Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Cell Metab ; 28(5): 689-705.e5, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30100196

ABSTRACT

It is clear that obesity increases the risk of many types of cancer, including breast cancer. However, the underlying molecular mechanisms by which obesity is linked to cancer risk remain to be defined. Herein, we report that circulating adipose fatty acid binding protein (A-FABP) promotes obesity-associated breast cancer development. Using clinical samples, we demonstrated that circulating A-FABP levels were significantly increased in obese patients with breast cancer in comparison with those without breast cancer. Circulating A-FABP released by adipose tissue directly targeted mammary tumor cells, enhancing tumor stemness and aggressiveness through activation of the IL-6/STAT3/ALDH1 pathway. Importantly, genetic deletion of A-FABP successfully reduced tumor ALHD1 activation and obesity-associated mammary tumor growth and development in different mouse models. Collectively, these data suggest circulating A-FABP as a new link between obesity and breast cancer risk, thereby revealing A-FABP as a potential new therapeutic target for treatment of obesity-associated cancers.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/etiology , Fatty Acid-Binding Proteins/blood , Obesity/complications , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/blood , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Models, Animal , Disease Progression , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Interleukin-6/metabolism , Isoenzymes/metabolism , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Obesity/blood , Obesity/metabolism , Obesity/pathology , Retinal Dehydrogenase/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
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 Invest Dermatol ; 138(9): 1925-1934, 2018 09.
Article in English | MEDLINE | ID: mdl-29559340

ABSTRACT

Skin lipids (e.g., fatty acids) are essential for normal skin functions. Epidermal FABP (E-FABP) is the predominant FABP expressed in skin epidermis. However, the role of E-FABP in skin homeostasis and pathology remains largely unknown. Herein, we utilized the 7,12-dimethylbenz(a)anthracene and 12-O-tetradecanolyphorbol-13-acetate-induced skin tumorigenesis model to assess the role of E-FABP in chemical-induced skin tumorigenesis. Compared to their wild-type littermates, mice deficient in E-FABP, but not adipose FABP, developed more skin tumors with higher incidence. 12-O-tetradecanolyphorbol-13-acetate functioning as a tumor promoter induced E-FABP expression and initiated extensive flaring inflammation in skin. Interestingly, 12-O-tetradecanolyphorbol-13-acetate -induced production of IFN-ß and IFN-λ in the skin tissue was dependent on E-FABP expression. Further protein and gene expression arrays demonstrated that E-FABP was critical in enhancing IFN-induced p53 responses and in suppressing SOX2 expression in keratinocytes. Thus, E-FABP expression in skin suppresses chemical-induced skin tumorigenesis through regulation of IFN/p53/SOX2 pathway. Collectively, our data suggest an unknown function of E-FABP in prevention of skin tumor development, and offer E-FABP as a therapeutic target for improving skin innate immunity in chemical-induced skin tumor prevention.


Subject(s)
Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Interferon-beta/genetics , Keratinocytes/metabolism , Neoplasm Proteins/genetics , SOXB1 Transcription Factors/genetics , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Epidermis/metabolism , Epidermis/pathology , Fatty Acid-Binding Proteins/biosynthesis , Interferon-beta/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasm Proteins/biosynthesis , Neoplasms, Experimental , RNA, Neoplasm/genetics , SOXB1 Transcription Factors/metabolism , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate/toxicity , Tumor Suppressor Protein p53/metabolism
4.
Cancer Res ; 78(9): 2343-2355, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29437708

ABSTRACT

Tumor-associated macrophages (TAM) play a critical role in cancer development and progression. However, the heterogeneity of TAM presents a major challenge to identify clinically relevant markers for protumor TAM. Here, we report that expression of adipocyte/macrophage fatty acid-binding protein (A-FABP) in TAM promotes breast cancer progression. Although upregulation of A-FABP was inversely associated with breast cancer survival, deficiency of A-FABP significantly reduced mammary tumor growth and metastasis. Furthermore, the protumor effect of A-FABP was mediated by TAM, in particular, in a subset of TAM with a CD11b+F4/80+MHCII-Ly6C- phenotype. A-FABP expression in TAM facilitated protumor IL6/STAT3 signaling through regulation of the NFκB/miR-29b pathway. Collectively, our results suggest A-FABP as a new functional marker for protumor TAM.Significance: These findings identify A-FABP as a functional marker for protumor macrophages, thus offering a new target for tumor immunotherapy. Cancer Res; 78(9); 2343-55. ©2018 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Immunohistochemistry , Macrophages/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Neoplasm Metastasis
5.
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
6.
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
7.
Oncotarget ; 7(23): 33783-95, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27177226

ABSTRACT

As a master metabolic sensor, AMP-activated protein kinase (AMPK) is involved in different fundamental cellular processes. Regulation of AMPK activity either by agonists (e.g., AICAR) or by antagonists (e.g., Compound C) has been widely employed to study the physiological functions of AMPK. However, mounting evidence indicates AMPK-independent effects for these chemicals and how they regulate immune cell functions remains largely unknown. Herein, using T cells from AMPK conditional knockout mice and their wild type littermates, we demonstrate that AICAR and Compound C can, indeed, activate or inhibit AMPK activity in T cells, respectively. Specifically, AICAR inhibits, but Compound C promotes, Ca2+-induced T cell death in an AMPK-dependent manner. In contrast, our data also demonstrate that AICAR and Compound C inhibit T cell activation and cytokine production in an AMPK-independent manner. Moreover, we find that the AMPK-independent activity of AICAR and Compound Cis mediated via the mTOR signaling pathway in activated T cells. Our results not only reveal the critical role of AMPK in regulating T cell survival and function, but also demonstrate AMPK-dependent and independent rolesof AICAR/Compound C in regulating T cell responses, thus suggesting a context-dependent effect of these "AMPK regulators".


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Enzyme Activators/pharmacology , Immunologic Factors/pharmacology , Lymphocyte Activation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ribonucleosides/pharmacology , T-Lymphocytes/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/pharmacology , Animals , Calcium Signaling/drug effects , Cell Death/drug effects , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Genotype , Mice, Knockout , Phenotype , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , TOR Serine-Threonine Kinases/metabolism
8.
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
9.
Oncotarget ; 6(10): 7944-58, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25760243

ABSTRACT

A number of studies have linked AMPK, a major metabolic sensor coordinating of multiple cellular functions, to tumor development and progression. However, the exact role of AMPK in tumor development is still controversial. Here we report that activation of AMPK promotes survival and anti-tumor function of T cells, in particular CD8+ T cells, resulting in superior tumor suppression in vivo. While AMPK expression is dispensable for T cell development, genetic deletion of AMPK promotes T cell death during in vitro activation and in vivo tumor development. Moreover, we demonstrate that protein phosphatases are the key mediators of AMPK-dependent effects on T cell death, and inhibition of phosphatase activity by okadaic acid successfully restores T cell survival and function. Altogether, our data suggest a novel mechanism by which AMPK regulates protein phosphatase activity in control of survival and function of CD8+ T cells, thereby enhancing their role in tumor immunosurveillance.


Subject(s)
AMP-Activated Protein Kinases/deficiency , CD8-Positive T-Lymphocytes/metabolism , Phosphoprotein Phosphatases/metabolism , AMP-Activated Protein Kinases/metabolism , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Cell Death/physiology , Cell Line, Tumor , Cell Survival/physiology , Humans
10.
Mol Cell Biol ; 35(6): 1055-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582199

ABSTRACT

Chronic inflammation in obese adipose tissue is linked to endoplasmic reticulum (ER) stress and systemic insulin resistance. Targeted deletion of the murine fatty acid binding protein (FABP4/aP2) uncouples obesity from inflammation although the mechanism underlying this finding has remained enigmatic. Here, we show that inhibition or deletion of FABP4/aP2 in macrophages results in increased intracellular free fatty acids (FFAs) and elevated expression of uncoupling protein 2 (UCP2) without concomitant increases in UCP1 or UCP3. Silencing of UCP2 mRNA in FABP4/aP2-deficient macrophages negated the protective effect of FABP loss and increased ER stress in response to palmitate or lipopolysaccharide (LPS). Pharmacologic inhibition of FABP4/aP2 with the FABP inhibitor HTS01037 also upregulated UCP2 and reduced expression of BiP, CHOP, and XBP-1s. Expression of native FABP4/aP2 (but not the non-fatty acid binding mutant R126Q) into FABP4/aP2 null cells reduced UCP2 expression, suggesting that the FABP-FFA equilibrium controls UCP2 expression. FABP4/aP2-deficient macrophages are resistant to LPS-induced mitochondrial dysfunction and exhibit decreased mitochondrial protein carbonylation and UCP2-dependent reduction in intracellular reactive oxygen species. These data demonstrate that FABP4/aP2 directly regulates intracellular FFA levels and indirectly controls macrophage inflammation and ER stress by regulating the expression of UCP2.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Inflammation/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Adipose Tissue/metabolism , Animals , Cell Line , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acids, Nonesterified/genetics , Inflammation/genetics , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Obesity/genetics , Obesity/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
11.
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
12.
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
13.
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
14.
J Leukoc Biol ; 94(6): 1113-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23883517

ABSTRACT

AMPK is a serine/threonine kinase that regulates energy homeostasis and metabolic stress in eukaryotes. Previous work from our laboratory, as well as by others, has provided evidence that AMPKα1 acts as a negative regulator of TLR-induced inflammatory function. Herein, we demonstrate that AMPKα1-deficient macrophages and DCs exhibit heightened inflammatory function and an enhanced capacity for antigen presentation favoring the promotion of Th1 and Th17 responses. Macrophages and DCs generated from AMPKα1-deficient mice produced higher levels of proinflammatory cytokines and decreased production of the anti-inflammatory cytokine IL-10 in response to TLR and CD40 stimulation as compared with WT cells. In assays of antigen presentation, AMPKα1 deficiency in the myeloid APC and T cell populations contributed to enhanced IL-17 and IFN-γ production. Focusing on the CD154-CD40 interaction, we found that CD40 stimulation resulted in increased phosphorylation of ERK1/2, p38, and NF-κB p65 and decreased activation of the anti-inflammatory Akt -GSK3ß-CREB pathway in DCs deficient for AMPKα1. Our data demonstrate that AMPKα1 serves to attenuate LPS and CD40-mediated proinflammatory activity of myeloid APCs and that AMPKα1 activity in both APC and T cells contributes to T cell functional polarization during antigen presentation.


Subject(s)
AMP-Activated Protein Kinases/immunology , Antigen Presentation , Antigen-Presenting Cells/immunology , CD40 Antigens/immunology , MAP Kinase Signaling System/immunology , Myeloid Cells/immunology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/enzymology , CD40 Antigens/genetics , CD40 Antigens/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/enzymology , Th1 Cells/cytology , Th1 Cells/enzymology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/enzymology , Th17 Cells/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism
15.
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
16.
J Nutr Biochem ; 24(9): 1609-15, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23618528

ABSTRACT

The therapeutic effects of probiotic treatment in alcoholic liver disease (ALD) have been studied in both patients and experimental animal models. Although the precise mechanisms of the pathogenesis of ALD are not fully understood, gut-derived endotoxin has been postulated to play a crucial role in hepatic inflammation. Previous studies have demonstrated that probiotic therapy reduces circulating endotoxin derived from intestinal gram-negative bacteria in ALD. In this study, we investigated the effects of probiotics on hepatic tumor necrosis factor-α (TNFα) production and inflammation in response to chronic alcohol ingestion. Mice were fed Lieber DeCarli liquid diet containing 5% alcohol for 8weeks, and Lactobacillus rhamnosus GG (LGG) was supplemented in the last 2 weeks. Eight-week alcohol feeding caused a significant increase in hepatic inflammation as shown by histological assessment and hepatic tissue myeloperoxidase activity assay. Two weeks of LGG supplementation reduced hepatic inflammation and liver injury and markedly reduced TNFα expression. Alcohol feeding increased hepatic mRNA expression of Toll-like receptors (TLRs) and CYP2E1 and decreased nuclear factor erythroid 2-related factor 2 expression. LGG supplementation attenuated these changes. Using human peripheral blood monocytes-derived macrophages, we also demonstrated that incubation with ethanol primes both lipopolysaccharide- and flagellin-induced TNFα production, and LGG culture supernatant reduced this induction in a dose-dependent manner. In addition, LGG treatment also significantly decreased alcohol-induced phosphorylation of p38 MAP kinase. In conclusion, probiotic LGG treatment reduced alcohol-induced hepatic inflammation by attenuation of TNFα production via inhibition of TLR4- and TLR5-mediated endotoxin activation.


Subject(s)
Inflammation/therapy , Lacticaseibacillus rhamnosus , Liver Diseases, Alcoholic/therapy , Probiotics/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Animals , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Endotoxins/adverse effects , Endotoxins/metabolism , Ethanol/metabolism , Flagellin/adverse effects , Flagellin/metabolism , Gram-Negative Bacteria/metabolism , Humans , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestines/microbiology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/adverse effects , Lipopolysaccharides/metabolism , Liver/pathology , Liver Diseases, Alcoholic/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Probiotics/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
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.

18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...