Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 7(11): e50069, 2012.
Article in English | MEDLINE | ID: mdl-23166823

ABSTRACT

BACKGROUND: There is an inverse secular trend between the incidence of obesity and gastric colonization with Helicobacter pylori, a bacterium that can affect the secretion of gastric hormones that relate to energy homeostasis. H. pylori strains that carry the cag pathogenicity island (PAI) interact more intimately with gastric epithelial cells and trigger more extensive host responses than cag(-) strains. We hypothesized that gastric colonization with H. pylori strains differing in cag PAI status exert distinct effects on metabolic and inflammatory phenotypes. METHODOLOGY/PRINCIPAL FINDINGS: To test this hypothesis, we examined metabolic and inflammatory markers in db/db mice and mice with diet-induced obesity experimentally infected with isogenic forms of H. pylori strain 26695: the cag PAI wild-type and its cag PAI mutant strain 99-305. H. pylori colonization decreased fasting blood glucose levels, increased levels of leptin, improved glucose tolerance, and suppressed weight gain. A response found in both wild-type and mutant H. pylori strain-infected mice included decreased white adipose tissue macrophages (ATM) and increased adipose tissue regulatory T cells (Treg) cells. Gene expression analyses demonstrated upregulation of gastric PPAR γ-responsive genes (i.e., CD36 and FABP4) in H. pylori-infected mice. The loss of PPAR γ in immune and epithelial cells in mice impaired the ability of H. pylori to favorably modulate glucose homeostasis and ATM infiltration during high fat feeding. CONCLUSIONS/SIGNIFICANCE: Gastric infection with some commensal strains of H. pylori ameliorates glucose homeostasis in mice through a PPAR γ-dependent mechanism and modulates macrophage and Treg cell infiltration into the abdominal white adipose tissue.


Subject(s)
Gastric Mucosa/microbiology , Genomic Islands/genetics , Helicobacter Infections/metabolism , Helicobacter pylori/growth & development , Homeostasis/physiology , Obesity/microbiology , PPAR gamma/metabolism , Adipose Tissue/cytology , Adipose Tissue/immunology , Animals , Blood Glucose , Body Weight , CD36 Antigens/metabolism , Enzyme-Linked Immunosorbent Assay , Fatty Acid-Binding Proteins/metabolism , Flow Cytometry , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gene Expression Profiling , Ghrelin/blood , Helicobacter Infections/immunology , Helicobacter pylori/genetics , Insulin/blood , Leptin/blood , Macrophages/immunology , Mice , T-Lymphocytes, Regulatory/immunology
2.
PLoS One ; 6(8): e24031, 2011.
Article in English | MEDLINE | ID: mdl-21904603

ABSTRACT

BACKGROUND: Treatments for inflammatory bowel disease (IBD) are modestly effective and associated with side effects from prolonged use. As there is no known cure for IBD, alternative therapeutic options are needed. Peroxisome proliferator-activated receptor-gamma (PPARγ) has been identified as a potential target for novel therapeutics against IBD. For this project, compounds were screened to identify naturally occurring PPARγ agonists as a means to identify novel anti-inflammatory therapeutics for experimental assessment of efficacy. METHODOLOGY/PRINCIPAL FINDINGS: Here we provide complementary computational and experimental methods to efficiently screen for PPARγ agonists and demonstrate amelioration of experimental IBD in mice, respectively. Computational docking as part of virtual screening (VS) was used to test binding between a total of eighty-one compounds and PPARγ. The test compounds included known agonists, known inactive compounds, derivatives and stereoisomers of known agonists with unknown activity, and conjugated trienes. The compound identified through VS as possessing the most favorable docked pose was used as the test compound for experimental work. With our combined methods, we have identified α-eleostearic acid (ESA) as a natural PPARγ agonist. Results of ligand-binding assays complemented the screening prediction. In addition, ESA decreased macrophage infiltration and significantly impeded the progression of IBD-related phenotypes through both PPARγ-dependent and -independent mechanisms in mice with experimental IBD. CONCLUSIONS/SIGNIFICANCE: This study serves as the first significant step toward a large-scale VS protocol for natural PPARγ agonist screening that includes a massively diverse ligand library and structures that represent multiple known target pharmacophores.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Linolenic Acids/therapeutic use , PPAR gamma/metabolism , Animals , Cell Line , Mice , PPAR gamma/agonists , Real-Time Polymerase Chain Reaction
3.
J Obes ; 2011: 897894, 2011.
Article in English | MEDLINE | ID: mdl-21253508

ABSTRACT

The prevalence of obesity and its associated comorbidities has grown to epidemic proportions in the US and worldwide. Thus, developing safe and effective therapeutic approaches against these widespread and debilitating diseases is important and timely. Activation of peroxisome proliferator-activated receptors (PPARs) α, γ, and δ through several classes of pharmaceuticals can prevent or treat a variety of metabolic and inflammatory diseases, including type II diabetes (T2D). Thus, PPARs represent important molecular targets for developing novel and better treatments for a wide range of debilitating and widespread obesity-related diseases and disorders. However, available PPAR γ agonistic drugs such as Avandia have significant adverse side effects, including weight gain, fluid retention, hepatotoxicity, and congestive heart failure. An alternative to synthetic agonists of PPAR γ is the discovery and development of naturally occurring and safer nutraceuticals that may be dual or pan PPAR agonists. The purpose of this paper is to summarize the health effects of three plant-derived PPAR agonists: abscisic acid (ABA), punicic acid (PUA), and catalpic acid (CAA) in the prevention and treatment of chronic inflammatory and metabolic diseases and disorders.

5.
J Biol Chem ; 286(4): 2504-16, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21088297

ABSTRACT

Abscisic acid (ABA) has shown efficacy in the treatment of diabetes and inflammation; however, its molecular targets and the mechanisms of action underlying its immunomodulatory effects remain unclear. This study investigates the role of peroxisome proliferator-activated receptor γ (PPAR γ) and lanthionine synthetase C-like 2 (LANCL2) as molecular targets for ABA. We demonstrate that ABA increases PPAR γ reporter activity in RAW 264.7 macrophages and increases ppar γ expression in vivo, although it does not bind to the ligand-binding domain of PPAR γ. LANCL2 knockdown studies provide evidence that ABA-mediated activation of macrophage PPAR γ is dependent on lancl2 expression. Consistent with the association of LANCL2 with G proteins, we provide evidence that ABA increases cAMP accumulation in immune cells. ABA suppresses LPS-induced prostaglandin E(2) and MCP-1 production via a PPAR γ-dependent mechanism possibly involving activation of PPAR γ and suppression of NF-κB and nuclear factor of activated T cells. LPS challenge studies in PPAR γ-expressing and immune cell-specific PPAR γ null mice demonstrate that ABA down-regulates toll-like receptor 4 expression in macrophages and T cells in vivo through a PPAR γ-dependent mechanism. Global transcriptomic profiling and confirmatory quantitative RT-PCR suggest novel candidate targets and demonstrate that ABA treatment mitigates the effect of LPS on the expression of genes involved in inflammation, metabolism, and cell signaling, in part, through PPAR γ. In conclusion, ABA decreases LPS-mediated inflammation and regulates innate immune responses through a bifurcating pathway involving LANCL2 and an alternative, ligand-binding domain-independent mechanism of PPAR γ activation.


Subject(s)
Abscisic Acid/pharmacology , Immunity, Innate/drug effects , Macrophages/metabolism , PPAR gamma/metabolism , Plant Growth Regulators/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Dinoprostone/biosynthesis , Dinoprostone/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Mutant Strains , PPAR gamma/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
6.
J Nutr Biochem ; 22(9): 812-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21109419

ABSTRACT

The phytohormone abscisic acid (ABA) has been shown to be effective in ameliorating chronic and acute inflammation. The objective of this study was to investigate whether ABA's anti-inflammatory efficacy in the gut is dependent on peroxisome proliferator-activated receptor γ (PPARγ) in T cells. PPARγ-expressing and T cell-specific PPARγ null mice were fed diets with or without ABA (100 mg/kg) for 35 days prior to challenge with 2.5% dextran sodium sulfate. The severity of clinical disease was assessed daily, and mice were euthanized on Day 7 of the dextran sodium sulfate challenge. Colonic inflammation was assessed through macroscopic and histopathological examination of inflammatory lesions and real-time quantitative RT-PCR-based quantification of inflammatory genes. Flow cytometry was used to phenotypically characterize leukocyte populations in the blood and mesenteric lymph nodes. Colonic sections were stained immunohistochemically to determine the effect of ABA on colonic regulatory T (T(reg)) cells. ABA's beneficial effects on disease activity were completely abrogated in T cell-specific PPARγ null mice. Additionally, ABA improved colon histopathology, reduced blood F4/80(+)CD11b(+) monocytes, increased the percentage of CD4(+) T cells expressing the inhibitory molecule cytotoxic T lymphocyte antigen 4 in blood and enhanced the number of T(reg) cells in the mesenteric lymph nodes and colons of PPARγ-expressing but not T cell-specific PPARγ null mice. We conclude that dietary ABA ameliorates experimental inflammatory bowel disease by enhancing T(reg) cell accumulation in the colonic lamina propria through a PPARγ-dependent mechanism.


Subject(s)
Abscisic Acid/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , PPAR gamma/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Animals, Genetically Modified , Down-Regulation , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Mice , Mice, Transgenic , PPAR gamma/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
7.
BMC Gastroenterol ; 10: 60, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20537136

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPAR gamma) is a nuclear receptor whose activation has been shown to modulate macrophage and T cell-mediated inflammation. The objective of this study was to investigate the mechanisms by which the deletion of PPAR gamma in T cells modulates immune cell distribution and colonic gene expression and the severity of experimental IBD. METHODS: PPAR gamma flfl; CD4 Cre+ (CD4cre) or Cre- (WT) mice were challenged with 2.5% dextran sodium sulfate in their drinking water for 0, 2, or 7 days. Mice were scored on disease severity both clinically and histopathologically. Flow cytometry was used to assess lymphocyte and macrophage populations in the blood, spleen, and mesenteric lymph nodes (MLN). Global gene expression in colonic mucosa was profiled using Affymetrix microarrays. RESULTS: The deficiency of PPAR gamma in T cells accelerated the onset of disease and body weight loss. Examination of colon histopathology revealed significantly greater epithelial erosion, leukocyte infiltration, and mucosal thickening in the CD4cre mice on day 7. CD4cre mice had more CD8+ T cells than WT mice and fewer CD4+ FoxP3+ regulatory T cells (Treg) and IL10+ CD4+ T cells in blood and MLN, respectively. Transcriptomic profiling revealed around 3000 genes being transcriptionally altered as a result of DSS challenge in CD4cre mice. These included up-regulated mRNA expression of adhesion molecules, proinflammatory cytokines interleukin-6 (IL-6) and IL-1beta, and suppressor of cytokine signaling 3 (SOCS-3) on day 7. Gene set enrichment analysis (GSEA) showed that the ribosome and Krebs cycle pathways were downregulated while the apoptosis pathway was upregulated in colons of mice lacking PPAR gamma in T cells. CONCLUSIONS: The expression of PPAR gamma in T cells is involved in preventing gut inflammation by regulating colonic expression of adhesion molecules and inflammatory mediators at later stages of disease while favoring the recruitment of Treg to the mucosal inductive sites.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , PPAR gamma/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Animals , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Disease Progression , Inflammatory Bowel Diseases/chemically induced , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
8.
PLoS One ; 5(4): e10215, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20422041

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptors are nuclear receptors highly expressed in intestinal epithelial cells (IEC) and immune cells within the gut mucosa and are implicated in modulating inflammation and immune responses. The objective of this study was to investigate the effect of targeted deletion of PPAR gamma in IEC on progression of experimental inflammatory bowel disease (IBD). METHODOLOGY/PRINCIPAL FINDINGS: In the first phase, PPAR gamma flfl; Villin Cre- (VC-) and PPAR gamma flfl; Villin Cre+ (VC+) mice in a mixed FVB/C57BL/6 background were challenged with 2.5% dextran sodium sulfate (DSS) in drinking water for 0, 2, or 7 days. VC+ mice express a transgenic recombinase under the control of the Villin-Cre promoter that causes an IEC-specific deletion of PPAR gamma. In the second phase, we generated VC- and VC+ mice in a C57BL/6 background that were challenged with 2.5% DSS. Mice were scored on disease severity both clinically and histopathologically. Flow cytometry was used to phenotypically characterize lymphocyte and macrophage populations in blood, spleen and mesenteric lymph nodes. Global gene expression analysis was profiled using Affymetrix microarrays. The IEC-specific deficiency of PPAR gamma in mice with a mixed background worsened colonic inflammatory lesions, but had no effect on disease activity (DAI) or weight loss. In contrast, the IEC-specific PPAR gamma null mice in C57BL/6 background exhibited more severe inflammatory lesions, DAI and weight loss in comparison to their littermates expressing PPAR gamma in IEC. Global gene expression profiling revealed significantly down-regulated expression of lysosomal pathway genes and flow cytometry results demonstrated suppressed production of IL-10 by CD4+ T cells in mesenteric lymph nodes (MLN) of IEC-specific PPAR gamma null mice. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that adequate expression of PPAR gamma in IEC is required for the regulation of mucosal immune responses and prevention of experimental IBD, possibly by modulation of lysosomal and antigen presentation pathways.


Subject(s)
Colon/pathology , Epithelial Cells/pathology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/pathology , PPAR gamma/immunology , Animals , Epithelial Cells/immunology , Gene Expression Profiling , Immunity , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout
9.
Clin Nutr ; 29(6): 824-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20236740

ABSTRACT

BACKGROUND & AIMS: Abscisic acid (ABA) has shown effectiveness in ameliorating inflammation in obesity, diabetes and cardiovascular disease models. The objective of this study was to determine whether ABA prevents or ameliorates experimental inflammatory bowel disease (IBD). METHODS: C57BL/6J mice were fed diets with or without ABA (100mg/kg) for 35 days prior to challenge with 2.5% dextran sodium sulfate (DSS). The severity of clinical disease was assessed daily. Colonic mucosal lesions were evaluated by histopathology, and cellular adhesion molecular and inflammatory markers were assayed by real-time quantitative PCR. Flow cytometry was used to quantify leukocyte populations in the blood, spleen, and mesenteric lymph nodes (MLN). The effect of ABA on cytotoxic T-lymphocyte antigen 4 (CTLA-4) expression in splenocytes was also investigated. RESULTS: ABA significantly ameliorated disease activity, colitis and reduced colonic leukocyte infiltration and inflammation. These improvements were associated with downregulation in vascular cell adhesion marker-1 (VCAM-1), E-selectin, and mucosal addressin adhesion marker-1 (MAdCAM-1) expression. ABA also increased CD4(+) and CD8(+) T-lymphocytes in blood and MLN and regulatory T cells in blood. In vitro, ABA increased CTLA-4 expression through a PPAR γ-dependent mechanism. CONCLUSIONS: We conclude that ABA ameliorates gut inflammation by modulating T cell distribution and adhesion molecule expression.


Subject(s)
Abscisic Acid/pharmacology , Cell Adhesion , Down-Regulation , Inflammatory Bowel Diseases/prevention & control , Animals , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Adhesion Molecules/metabolism , Disease Models, Animal , E-Selectin/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mucoproteins , Obesity/pathology , PPAR gamma/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
10.
Clin Nutr ; 29(5): 646-53, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20207056

ABSTRACT

BACKGROUND & AIMS: Abscisic acid (ABA) is effective in preventing insulin resistance and obesity-related inflammation through a PPAR γ-dependent mechanism. The objective of this study was to assess the efficacy ABA in improving glucose homeostasis and suppress inflammation when administered in combination with rosiglitazone (Ros) and to determine whether PPAR γ activation by ABA is initiated via cAMP/protein kinase A (PKA) signaling. METHODS: Obese db/db mice were fed high-fat diets containing 0, 10, or 70 mg/kg Ros with and without racemic ABA (100 mg/kg) for 60 days. Glucose tolerance and fasting insulin levels were assessed at 6 and 8 weeks, respectively, and adipose tissue macrophage (ATM) infiltration was examined by flow cytometry. Gene expression was examined on white adipose tissue (WAT) and stromal vascular cells (SVCs) cultured with ABA, Ros, or an ABA/Ros combination. RESULTS: Both Ros and ABA improved glucose tolerance, and ABA decreased plasma insulin levels while having no effect on Ros-induced weight gain. ABA in combination with low-dose Ros (10 mg/kg; Roslo) synergistically inhibited ATM infiltration. Treatment of SVCs with Ros, ABA or ABA/Ros suppressed expression of the M1 marker CCL17. ABA and Ros synergistically increased PPAR γ activity and pretreatment with a cAMP-inhibitor or a PKA-inhibitor abrogated ABA-induced PPAR γ activation. CONCLUSIONS: ABA and Ros act synergistically to modulate PPAR γ activity and macrophage accumulation in WAT and ABA enhances PPAR γ activity through a membrane-initiated mechanism dependent on cAMP/PKA signaling.


Subject(s)
Abscisic Acid/pharmacokinetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose Intolerance/metabolism , Inflammation/metabolism , PPAR gamma/metabolism , Thiazolidinediones/pharmacokinetics , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Data Collection , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Glucose Intolerance/drug therapy , Inflammation/drug therapy , Insulin/blood , Insulin/metabolism , Macrophages/metabolism , Mice , Mice, Obese , Obesity/drug therapy , Obesity/metabolism , Rosiglitazone
11.
J Nutr Biochem ; 21(12): 1178-85, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20092994

ABSTRACT

Abscisic acid (ABA) is a natural phytohormone which improves insulin sensitivity and reduces adipose tissue inflammation when supplemented into diets of obese mice. The objective of this study was to investigate the mechanisms by which ABA prevents or ameliorates atherosclerosis. apolipoprotein E-deficient (ApoE(-/-)) mice were fed high-fat diets with or without ABA for 84 days. Systolic blood pressure was assessed on Days 0, 28, 56 and 72. Gene expression, immune cell infiltration and histological lesions were evaluated in the aortic root wall. Human aortic endothelial cells were used to examine the effect of ABA on 3',5'-cyclic adenosine monophosphate (cAMP) and nitric oxide (NO) production in vitro. We report that ABA-treated mice had significantly improved systolic blood pressure and decreased accumulation of F4/80(+)CD11b(+) macrophages and CD4(+) T cells in aortic root walls. At the molecular level, ABA significantly enhanced aortic endothelial nitric oxide synthase (eNOS) and tended to suppress aortic vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) expression and plasma MCP-1 concentrations. ABA also caused a dose-dependent increase in intracellular concentrations of cAMP and NO and up-regulated eNOS mRNA expression in human aortic endothelial cells. This is the first report showing that ABA prevents or ameliorates atherosclerosis-induced hypertension, immune cell recruitment into the aortic root wall and up-regulates aortic eNOS expression in ApoE(-/-) mice.


Subject(s)
Abscisic Acid/pharmacology , Aorta/immunology , Atherosclerosis/immunology , CD4-Positive T-Lymphocytes/metabolism , Macrophages/metabolism , Abscisic Acid/immunology , Animals , Aorta/metabolism , Atherosclerosis/prevention & control , Blood Pressure/drug effects , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Hypertension/drug therapy , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism
12.
J Nutr ; 140(3): 515-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20089779

ABSTRACT

Conjugated linoleic acid (CLA) exerts a protective effect on experimental inflammatory bowel disease and shows promise as a chemopreventive agent against colorectal cancer (CRC) in mice, although the mechanisms by which it exerts its beneficial effects against malignancies in the gut are not completely understood. Mice lacking PPARgamma in immune and epithelial cells and PPARgamma-expressing littermates were fed either control or CLA-supplemented (1 g CLA/100 g) diets to determine the role of PPARgamma in inflammation-induced CRC. To induce tumor formation and colitis, mice were treated with azoxymethane and then challenged with 2% dextran sodium sulfate, respectively. Dietary CLA ameliorated disease activity, decreased colitis, and prevented adenocarcinoma formation in the PPARgamma-expressing floxed mice but not in the tissue-specific PPARgamma-null mice. Dietary CLA supplementation significantly decreased the percentages of macrophages in the mesenteric lymph nodes (MLN) regardless of the genotype and increased regulatory T cell numbers in MLN of PPARgamma-expressing, but not in the tissue-specific, PPARgamma-null mice. Colonic tumor necrosis factor-alpha mRNA expression was significantly suppressed in CLA-fed, PPARgamma-expressing mice. This study suggests CLA ameliorates colitis and prevents tumor formation in part through a PPARgamma-dependent mechanism.


Subject(s)
Colorectal Neoplasms/drug therapy , Gene Expression Regulation/drug effects , Inflammation/complications , Linoleic Acids, Conjugated/pharmacology , PPAR gamma/metabolism , Animals , Cell Line , Diet , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/drug therapy , Linoleic Acids, Conjugated/administration & dosage , Lymph Nodes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout
13.
Cell Immunol ; 258(2): 138-46, 2009.
Article in English | MEDLINE | ID: mdl-19423085

ABSTRACT

Macrophage infiltration into adipose tissue is a hallmark of obesity. We recently reported two phenotypically distinct subsets of adipose tissue macrophages (ATM) based on the surface expression of the glycoprotein F4/80 and responsiveness to treatment with a peroxisome proliferator-activated receptor (PPAR) gamma agonist. Hence, we hypothesized that F4/80(hi) and F4/80(lo) ATM differentially express PPAR gamma. This study phenotypically and functionally characterizes F4/80(hi) and F4/80(lo) ATM subsets during obesity. Changes in gene expression were also examined on sorted F4/80(lo) and F4/80(hi) ATM by quantitative real-time RT-PCR. We show that while F4/80(lo) macrophages predominate in adipose tissue of lean mice, obesity causes accumulation of both F4/80(lo) and F4/80(hi) ATM. Moreover, accumulation of F4/80(hi) ATM in adipose tissue is associated with impaired glucose tolerance. Phenotypically, F4/80(hi) ATM express greater amounts of CD11c, MHC II, CD49b, and CX3CR1 and produce more TNF-alpha, MCP-1, and IL-10 than F4/80(lo) ATM. Gene expression analyses of the sorted populations revealed that only the F4/80(lo) population produced IL-4, whereas the F4/80(hi) ATM expressed greater amounts of PPAR gamma, delta, CD36 and toll-like receptor-4. In addition, the deficiency of PPAR gamma in immune cells favors expression of M1 and impairs M2 macrophage marker expression in adipose tissue. Thus, PPAR gamma is differentially expressed in F4/80(hi) versus F4/80(low) ATM subsets and its deficiency favors a predominance of M1 markers in WAT.


Subject(s)
Adipose Tissue, White/immunology , Inflammation/immunology , Macrophage Activation , Macrophages/metabolism , Obesity/immunology , PPAR gamma/metabolism , Animals , Antigens, Differentiation/metabolism , CD36 Antigens/metabolism , Cell Count , Chemokine CCL1/metabolism , Chemokine CCL17/metabolism , Cytokines/analysis , Cytokines/metabolism , Glucose/metabolism , Interleukin-4/biosynthesis , Macrophages/immunology , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/deficiency , PPAR gamma/genetics
14.
J Nutrigenet Nutrigenomics ; 1(3): 126-35, 2008.
Article in English | MEDLINE | ID: mdl-19776622

ABSTRACT

In its simplest form, obesity is a state characterized by nutrient overabundance leading to hypertrophy of storage cells in white adipose tissue and the deposition of excess lipids into key metabolic regions, such as skeletal muscle and liver. Ever so steadily, this condition begins to manifest itself as progressive insulin resistance and thus ensues a myriad of other chronic diseases, such as type 2 diabetes, cardiovascular disease, and hypertension, which all fall into the realm of the metabolic syndrome. To offset imbalances in nutrient availability, however, it appears that nature has developed the peroxisome proliferator-activated receptors (PPARs), a family of endogenous lipid sensors that adeptly modulate our rates of macronutrient oxidation and regulate the systemic inflammatory response, which itself is tightly linked to the development of obesity-induced chronic disease. By understanding how PPARs alpha, delta and gamma act jointly to maintain metabolic homeostasis and reduce the chronic inflammation associated with obesity, we may one day discover that the machinery needed to defeat obesity and control the devastating consequences of the metabolic syndrome have been with us the entire time.


Subject(s)
Diet , Metabolic Syndrome/prevention & control , Metabolic Syndrome/therapy , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Adipocytes/pathology , Animals , Glucose/metabolism , Humans , Hypertrophy , Inflammation/complications , Inflammation/metabolism , Insulin Resistance/physiology , Lipid Metabolism/physiology , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Nutritional Physiological Phenomena , Obesity/etiology , Obesity/pathology , Peroxisome Proliferator-Activated Receptors/metabolism
15.
J Nutr Biochem ; 19(4): 216-28, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17618105

ABSTRACT

Abscisic acid (ABA) is a natural phytohormone and peroxisome proliferator-activated receptor gamma (PPARgamma) agonist that significantly improves insulin sensitivity in db/db mice. Although it has become clear that obesity is associated with macrophage infiltration into white adipose tissue (WAT), the phenotype of adipose tissue macrophages (ATMs) and the mechanisms by which insulin-sensitizing compounds modulate their infiltration remain unknown. We used a loss-of-function approach to investigate whether ABA ameliorates insulin resistance through a mechanism dependent on immune cell PPARgamma. We characterized two phenotypically distinct ATM subsets in db/db mice based on their surface expression of F4/80. F4/80(hi) ATMs were more abundant and expressed greater concentrations of chemokine receptor (CCR) 2 and CCR5 when compared to F4/80(lo) ATMs. ABA significantly decreased CCR2(+) F4/80(hi) infiltration into WAT and suppressed monocyte chemoattractant protein-1 (MCP-1) expression in WAT and plasma. Furthermore, the deficiency of PPARgamma in immune cells, including macrophages, impaired the ability of ABA to suppress the infiltration of F4/80(hi) ATMs into WAT, to repress WAT MCP-1 expression and to improve glucose tolerance. We provide molecular evidence in vivo demonstrating that ABA improves insulin sensitivity and obesity-related inflammation by inhibiting MCP-1 expression and F4/80(hi) ATM infiltration through a PPARgamma-dependent mechanism.


Subject(s)
Abscisic Acid/pharmacology , Adipose Tissue, White/immunology , Chemokine CCL2/metabolism , Inflammation/metabolism , Insulin Resistance , Macrophages/immunology , PPAR gamma/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Chemokine CCL2/genetics , Inflammation/immunology , Liver/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred Strains , Obesity/metabolism , PPAR gamma/deficiency , Phenotype , Triglycerides/blood
16.
Am J Clin Nutr ; 85(3): 824-36, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17344506

ABSTRACT

BACKGROUND: Whereas the immunomodulatory effects of feeding either arachidonic acid (AA) or docosahexaenoic acid (DHA) separately have been previously investigated, little is known about the immunomodulatory efficacy of AA or DHA when they are fed in combination as infant formula ingredients. OBJECTIVE: The objective of this study was to investigate the ability of AA- and DHA(AA/DHA)-enriched infant formula to modulate immune responses in the neonate in response to an inactivated influenza virus vaccine. DESIGN: Neonatal piglets (n = 48) were weaned on day 2 of age and distributed into 16 blocks of 3 littermate piglets each. Within each block, piglets were randomly assigned to a control formula, AA/DHA-enriched formula (0.63% AA and 0.34% DHA), or sow milk for 30 d. On day 9, 8 blocks of piglets were immunized with an inactivated influenza virus vaccine. On days 0, 9, 16, 23, and 30 after weaning, we measured influenza virus-specific T cell proliferation and phenotype of T subsets in peripheral blood. A delayed-type hypersensitivity reaction test was administered on day 28. Cytokine messenger RNA expression was determined by quantitative real time reverse transcriptase-polymerase chain reaction on day 30. RESULTS: The influenza virus-specific CD4(+) and CD8(+) T cell ex vivo lymphoproliferative responses were significantly lower on day 23 after immunization in piglets receiving dietary AA/DHA supplementation and sow milk than in those receiving the unsupplemented control formula. The immunomodulatory effects of AA/DHA-enriched formulas were consistent with up-regulation of interleukin 10 in peripheral blood mononuclear cells. CONCLUSION: Overall, it appears that the AA/DHA-enriched formula modulated antigen-specific T cell responses in part through an interleukin 10-dependent mechanism.


Subject(s)
Arachidonic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Administration, Oral , Animals , Animals, Newborn , Arachidonic Acid/administration & dosage , Docosahexaenoic Acids/administration & dosage , Models, Animal , Swine , T-Lymphocytes/drug effects
17.
Clin Nutr ; 26(1): 107-16, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17000034

ABSTRACT

BACKGROUND & AIMS: Despite their efficacy in improving insulin sensitivity, thiazolidinediones (TZDs) are associated with a number of side effects (i.e. weight gain, hepatotoxicity, congestive heart failure) that have limited their use by millions of diabetic patients. We have investigated whether abscisic acid (ABA), a naturally occurring phytochemical with structural similarities to TZDs, could be used as an alternative to TZDs to improve glucose homeostasis. METHODS: We first examined whether ABA, similar to TZDs, activates PPARgamma in vitro. We next determined the lowest effective dose of dietary ABA (100 mg/kg) and assessed its effect on glucose tolerance, obesity-related inflammation, and mRNA expression of PPARgamma and its responsive genes in white adipose tissue (WAT) of db/db mice fed high-fat diets. RESULTS: We found that ABA induced transactivation of PPARgamma in 3T3-L1 pre-adipocytes in vitro. Dietary ABA-supplementation for 36 days decreased fasting blood glucose concentrations, ameliorated glucose tolerance, and increased mRNA expression of PPARgamma and its responsive genes (i.e., adiponectin, aP2, and CD36) in WAT. We also found that adipocyte hypertrophy, tumor necrosis factor-alpha (TNF-alpha) expression, and macrophage infiltration in WAT were significantly attenuated in ABA-fed mice. CONCLUSIONS: These findings suggest that ABA could be used as a nutritional intervention against type II diabetes and obesity-related inflammation.


Subject(s)
Abscisic Acid/administration & dosage , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/prevention & control , Inflammation/prevention & control , PPAR gamma/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Tolerance Test , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred Strains , Obesity/complications , PPAR gamma/drug effects , PPAR gamma/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Thiazolidinediones
18.
Clin Nutr ; 25(6): 871-85, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17052808

ABSTRACT

Over recent years, obesity rates and the onset of obesity-induced chronic diseases have risen dramatically. The more we learn about the physiological and morphological changes that occur during obesity, the more it is becoming clear that obesity-related disorders can be traced back to adipocyte hypertrophy and inflammation at white adipose tissue (WAT). To combat this problem, the body has developed a regulatory system specifically designed at mediating the systemic response to obesity, utilizing free fatty acids (FFAs) and their metabolites as nutrient messengers to signal adaptations from peripheral tissues. These messages are predominantly interceded through the peroxisome proliferator-activated receptors (PPARs), a family of ligand-induced transcription factors that serve as a net of lipid sensors throughout the body. Understanding how and why nutrients, nutrient derivatives and metabolites exert their physiological effects are the key goals in the study of molecular nutrition. By learning about the mechanisms and tissue-specific effects of endogenous PPAR ligands and expanding our knowledge of the body's integrated homeostatic system, we will significantly increase our odds of designing safe and effective preventive and therapeutic interventions that keep us one step ahead of obesity-related diseases.


Subject(s)
Adipocytes/cytology , Lipid Metabolism , Obesity/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Transcription Factors/physiology , Animals , Cell Division , Fatty Acids, Nonesterified/metabolism , Humans , Inflammation , Metabolic Syndrome/metabolism , Nutritional Physiological Phenomena , Organ Specificity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...