Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Nutr ; 106(6): 878-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21736821

ABSTRACT

The goal of the present study was to elucidate the mechanisms of immunoregulation by which dietary punicic acid (PUA) prevents or ameliorates experimental inflammatory bowel disease (IBD). The expression of PPARγ and δ, their responsive genes and pro-inflammatory cytokines was assayed in the colonic mucosa. Immune cell-specific PPARγ null, PPARδ knockout and wild-type mice were treated with PUA and challenged with 2·5 % dextran sodium sulphate (DSS). The prophylactic efficacy of PUA was examined in an IL-10(-/-) model of IBD. The effect of PUA on the regulatory T-cell (Treg) compartment was also examined in mice with experimental IBD. PUA ameliorated spontaneous pan-enteritis in IL-10(-/-) mice and DSS colitis, up-regulated Foxp3 expression in Treg and suppressed TNF-α, but the loss of functional PPARγ or δ impaired these anti-inflammatory effects. At the cellular level, the macrophage-specific deletion of PPARγ caused a complete abrogation of the protective effect of PUA, whereas the deletion of PPARδ or intestinal epithelial cell-specific PPARγ decreased its anti-inflammatory efficacy. We provide in vivo molecular evidence demonstrating that PUA ameliorates experimental IBD by regulating macrophage and T-cell function through PPARγ- and δ-dependent mechanisms.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Linolenic Acids/pharmacology , PPAR delta/metabolism , PPAR gamma/metabolism , Animal Feed , Animals , Anti-Inflammatory Agents/pharmacology , Gene Deletion , Inflammation , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology
2.
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
3.
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
SELECTION OF CITATIONS
SEARCH DETAIL