukin-12/pharmacology , Interleukin-15/metabolism , Macrophages/drug effects , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cytokines/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-12/administration & dosage , Interleukin-15/genetics , Interleukin-15/immunology , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Tumor Burden/drug effects
20.
J Immunol ; 181(12): 8633-41, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050283

ABSTRACT

Herein, we demonstrate a role of AMP-activated protein kinase (AMPK) as a potent counterregulator of inflammatory signaling pathways in macrophages. Stimulation of macrophages with anti-inflammatory cytokines (i.e., IL-10 and TGFbeta) resulted in the rapid phosphorylation/activation of AMPK, whereas stimulation of macrophages with a proinflammatory stimulus (LPS) resulted in AMPK dephosphorylation/inactivation. Inhibition of AMPKalpha expression by RNA interference dramatically increased the mRNA levels of LPS-induced TNF-alpha, IL-6, and cyclooxygenase-2. Likewise, expression of a dominant negative AMPKalpha1 in macrophages enhanced TNF-alpha and IL-6 protein synthesis in response to LPS stimulation, while diminishing the production of IL-10. In contrast, transfection of macrophages with a constitutively active form of AMPKalpha1 resulted in decreased LPS-induced TNF-alpha and IL-6 production, and heightened production of IL-10. In addition, we found that AMPK negatively regulated LPS-induced IkappaB-alpha degradation and positively regulated Akt activation, accompanied by inhibition of glycogen synthase kinase beta and activation of CREB. Thus, AMPK directs signaling pathways in macrophages in a manner that suppresses proinflammatory responses and promotes macrophage polarization to an anti-inflammatory functional phenotype.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cell Polarity/immunology , Immunophenotyping , Inflammation Mediators/physiology , Macrophages/enzymology , Macrophages/immunology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Cell Line , Cell Polarity/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/physiology , Macrophages/cytology , Mice , Monocytes/cytology , Monocytes/enzymology , Monocytes/immunology , Signal Transduction/genetics , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL