Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 570425, 2021.
Article in English | MEDLINE | ID: mdl-33732230

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR)-δ is a nuclear receptor that functions to maintain metabolic homeostasis, regulate cell growth, and limit the development of excessive inflammation during immune responses. Previously, we reported that PPAR-δ-deficient mice develop a more severe clinical course of experimental autoimmune encephalomyelitis (EAE); however, it was difficult to delineate the role that microglia played in this disease phenotype since PPAR-δ-deficient mice exhibited a number of immune defects that enhanced CNS inflammation upstream of microglia activation. Here, we specifically investigated the role of PPAR-δ in microglia during EAE by using mice where excision of a floxed Ppard allele was driven by expression of a tamoxifen (TAM)-inducible CX3C chemokine receptor 1 promoter-Cre recombinase transgene (Cx3cr1CreERT2: Ppardfl/fl). We observed that by 30 days of TAM treatment, Cx3cr1CreERT2: Ppardfl/fl mice exhibited Cre-mediated deletion primarily in microglia and this was accompanied by efficient knockdown of Ppard expression in these cells. Upon induction of EAE, TAM-treated Cx3cr1CreERT2: Ppardfl/fl mice presented with an exacerbated course of disease compared to TAM-treated Ppardfl/fl controls. Histopathological and magnetic resonance (MR) studies on the spinal cord and brains of EAE mice revealed increased Iba-1 immunoreactivity, axonal injury and CNS tissue loss in the TAM-treated Cx3cr1CreERT2: Ppardfl/fl group compared to controls. In early EAE, a time when clinical scores and the infiltration of CD45+ leukocytes was equivalent between Cx3cr1CreERT2: Ppardfl/fl and Ppardfl/fl mice, Ppard-deficient microglia exhibited a more reactive phenotype as evidenced by a shorter maximum process length and lower expression of genes associated with a homeostatic microglia gene signature. In addition, Ppard-deficient microglia exhibited increased expression of genes associated with reactive oxygen species generation, phagocytosis and lipid clearance, M2-activation, and promotion of inflammation. Our results therefore suggest that PPAR-δ has an important role in microglia in limiting bystander tissue damage during neuroinflammation.


Subject(s)
Axons/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Microglia/immunology , Microglia/metabolism , PPAR delta/deficiency , Animals , Axons/pathology , Cells, Cultured , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Lymphocyte Activation/immunology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Microglia/pathology , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Cell Death Differ ; 27(9): 2668-2680, 2020 09.
Article in English | MEDLINE | ID: mdl-32313198

ABSTRACT

The incidence of nonmelanoma skin cancer (NMSC) has been increasing worldwide. Most studies have highlighted the importance of cancer-associated fibroblasts (CAFs) in NMSC progression. However much less is known about the communication between normal fibroblasts and epithelia; disruption of this communication affects tumor initiation and the latency period in the emergence of tumors. Delineating the mechanism that mediates this epithelial-mesenchymal communication in NMSC could identify more effective targeted therapies. The nuclear receptor PPARß/δ in fibroblasts has been shown to modulate adjacent epithelial cell behavior, however, its role in skin tumorigenesis remains unknown. Using chemically induced skin carcinogenesis, we showed that FSPCre-Pparb/dex4 mice, whose Pparb/d gene was selectively deleted in fibroblasts, had delayed emergence and reduced tumor burden compared with control mice (Pparb/dfl/fl). However, FSPCre-Pparb/dex4-derived tumors showed increased proliferation, with no difference in differentiation, suggesting delayed tumor initiation. Network analysis revealed a link between dermal Pparb/d and TGF-ß1 with epidermal NRF2 and Nox4. In vitro investigations showed that PPARß/δ deficiency in fibroblasts increased epidermal Nox4-derived H2O2 production, which triggered an NRF2-mediated antioxidant response. We further showed that H2O2 upregulated NRF2 mRNA via the B-Raf-MEK1/2 pathway. The enhanced NRF2 response altered the activities of PTEN, Src, and AKT. In vivo, we detected the differential phosphorylation profiles of B-Raf, MEK1/2, PTEN, Src, and AKT in the vehicle-treated and chemically treated epidermis of FSPCre-Pparb/dex4 mice compared with that in Pparb/dfl/fl mice, prior to the first appearance of tumors in Pparb/dfl/fl. Our study revealed a role for fibroblast PPARß/δ in the epithelial-mesenchymal communication involved in cellular redox homeostasis.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , PPAR delta/deficiency , PPAR-beta/deficiency , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Epidermis/pathology , Gene Regulatory Networks , Glycoproteins/metabolism , Keratinocytes/metabolism , Kinetics , Melanoma/metabolism , Melanoma/pathology , Mice, Transgenic , NADPH Oxidase 4/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction , Skin Neoplasms/genetics , Transforming Growth Factor beta1/metabolism , Tumor Burden
3.
Diabetes ; 65(10): 3185-99, 2016 10.
Article in English | MEDLINE | ID: mdl-27486236

ABSTRACT

Fibroblast growth factor 21 (FGF21), a peptide hormone with pleiotropic effects on carbohydrate and lipid metabolism, is considered a target for the treatment of diabetes. We investigated the role of peroxisome proliferator-activated receptor (PPAR) ß/δ deficiency in hepatic FGF21 regulation. Increased Fgf21 expression was observed in the livers of PPARß/δ-null mice and in mouse primary hepatocytes when this receptor was knocked down by small interfering RNA (siRNA). Increased Fgf21 was associated with enhanced protein levels in the heme-regulated eukaryotic translation initiation factor 2α (eIF2α) kinase (HRI). This increase caused enhanced levels of phosphorylated eIF2α and activating transcription factor (ATF) 4, which is essential for Fgf21-induced expression. siRNA analysis demonstrated that HRI regulates Fgf21 expression in primary hepatocytes. Enhanced Fgf21 expression attenuated tunicamycin-induced endoplasmic reticulum stress, as demonstrated by using a neutralizing antibody against FGF21. Of note, increased Fgf21 expression in mice fed a high-fat diet or hepatocytes exposed to palmitate was accompanied by reduced PPARß/δ and activation of the HRI-eIF2α-ATF4 pathway. Moreover, pharmacological activation of HRI increased Fgf21 expression and reduced lipid-induced hepatic steatosis and glucose intolerance, but these effects were not observed in Fgf21-null mice. Overall, these findings suggest that HRI is a potential target for regulating hepatic FGF21 levels.


Subject(s)
Fibroblast Growth Factors/metabolism , Liver/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Fibroblast Growth Factors/genetics , Immunoblotting , Male , Mice , Mice, Knockout , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , Phosphorylation/genetics , Phosphorylation/physiology , Reverse Transcriptase Polymerase Chain Reaction , eIF-2 Kinase/genetics
4.
Arterioscler Thromb Vasc Biol ; 36(8): 1534-48, 2016 08.
Article in English | MEDLINE | ID: mdl-27283742

ABSTRACT

OBJECTIVE: Drug-eluting coronary stents reduce restenosis rate and late lumen loss compared with bare-metal stents; however, drug-eluting coronary stents may delay vascular healing and increase late stent thrombosis. The peroxisome proliferator-activated receptor-delta (PPARδ) exhibits actions that could favorably influence outcomes after drug-eluting coronary stents placement. APPROACH AND RESULTS: Here, we report that PPARδ ligand-coated stents strongly reduce the development of neointima and luminal narrowing in a rabbit model of experimental atherosclerosis. Inhibition of inflammatory gene expression and vascular smooth muscle cell (VSMC) proliferation and migration, prevention of thrombocyte activation and aggregation, and proproliferative effects on endothelial cells were identified as key mechanisms for the prevention of restenosis. Using normal and PPARδ-depleted VSMCs, we show that the observed effects of PPARδ ligand GW0742 on VSMCs and thrombocytes are PPARδ receptor dependent. PPARδ ligand treatment induces expression of pyruvate dehydrogenase kinase isozyme 4 and downregulates the glucose transporter 1 in VSMCs, thus impairing the ability of VSMCs to provide the increased energy demands required for growth factor-stimulated proliferation and migration. CONCLUSIONS: In contrast to commonly used drugs for stent coating, PPARδ ligands not only inhibit inflammatory response and proliferation of VSMCs but also prevent thrombocyte activation and support vessel re-endothelialization. Thus, pharmacological PPARδ activation could be a promising novel strategy to improve drug-eluting coronary stents outcomes.


Subject(s)
Angioplasty, Balloon/instrumentation , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cardiovascular Agents/administration & dosage , Drug-Eluting Stents , PPAR delta/agonists , Steroids/administration & dosage , Thrombosis/prevention & control , Angioplasty, Balloon/adverse effects , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blood Platelets/drug effects , Blood Platelets/metabolism , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Artery Disease/therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima , PPAR delta/deficiency , PPAR delta/genetics , PPAR delta/metabolism , Platelet Activation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Rats, Sprague-Dawley , Re-Epithelialization/drug effects , Recurrence , Signal Transduction/drug effects , Thrombosis/etiology , Thrombosis/metabolism , Thrombosis/pathology , Time Factors
5.
J Allergy Clin Immunol ; 135(6): 1625-35.e5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25556996

ABSTRACT

BACKGROUND: Remodeling of quiescent vessels with increases in permeability, vasodilatation, and edema are hallmarks of inflammatory disorders. Factors involved in this type of remodeling represent potential therapeutic targets. OBJECTIVES: We investigated whether the nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) ß/δ, a regulator of metabolism, fibrosis, and skin homeostasis, is involved in regulation of this type of remodeling. METHODS: Wild-type and various Pparb/d mutant mice were used to monitor dermal acute vascular hyperpermeability (AVH) and passive systemic anaphylaxis-induced hypothermia and edema. PPARß/δ-dependent kinase activation and remodeling of endothelial cell-cell junctions were addressed by using human endothelial cells. RESULTS: AVH and dilatation of dermal microvessels stimulated by vascular endothelial growth factor A, histamine, and thrombin are severely compromised in PPARß/δ-deficient mice. Selective deletion of the Pparb/d-encoding gene in endothelial cells in vivo similarly limits dermal AVH and vasodilatation, providing evidence that endothelial PPARß/δ is the major player in regulating acute dermal microvessel remodeling. Furthermore, endothelial PPARß/δ regulatory functions are not restricted to the skin vasculature because its deletion in the endothelium, but not in smooth muscle cells, also leads to reduced systemic anaphylaxis, the most severe form of allergic reaction, in which an acute vascular response plays a key role. PPARß/δ-dependent AVH activation likely involves the activation of mitogen-activated protein kinase and Akt pathways and leads to downstream destabilization of endothelial cell-cell junctions. CONCLUSION: These results unveil not only a novel function of PPARß/δ as a direct regulator of acute vessel permeability and dilatation but also provide evidence that antagonizing PPARß/δ represents an important strategy to consider for moderating diseases with altered endothelial integrity, such as acute inflammatory and allergic disorders.


Subject(s)
Anaphylaxis/immunology , Capillary Permeability/immunology , Endothelial Cells/immunology , PPAR delta/immunology , PPAR-beta/immunology , Skin/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Capillary Permeability/drug effects , Edema/genetics , Edema/immunology , Edema/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Expression Regulation , Histamine/pharmacology , Hypothermia/genetics , Hypothermia/immunology , Hypothermia/pathology , Intercellular Junctions/drug effects , Intercellular Junctions/immunology , Intercellular Junctions/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Skin/blood supply , Skin/drug effects , Skin/pathology , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
6.
Diabetologia ; 57(10): 2126-35, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25063273

ABSTRACT

AIM/HYPOTHESIS: Endoplasmic reticulum (ER) stress, which is involved in the link between inflammation and insulin resistance, contributes to the development of type 2 diabetes mellitus. In this study, we assessed whether peroxisome proliferator-activated receptor (PPAR)ß/δ prevented ER stress-associated inflammation and insulin resistance in skeletal muscle cells. METHODS: Studies were conducted in mouse C2C12 myotubes, in the human myogenic cell line LHCN-M2 and in skeletal muscle from wild-type and PPARß/δ-deficient mice and mice exposed to a high-fat diet. RESULTS: The PPARß/δ agonist GW501516 prevented lipid-induced ER stress in mouse and human myotubes and in skeletal muscle of mice fed a high-fat diet. PPARß/δ activation also prevented thapsigargin- and tunicamycin-induced ER stress in human and murine skeletal muscle cells. In agreement with this, PPARß/δ activation prevented ER stress-associated inflammation and insulin resistance, and glucose-intolerant PPARß/δ-deficient mice showed increased phosphorylated levels of inositol-requiring 1 transmembrane kinase/endonuclease-1α in skeletal muscle. Our findings demonstrate that PPARß/δ activation prevents ER stress through the activation of AMP-activated protein kinase (AMPK), and the subsequent inhibition of extracellular-signal-regulated kinase (ERK)1/2 due to the inhibitory crosstalk between AMPK and ERK1/2, since overexpression of a dominant negative AMPK construct (K45R) reversed the effects attained by PPARß/δ activation. CONCLUSIONS/INTERPRETATION: Overall, these findings indicate that PPARß/δ prevents ER stress, inflammation and insulin resistance in skeletal muscle cells by activating AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/physiology , Inflammation/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , PPAR delta/physiology , PPAR-beta/physiology , Animals , Cell Line , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress/genetics , Humans , In Vitro Techniques , Inflammation/etiology , Inflammation/genetics , Insulin Resistance/genetics , Mice , Muscle Fibers, Skeletal/metabolism , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics
7.
J Biol Chem ; 289(29): 20102-19, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24898257

ABSTRACT

Endoplasmic reticulum (ER) stress and ER stress-associated unfolded protein response (UPR) can promote cancer cell survival, but it remains unclear whether they can influence oncogene-induced senescence. The present study examined the role of ER stress in senescence using oncogene-dependent models. Increased ER stress attenuated senescence in part by up-regulating phosphorylated protein kinase B (p-AKT) and decreasing phosphorylated extracellular signal-regulated kinase (p-ERK). A positive feed forward loop between p-AKT, ER stress, and UPR was discovered whereby a transient increase of ER stress caused reduced senescence and promotion of tumorigenesis. Decreased ER stress was further correlated with increased senescence in both mouse and human tumors. Interestingly, H-RAS-expressing Pparß/δ null cells and tumors having increased cell proliferation exhibited enhanced ER stress, decreased cellular senescence, and/or enhanced tumorigenicity. Collectively, these results demonstrate a new role for ER stress and UPR that attenuates H-RAS-induced senescence and suggest that PPARß/δ can repress this oncogene-induced ER stress to promote senescence in accordance with its role as a tumor modifier that suppresses carcinogenesis.


Subject(s)
Cellular Senescence/genetics , Cellular Senescence/physiology , Endoplasmic Reticulum Stress , Genes, ras , PPAR delta/metabolism , PPAR-beta/metabolism , Activating Transcription Factor 4/genetics , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression , Gene Knockdown Techniques , Genes, p53 , Heat-Shock Proteins/genetics , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Models, Biological , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Regulatory Factor X Transcription Factors , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Unfolded Protein Response
8.
J Lipid Res ; 54(7): 1949-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23606724

ABSTRACT

The surface of lipid droplets (LDs) in various cell types is coated with perilipin proteins encoded by the Plin genes. Perilipins regulate LD metabolism by selectively recruiting lipases and other proteins to LDs. We have studied the expression of perilipins in mouse muscle. The glycolytic fiber-enriched gastrocnemius muscle expresses predominantly Plin2-4. The oxidative fiber-enriched soleus muscle expresses Plin2-5. Expression of Plin2 and Plin4-5 is elevated in gastrocnemius and soleus muscles from mice fed a high-fat diet. This effect is preserved in peroxisome proliferator-activated receptor (PPAR)α-deficient mice. Mouse muscle derived C2C12 cells differentiated into glycolytic fibers increase transcription of these Plins when exposed to various long chain fatty acids (FAs). To understand how FAs regulate Plin genes, we used specific activators and antagonists against PPARs, Plin promoter reporter assays, chromatin immunoprecipitation, siRNA, and animal models. Our analyses demonstrate that FAs require PPARδ to induce transcription of Plin4 and Plin5. We further identify a functional PPAR binding site in the Plin5 gene and establish Plin5 as a novel direct PPARδ target in muscle. Our study reveals that muscle cells respond to elevated FAs by increasing transcription of several perilipin LD-coating proteins. This induction renders the muscle better equipped to sequester incoming FAs into cytosolic LDs.


Subject(s)
Fatty Acids/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , PPAR delta/metabolism , Animals , Binding Sites/drug effects , Cells, Cultured , Fatty Acids/administration & dosage , Gene Silencing/drug effects , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , PPAR delta/chemistry , PPAR delta/deficiency , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
J Neuroinflammation ; 10: 20, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23374874

ABSTRACT

BACKGROUND: Palmitoylethanolamide (PEA) is an endogenous fatty acid amide displaying anti-inflammatory and analgesic actions. Moreover, several data have suggested that PEA reduced inflammation and tissue injury associated with spinal cord trauma and showed a regulatory role for peroxisome proliferator-activated receptor (PPAR)-α signaling in the neuroprotective effect of PEA. However, several other mechanisms could explain the anti-inflammatory and anti-hyperalgesic effects of PEA, including the activation of PPAR-δ and PPAR-γ. The aim of the present study was to carefully investigate the exact contribution of PPAR-δ and PPAR-γ in addition to PPAR-α, in the protective effect of PEA on secondary inflammatory damage associated with an experimental model of spinal cord injury (SCI). METHODS: SCI was induced in mice through a spinal cord compression by the application of vascular clips (force of 24 g) to the dura via a four-level T5 to T8 laminectomy, and PEA (10 mg/kg, intraperitoneally, 1 and 6 hours after SCI) was injected into wildtype mice and into mice lacking PPAR-α (PPAR-αKO). To deepen the ability of specific PPAR-δ and PPAR-γ antagonists to reverse the effect of PEA, mice were administered GSK0660 or GW9662, 30 minutes before PEA injection. RESULTS: Genetic ablation of PPAR-α in mice exacerbated spinal cord damage, while PEA-induced neuroprotection seemed be abolished in PPARαKO mice. Twenty-four hours after spinal cord damage, immunohistological and biochemical studies were performed on spinal cord tissue. Our results indicate that PPAR-δ and PPAR-γ also mediated the protection induced by PEA. In particular, PEA was less effective in PPAR-αKO, GSK0660-treated or GW9662-pretreated mice, as evaluated by the degree of spinal cord inflammation and tissue injury, neutrophil infiltration, proinflammmatory cytokine, inducible nitric oxide synthase expression and motor function. PEA is also able to restore PPAR-δ and PPAR-γ expression in spinal cord tissue. CONCLUSION: This study indicates that PPAR-δ and PPAR-γ can also contribute to the anti-inflammatory activity of PEA in SCI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Endocannabinoids/therapeutic use , Ethanolamines/therapeutic use , Neuroprotective Agents/therapeutic use , PPAR delta/deficiency , PPAR gamma/deficiency , Palmitic Acids/therapeutic use , Spinal Cord Injuries/prevention & control , Amides , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Endocannabinoids/metabolism , Ethanolamines/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Neuroprotective Agents/metabolism , PPAR delta/genetics , PPAR gamma/genetics , Palmitic Acids/metabolism , Random Allocation , Spinal Cord Injuries/genetics , Time Factors
10.
Nat Med ; 18(9): 1350-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22902876

ABSTRACT

Stem-cell function is an exquisitely regulated process. Thus far, the contribution of metabolic cues to stem-cell function has not been well understood. Here we identify a previously unknown promyelocytic leukemia (PML)­peroxisome proliferator-activated receptor δ (PPAR-δ)­fatty-acid oxidation (FAO) pathway for the maintenance of hematopoietic stem cells (HSCs). We have found that loss of PPAR-δ or inhibition of mitochondrial FAO induces loss of HSC maintenance, whereas treatment with PPAR-δ agonists improved HSC maintenance. PML exerts its essential role in HSC maintenance through regulation of PPAR signaling and FAO. Mechanistically, the PML­PPAR-δ­FAO pathway controls the asymmetric division of HSCs. Deletion of Ppard or Pml as well as inhibition of FAO results in the symmetric commitment of HSC daughter cells, whereas PPAR-δ activation increased asymmetric cell division. Thus, our findings identify a metabolic switch for the control of HSC cell fate with potential therapeutic implications.


Subject(s)
Cell Division/physiology , Fatty Acids/metabolism , Hematopoietic Stem Cells/physiology , Metabolic Networks and Pathways/physiology , Models, Biological , Nuclear Proteins/metabolism , PPAR delta/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Colony-Forming Units Assay , Epoxy Compounds/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Hematopoietic Stem Cells/metabolism , Immunoprecipitation , Metabolic Networks and Pathways/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Oxidation-Reduction/drug effects , PPAR delta/agonists , PPAR delta/deficiency , Promyelocytic Leukemia Protein , Real-Time Polymerase Chain Reaction , Thiazoles/pharmacology
11.
Diabetes ; 60(7): 1990-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21617181

ABSTRACT

OBJECTIVE: It has been suggested that interleukin (IL)-6 is one of the mediators linking obesity-derived chronic inflammation with insulin resistance through activation of STAT3, with subsequent upregulation of suppressor of cytokine signaling 3 (SOCS3). We evaluated whether peroxisome proliferator-activated receptor (PPAR)-ß/-δ prevented activation of the IL-6-STAT3-SOCS3 pathway and insulin resistance in adipocytes. RESEARCH DESIGN AND METHODS: Adipocytes and white adipose tissue from wild-type and PPAR-ß/-δ-null mice were used to evaluate the effect of PPAR-ß/-δ on the IL-6-STAT3-SOCS3 pathway. RESULTS: First, we observed that the PPAR-ß/-δ agonist GW501516 prevented both IL-6-dependent reduction in insulin-stimulated Akt phosphorylation and glucose uptake in adipocytes. In addition, this drug treatment abolished IL-6-induced SOCS3 expression in differentiated 3T3-L1 adipocytes. This effect was associated with the capacity of the drug to prevent IL-6-induced STAT3 phosphorylation on Tyr(705) and Ser(727) residues in vitro and in vivo. Moreover, GW501516 prevented IL-6-dependent induction of extracellular signal-related kinase (ERK)1/2, a serine-threonine-protein kinase involved in serine STAT3 phosphorylation. Furthermore, in white adipose tissue from PPAR-ß/-δ-null mice, STAT3 phosphorylation (Tyr(705) and Ser(727)), STAT3 DNA-binding activity, and SOCS3 protein levels were higher than in wild-type mice. Several steps in STAT3 activation require its association with heat shock protein 90 (Hsp90), which was prevented by GW501516 as revealed in immunoprecipitation studies. Consistent with this finding, the STAT3-Hsp90 association was enhanced in white adipose tissue from PPAR-ß/-δ-null mice compared with wild-type mice. CONCLUSIONS: Collectively, our findings indicate that PPAR-ß/-δ activation prevents IL-6-induced STAT3 activation by inhibiting ERK1/2 and preventing the STAT3-Hsp90 association, an effect that may contribute to the prevention of cytokine-induced insulin resistance in adipocytes.


Subject(s)
Adipocytes/metabolism , Insulin/physiology , PPAR delta/metabolism , PPAR-beta/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/metabolism , Animals , Glucose/metabolism , HSP90 Heat-Shock Proteins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/pharmacology , Male , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , PPAR delta/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Rats , Thiazoles/pharmacology
12.
Arterioscler Thromb Vasc Biol ; 31(3): 574-81, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21205987

ABSTRACT

OBJECTIVE: To investigate the effects of peroxisome proliferator-activated receptor (PPAR)δ in the cerebral vasculature following stroke-induced brain injury. METHODS AND RESULTS: Here, we report a novel finding that selective PPARδ genetic deletion in vascular smooth muscle cells (VSMCs) resulted in increased cerebrovascular permeability and brain infarction in mice after middle cerebral artery occlusion (MCAO). Mechanistically, we revealed for the first time that PPARδ expression is reduced, but matrix metalloproteinase (MMP)-9 activity is increased in cultured VSMCs after oxygen-glucose deprivation and also in the cerebral cortex of mice following MCAO. Moreover, gain- and loss of PPARδ function in VSMCs significantly reduces and increases oxygen-glucose deprivation-induced MMP-9 activity, respectively. We have further identified that MMP-9 is a direct target of PPARδ-mediated transrepression by chromatin immunoprecipitation and PPARδ transcriptional activity assays. Furthermore, inhibition of MMP-9 activity by lentiviral MMP-9 short hairpin RNA effectively improves cerebrovascular permeability and reduces brain infarction in VSMC-selective PPARδ conditional knockout mice after MCAO. CONCLUSIONS: Our data demonstrate that PPARδ in VSMCs can prevent ischemic brain injury by inhibition of MMP-9 activation and attenuation of postischemic inflammation. The pharmacological activation of PPARδ may provide a new therapeutic strategy to treat stroke-induced vascular and neuronal damage.


Subject(s)
Capillary Permeability , Cerebral Cortex/blood supply , Infarction, Middle Cerebral Artery/metabolism , Muscle, Smooth, Vascular/metabolism , PPAR delta/metabolism , Reperfusion Injury/prevention & control , Animals , Binding Sites , Cell Hypoxia , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chromatin Immunoprecipitation , Disease Models, Animal , Glucose/deficiency , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Inflammation/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Oxygen/metabolism , PPAR delta/deficiency , PPAR delta/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Time Factors , Transcriptional Activation , Transfection , Up-Regulation
13.
Brain Res ; 1376: 101-12, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21192919

ABSTRACT

Multiple sclerosis (MS) is a neurological disorder that affects more than a million people worldwide. The etiology of MS is not known and there is no medical treatment that can cure MS. Earlier studies have shown that peroxisome proliferator-activated receptor (PPARs) agonists ameliorate MS-like disease in experimental allergic encephalomyelitis (EAE). In this study we have used PPARδ deficient mice to determine its physiological role in the regulation of CNS EAE and MS. We found that PPARδ(-/-) mice develop EAE with similar day of onset and disease incidence compared to C57BL/6 wild type mice. Interestingly, both male and female PPARδ(-/-) mice showed prolonged EAE with resistance to remission and recovery. PPARδ(-/-) mice with EAE expressed elevated levels of IFNγ and IL-17 along with IL-12p35 and IL-12p40 in the brain and spleen. PPARδ(-/-) mice also developed augmented neural antigen-specific Th1/Th17 responses and impaired Th2/Treg responses compared to wild type mice. These findings indicate that PPARδ(-/-) mice develop prolonged EAE in association with augmented Th1/Th17 responses, suggesting a critical physiological role for PPARδ in the remission and recovery of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , PPAR delta/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Cell Separation , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/deficiency , PPAR delta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology , Spinal Cord/pathology
14.
Circ Res ; 106(11): 1712-21, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20378851

ABSTRACT

RATIONALE: Although dietary fatty acids are a major fuel for the heart, little is known about the direct effects of dietary fatty acids on gene regulation in the intact heart. OBJECTIVE: To study the effect of dietary fatty acids on cardiac gene expression and explore the functional consequences. METHODS AND RESULTS: Oral administration of synthetic triglycerides composed of one single fatty acid altered cardiac expression of numerous genes, many of which are involved in the oxidative stress response. The gene most significantly and consistently upregulated by dietary fatty acids encoded Angiopoietin-like protein (Angptl)4, a circulating inhibitor of lipoprotein lipase expressed by cardiomyocytes. Induction of Angptl4 by the fatty acid linolenic acid was specifically abolished in peroxisome proliferator-activated receptor (PPAR)beta/delta(-/-) and not PPARalpha(-/-) mice and was blunted on siRNA-mediated PPARbeta/delta knockdown in cultured cardiomyocytes. Consistent with these data, linolenic acid stimulated binding of PPARbeta/delta but not PPARalpha to the Angptl4 gene. Upregulation of Angptl4 resulted in decreased cardiac uptake of plasma triglyceride-derived fatty acids and decreased fatty acid-induced oxidative stress and lipid peroxidation. In contrast, Angptl4 deletion led to enhanced oxidative stress in the heart, both after an acute oral fat load and after prolonged high fat feeding. CONCLUSIONS: Stimulation of cardiac Angptl4 gene expression by dietary fatty acids and via PPARbeta/delta is part of a feedback mechanism aimed at protecting the heart against lipid overload and consequently fatty acid-induced oxidative stress.


Subject(s)
Angiopoietins/metabolism , Cardiomyopathies/prevention & control , Dietary Fats/metabolism , Fatty Acids, Unsaturated/metabolism , Myocardium/metabolism , Oxidative Stress , PPAR delta/metabolism , PPAR-beta/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins/deficiency , Angiopoietins/genetics , Animals , Animals, Newborn , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cells, Cultured , Cytoprotection , Dietary Fats/administration & dosage , Dietary Fats/blood , Dietary Fats/toxicity , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/toxicity , Feedback, Physiological , Linoleic Acid/metabolism , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oleic Acid/metabolism , Oxidative Stress/genetics , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , RNA Interference , Time Factors , Up-Regulation , alpha-Linolenic Acid/metabolism
15.
Immunology ; 130(4): 572-88, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20406305

ABSTRACT

Multiple sclerosis (MS) is a neurological disorder that affects more than a million people world-wide. The aetiology of MS is not known and there is no medical treatment available that can cure MS. Experimental autoimmune encephalomyelitis (EAE) is a T-cell-mediated autoimmune disease model of MS. The pathogenesis of EAE/MS is a complex process involving activation of immune cells, secretion of inflammatory cytokines and destruction of myelin sheath in the central nervous system (CNS). Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptor transcription factors that regulate cell growth, differentiation and homeostasis. PPAR agonists have been used in the treatment of obesity, diabetes, cancer and inflammation. We and others have shown that PPARgamma, alpha and delta agonists inhibit CNS inflammation and demyelination in the EAE model of MS. In this study we show that the PPARdelta agonists GW501516 and L165041 ameliorate MOGp35-55-induced EAE in C57BL/6 mice by blocking interferon (IFN)-gamma and interleukin (IL)-17 production by T helper type 1 (Th1) and Th17 cells. The inhibition of EAE by PPARdelta agonists was also associated with a decrease in IL-12 and IL-23 and an increase in IL-4 and IL-10 expression in the CNS and lymphoid organs. These findings indicate that PPARdelta agonists modulate Th1 and Th17 responses in EAE and suggest their use in the treatment of MS and other autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , PPAR delta/agonists , Phenoxyacetates/pharmacology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Thiazoles/pharmacology , Animals , Cell Polarity , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/deficiency , PPAR delta/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , Th1 Cells/cytology , Th1 Cells/drug effects
16.
Circ Res ; 106(5): 911-9, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20075336

ABSTRACT

RATIONALE: Peroxisome proliferator-activated receptors (PPARs) (alpha, gamma, and delta/beta) are nuclear hormone receptors and ligand-activated transcription factors that serve as key determinants of myocardial fatty acid metabolism. Long-term cardiomyocyte-restricted PPARdelta deficiency in mice leads to depressed myocardial fatty acid oxidation, bioenergetics, and premature death with lipotoxic cardiomyopathy. OBJECTIVE: To explore the essential role of PPARdelta in the adult heart. METHODS AND RESULTS: We investigated the consequences of inducible short-term PPARdelta knockout in the adult mouse heart. In addition to a substantial transcriptional downregulation of lipid metabolic proteins, short-term PPARdelta knockout in the adult mouse heart attenuated cardiac expression of both Cu/Zn superoxide dismutase and manganese superoxide dismutase, leading to increased oxidative damage to the heart. Moreover, expression of key mitochondrial biogenesis determinants such as PPARgamma coactivator-1 were substantially decreased in the short-term PPARdelta deficient heart, concomitant with a decreased mitochondrial DNA copy number. Rates of palmitate and glucose oxidation were markedly depressed in cardiomyocytes of PPARdelta knockout hearts. Consequently, PPARdelta deficiency in the adult heart led to depressed cardiac performance and cardiac hypertrophy. CONCLUSIONS: PPARdelta is an essential regulator of cardiac mitochondrial protection and biogenesis and PPARdelta activation can be a potential therapeutic target for cardiac disorders.


Subject(s)
Energy Metabolism/genetics , Lipid Metabolism/genetics , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , PPAR delta/metabolism , RNA, Messenger/biosynthesis , Transcription, Genetic , Aging , Animals , Antioxidants/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cells, Cultured , DNA, Mitochondrial/metabolism , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress/genetics , PPAR delta/deficiency , PPAR delta/genetics , Palmitic Acid/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism
17.
J Lipid Res ; 51(6): 1370-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19965574

ABSTRACT

The peroxisome proliferator-activated receptor delta (PPARdelta) is implicated in regulation of mitochondrial processes in a number of tissues, and PPARdelta activation is associated with decreased susceptibility to ectopic lipid deposition and metabolic disease. Here, we show that PPARdelta is the PPAR subtype expressed at the highest level in insulinoma cells and rat pancreatic islets. Furthermore, PPARdelta displays high transcriptional activity and acts in pronounced synergy with retinoid-X-receptor (RXR). Interestingly, unsaturated fatty acids mimic the effects of synthetic PPARdelta agonists. Using short hairpin RNA-mediated knockdown, we demonstrate that the ability of unsaturated fatty acids to stimulate fatty acid metabolism is dependent on PPARdelta. Activation of PPARdelta increases the fatty acid oxidation capacity in INS-1E beta-cells, enhances glucose-stimulated insulin secretion (GSIS) from islets, and protects GSIS against adverse effects of prolonged fatty acid exposure. The presented results indicate that the nuclear receptor PPARdelta is a fatty acid sensor that adapts beta-cell mitochondrial function to long-term changes in unsaturated fatty acid levels. As maintenance of mitochondrial metabolism is essential to preserve beta-cell function, these data indicate that dietary or pharmacological activation of PPARdelta and RXR may be beneficial in the prevention of beta-cell dysfunction.


Subject(s)
Fatty Acids/metabolism , Fatty Acids/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , PPAR delta/metabolism , Animals , Cell Line, Tumor , Clone Cells , Drug Synergism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Mitochondria/drug effects , Oleic Acid/pharmacology , Oxidation-Reduction , PPAR delta/agonists , PPAR delta/deficiency , PPAR delta/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Retinoid X Receptors/agonists , Retinoid X Receptors/metabolism , Transcription, Genetic/drug effects
18.
Physiol Genomics ; 41(1): 42-52, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20009009

ABSTRACT

Little is known about the role of the transcription factor peroxisome proliferator-activated receptor (PPAR) beta/delta in liver. Here we set out to better elucidate the function of PPARbeta/delta in liver by comparing the effect of PPARalpha and PPARbeta/delta deletion using whole genome transcriptional profiling and analysis of plasma and liver metabolites. In fed state, the number of genes altered by PPARalpha and PPARbeta/delta deletion was similar, whereas in fasted state the effect of PPARalpha deletion was much more pronounced, consistent with the pattern of gene expression of PPARalpha and PPARbeta/delta. Minor overlap was found between PPARalpha- and PPARbeta/delta-dependent gene regulation in liver. Pathways upregulated by PPARbeta/delta deletion were connected to innate immunity and inflammation. Pathways downregulated by PPARbeta/delta deletion included lipoprotein metabolism and various pathways related to glucose utilization, which correlated with elevated plasma glucose and triglycerides and reduced plasma cholesterol in PPARbeta/delta-/- mice. Downregulated genes that may underlie these metabolic alterations included Pklr, Fbp1, Apoa4, Vldlr, Lipg, and Pcsk9, which may represent novel PPARbeta/delta target genes. In contrast to PPARalpha-/- mice, no changes in plasma free fatty acid, plasma beta-hydroxybutyrate, liver triglycerides, and liver glycogen were observed in PPARbeta/delta-/- mice. Our data indicate that PPARbeta/delta governs glucose utilization and lipoprotein metabolism and has an important anti-inflammatory role in liver. Overall, our analysis reveals divergent roles of PPARalpha and PPARbeta/delta in regulation of gene expression in mouse liver.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Liver/metabolism , PPAR alpha/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Transcription, Genetic , Animals , Gene Deletion , Immunity/genetics , Inflammation/genetics , Metabolome/genetics , Mice , Oligonucleotide Array Sequence Analysis , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics
19.
Toxicol Sci ; 113(1): 27-36, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19748995

ABSTRACT

Ligand activation of peroxisome proliferator-activated receptor (PPAR)-beta/delta and inhibition of cyclooxygenase-2 (COX-2) activity by nonsteroidal anti-inflammatory drugs can attenuate skin tumorigenesis. There is also evidence that attenuation of skin tumorigenesis by inhibition of COX-2 activity occurs through PPARbeta/delta-independent mechanisms. The present study examined the hypothesis that combining ligand activation of PPARbeta/delta with inhibition of COX-2 activity will cooperatively inhibit chemically induced skin tumor progression using both in vivo and ex vivo models. A two-stage chemical carcinogenesis bioassay was performed in wild-type and Pparbeta/delta-null mice. After 22 weeks, cohorts of both mouse lines were divided into four experimental groups: (1) control, (2) topical application of the PPARbeta/delta ligand GW0742, (3) dietary administration of the COX-2 inhibitor nimesulide, or (4) both GW0742 and nimesulide. Ligand activation of PPARbeta/delta did not influence skin tumor progression, while a modest decrease in skin tumor multiplicity was observed with dietary nimesulide. Interestingly, the combined treatment of GW0742 and nimesulide increased the efficacy of the decrease in papilloma multiplicity for 6 weeks in wild-type mice, but this effect was not found at later time points and was not found in similarly treated Pparbeta/delta-null mice. Neoplastic keratinocyte lines cultured with GW0742 and nimesulide also exhibited enhanced inhibition of cell proliferation coincident with increased expression of Keratin messenger RNAs. Results from these studies support the hypothesis that combining ligand activation of PPARbeta/delta with inhibition of COX-2 activity can inhibit chemically induced skin tumor progression by modulating differentiation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , PPAR delta/agonists , PPAR-beta/agonists , Skin Neoplasms/prevention & control , Sulfonamides/pharmacology , Thiazoles/pharmacology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/prevention & control , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dinoprostone/metabolism , Disease Models, Animal , Female , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/pathology , Keratins/genetics , Keratoacanthoma/enzymology , Keratoacanthoma/prevention & control , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/deficiency , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/deficiency , PPAR-beta/genetics , PPAR-beta/metabolism , Papilloma/enzymology , Papilloma/prevention & control , RNA, Messenger/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Time Factors
20.
Nat Med ; 15(11): 1266-72, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19838202

ABSTRACT

Macrophages rapidly engulf apoptotic cells to limit the release of noxious cellular contents and to restrict autoimmune responses against self antigens. Although factors participating in recognition and engulfment of apoptotic cells have been identified, the transcriptional basis for the sensing and the silent disposal of apoptotic cells is unknown. Here we show that peroxisome proliferator-activated receptor-delta (PPAR-delta) is induced when macrophages engulf apoptotic cells and functions as a transcriptional sensor of dying cells. Genetic deletion of PPAR-delta decreases expression of opsonins such as complement component-1qb (C1qb), resulting in impairment of apoptotic cell clearance and reduction in anti-inflammatory cytokine production. This increases autoantibody production and predisposes global and macrophage-specific Ppard(-/-) mice to autoimmune kidney disease, a phenotype resembling the human disease systemic lupus erythematosus. Thus, PPAR-delta has a pivotal role in orchestrating the timely disposal of apoptotic cells by macrophages, ensuring that tolerance to self is maintained.


Subject(s)
Apoptosis/physiology , Autoimmunity/physiology , Immune Tolerance/immunology , PPAR delta/metabolism , Animals , Apoptosis/drug effects , Autoantibodies/immunology , Autoantibodies/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/physiopathology , Autoimmunity/drug effects , CD11b Antigen/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Fluoresceins , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Hyaluronan Receptors/metabolism , Immune Tolerance/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mitochondrial Proteins , Opsonin Proteins/genetics , Opsonin Proteins/metabolism , PPAR delta/agonists , PPAR delta/deficiency , PPAR delta/genetics , Phagocytosis/drug effects , Phagocytosis/immunology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Thiazoles/pharmacology , Thymus Gland/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...