Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638886

ABSTRACT

Peroxisome proliferator-activated receptor α is a potent regulator of systemic and cellular metabolism and energy homeostasis, but it also suppresses various inflammatory reactions. In this review, we focus on its role in the regulation of innate immunity; in particular, we discuss the PPARα interplay with inflammatory transcription factor signaling, pattern-recognition receptor signaling, and the endocannabinoid system. We also present examples of the PPARα-specific immunomodulatory functions during parasitic, bacterial, and viral infections, as well as approach several issues associated with innate immunity processes, such as the production of reactive nitrogen and oxygen species, phagocytosis, and the effector functions of macrophages, innate lymphoid cells, and mast cells. The described phenomena encourage the application of endogenous and pharmacological PPARα agonists to alleviate the disorders of immunological background and the development of new solutions that engage PPARα activation or suppression.


Subject(s)
Energy Metabolism/immunology , Homeostasis/immunology , Immunity, Innate/immunology , Inflammation/immunology , PPAR alpha/immunology , Signal Transduction/immunology , Adaptive Immunity/immunology , Animals , Humans , Macrophages/immunology , PPAR alpha/metabolism
2.
Aging (Albany NY) ; 13(18): 22040-22058, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34544906

ABSTRACT

N-retinylidene-N-retinylethanolamine (A2E) plays a central role in age-related macular degeneration (AMD) by inducing angiogenesis and inflammation. A2E effects are mediated at least partly via the retinoic acid receptor (RAR)-α. Here we show that A2E binds and transactivates also peroxisome proliferator-activated receptors (PPAR) and retinoid X receptors (RXR). 9'-cis-norbixin, a di-apocarotenoid is also a ligand of these nuclear receptors (NR). Norbixin inhibits PPAR and RXR transactivation induced by A2E. Moreover, norbixin reduces protein kinase B (AKT) phosphorylation, NF-κB and AP-1 transactivation and mRNA expression of the inflammatory interleukins (IL) -6 and -8 and of vascular endothelial growth factor (VEGF) enhanced by A2E. By contrast, norbixin increases matrix metalloproteinase 9 (MMP9) and C-C motif chemokine ligand 2 (CCL2) mRNA expression in response to A2E. Selective PPAR-α, -ß/δ and -γ antagonists inhibit the expression of IL-6 and IL-8 while only the antagonist of PPAR-γ inhibits the transactivation of NF-κB following A2E exposure. In addition, a cocktail of all three PPARs antagonists and also HX531, an antagonist of RXR reproduce norbixin effects on inflammation. Altogether, A2E's deleterious biological effects could be inhibited through PPAR and RXR regulation. Moreover, the modulation of these NR by norbixin may open new avenues for the treatment of AMD.


Subject(s)
Carotenoids/administration & dosage , Macular Degeneration/drug therapy , PPAR alpha/immunology , PPAR delta/immunology , PPAR gamma/immunology , PPAR-beta/immunology , Retinal Pigment Epithelium/drug effects , Retinoids/immunology , Angiogenesis Inhibitors/administration & dosage , Animals , Humans , Macular Degeneration/chemically induced , Macular Degeneration/genetics , Macular Degeneration/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , PPAR alpha/genetics , PPAR delta/genetics , PPAR gamma/genetics , PPAR-beta/genetics , Retinal Pigment Epithelium/immunology , Retinoid X Receptors/agonists , Retinoid X Receptors/genetics , Retinoid X Receptors/immunology , Retinoids/adverse effects , Swine , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology
3.
Front Immunol ; 12: 618365, 2021.
Article in English | MEDLINE | ID: mdl-34434187

ABSTRACT

The current therapeutic options for Inflammatory Bowel Diseases (IBD) are limited. Even using common anti-inflammatory, immunosuppressive or biological therapies, many patients become unresponsive to the treatments, immunosuppressed or unable to restrain secondary infections. Statins are cholesterol-lowering drugs with non-canonical anti-inflammatory properties, whose underlying mechanisms of action still remain poorly understood. Here, we described that in vitro atorvastatin (ATO) treatment was not toxic to splenocytes, constrained cell proliferation and modulated IL-6 and IL-10 production in a dose-dependent manner. Mice exposed to dextran sulfate sodium (DSS) for colitis induction and treated with ATO shifted their immune response from Th17 towards Th2, improved the clinical and histological aspects of intestinal inflammation and reduced the number of circulating leukocytes. Both experimental and in silico analyses revealed that PPAR-α expression is reduced in experimental colitis, which was reversed by ATO treatment. While IBD patients also downregulate PPAR-α expression, the responsiveness to biological therapy relied on the restoration of PPAR-α levels. Indeed, the in vitro and in vivo effects induced by ATO treatment were abrogated in Ppara-/- mice or leukocytes. In conclusion, the beneficial effects of ATO in colitis are dependent on PPAR-α, which could also be a potential predictive biomarker of therapy responsiveness in IBD.


Subject(s)
Atorvastatin/pharmacology , Colitis/drug therapy , PPAR alpha/immunology , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Dextran Sulfate/toxicity , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Mice, Knockout , PPAR alpha/genetics , Th17 Cells/immunology , Th2 Cells/immunology
4.
Int J Biol Macromol ; 182: 179-186, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33838185

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) play a key role in the regulation of metabolic homeostasis, inflammation, cellular growth, and differentiation. To further explore the potential role of PPARα in the energy homeostasis of fatty liver hemorrhagic syndrome (FLHS), we reported the prokaryotic expression and purification of chicken PPARα subunit protein, and successfully prepared a polyclonal antibody against PPARα recombinant protein. The 987 bp PPARα subunit genes were cloned into the pEASY-T3 clone vector. Then the plasmid PCR products encoding 329 amino acids were ligated to pEASY-Blunt E2 vector and transformed into BL21 to induce expression. The recombinant PPARα subunit protein, containing His-tag, was purified by affinity column chromatography using Ni-NTA affinity column. Rabbit antiserum was generated by using the concentration of recombinant PPARα subunit protein as the antigen. The results of western blotting showed that the antiserum can specifically recognize chicken endogenous PPARα protein. Immunohistochemistry and immunofluorescence showed that the PPARα mainly existed in the nucleus of hepatocytes, renal epithelial cells and hypothalamic endocrine nerve cells. More importantly, western blotting and real-time quantitative PCR indicated that FLHS significantly decreased the expression of PPARα.


Subject(s)
Antibodies/immunology , Fatty Liver/veterinary , Hemorrhage/veterinary , PPAR alpha/metabolism , Poultry Diseases/metabolism , Animals , Antigen-Antibody Reactions , Blotting, Western/methods , Cells, Cultured , Chickens , Fatty Liver/metabolism , Female , Hemorrhage/metabolism , Hepatocytes/metabolism , Hypothalamus/metabolism , Immunohistochemistry/methods , Kidney/metabolism , PPAR alpha/genetics , PPAR alpha/immunology , Syndrome
5.
Fish Shellfish Immunol ; 102: 257-266, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32315742

ABSTRACT

Peroxisome proliferative-activated receptor α (PPARα) belongs to the superfamily of nuclear receptors (NR). Studies have demonstrated that PPARα functions in energy metabolism, hepatic function, immune response, cell cycle, and apoptosis. In teleost fish, few studies have investigated the role of PPARα in the immune response. In this study, the grouper PPARα gene (EcPPARα) was investigated for its role in viral infection. The open reading frame of EcPPARα encoded a protein of 469 amino acids and contained an N-terminal domain (NTD), a DNA-binding domain (DBD), a hinge region, and a C-terminal ligand-binding domain (LBD). Phylogenetic analysis revealed that EcPPARα was most closely related to homologous genes in Sander lucioperca and Perca flavescens. Upon challenge with SGIV (Singapore grouper iridovirus) and RGNNV (Red-spotted grouper nervous necrosis virus), EcPPARα expression levels were significantly upregulated in different tissues. Subcellular localization analysis showed that the EcPPARα protein localized throughout the cytoplasm and nucleus with diffuse intracellular expression patterns, which is consistent with the localization pattern of mammalian PPARs. Based on morphological observation of cytopathic effect (CPEs), viral gene expression mRNAs, and virus titer assays, the results presented here showed that an overexpression of EcPPARα promoted SGIV production in grouper spleen cells. Overexpression of EcPPARα significantly inhibited the expression of several cytokines, including interferon-related genes (IFN-γ, ISG15, MXI, MXII, MAVS and MDA5), inflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α) and Toll like receptor adaptors (TRAF6 and MyD88). Luciferase activity of IFN-α, IFN-γ, ISRE and NF-κB promoters was also significantly decreased in EcPPARα overexpression cells. Due to these detected interferon-related genes and inflammatory cytokines play important antiviral effect against SGIV in grouper, we speculated that the promotion effect of EcPPARα on SGIV replication may be caused by down-regulation of interferon and inflammatory response. In addition, through apoptotic body observation, capspase-3 activity detection, and flow cytometry analysis, it was found that overexpression of EcPPARα promoted SGIV-induced apoptosis in fathead minnow (FHM) cells. These data may increase an understanding of the role of PPARα in fish antiviral immune responses and apoptosis.


Subject(s)
Bass/genetics , Bass/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , PPAR alpha/genetics , PPAR alpha/immunology , Amino Acid Sequence , Animals , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Nodaviridae/physiology , PPAR alpha/chemistry , Phylogeny , RNA Virus Infections/immunology , RNA Virus Infections/veterinary , Ranavirus/physiology , Sequence Alignment/veterinary
6.
Biochem Biophys Res Commun ; 524(2): 308-316, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31987499

ABSTRACT

BACKGROUND: Atherosclerotic plaque rupture is the major trigger of acute cardiovascular risk events, and manipulation of M1/M2 macrophage homeostasis is an effective strategy for regulating atherosclerotic plaque stability. This study was aimed to illuminate the effects of oleoylethanolamide (OEA) on macrophage polarization and plaque stability. METHODS: Macrophages derived from THP-1 were treated with OEA followed by LPS/IFN-γ, and the markers of M1, M2 macrophages were monitored by western blot, real-time PCR and immunofluorescence staining. The effect of OEA on macrophage polarization in the arch of aortic arteries was tested by immunofluorescence staining and western blot, and the plaque stability was completed by Masson's trichrome and hematoxylin and eosin (HE) in apolipoprotein E (ApoE)-/- mice. RESULTS: OEA treatment enhanced the expression of two classic M2 macrophage markers, macrophage mannose receptor (CD206) and transforming growth factor (TGF-ß), while the expression of iNOS (M1 macrophages) was decreased in THP-1-derived macrophages. Blocking of PPARα using siRNA and inhibition of AMP-activated protein kinase (AMPK) by its inhibitor compound C attenuated the OEA-induced expression of M2 macrophage markers. In addition, OEA significantly suppressed M1, promoted M2 macrophage polarization, increased collagen content and decreased necrotic core size in atherosclerotic plaques in ApoE-/- mice, which were linked with the expression of PPARα. CONCLUSIONS: OEA improved atherosclerotic plaque stability through regulating macrophage polarization via AMPK-PPARα pathway.


Subject(s)
AMP-Activated Protein Kinases/immunology , Anti-Inflammatory Agents/therapeutic use , Endocannabinoids/therapeutic use , Macrophages/drug effects , Oleic Acids/therapeutic use , PPAR alpha/immunology , Plaque, Atherosclerotic/drug therapy , Animals , Humans , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Signal Transduction/drug effects , THP-1 Cells
7.
Front Immunol ; 10: 1769, 2019.
Article in English | MEDLINE | ID: mdl-31447832

ABSTRACT

Glucocorticoids (GCs) act via the glucocorticoid receptor (NR3C1, GRα) to combat overshooting responses to infectious stimuli, including lipopolysaccharide (LPS). As such, GCs inhibit the activity of downstream effector cytokines, such as tumor necrosis factor (TNF). PPARα (NR1C1) is a nuclear receptor described to function on the crossroad between lipid metabolism and control of inflammation. In the current work, we have investigated the molecular mechanism by which GCs and PPARα agonists cooperate to jointly inhibit NF-κB-driven expression in A549 cells. We discovered a nuclear mechanism that predominantly targets Mitogen- and Stress-activated protein Kinase-1 activation upon co-triggering GRα and PPARα. In vitro GST-pull down data further support that the anti-inflammatory mechanism may additionally involve a non-competitive physical interaction between the p65 subunit of NF-κB, GRα, and PPARα. Finally, to study metabolic effector target cells common to both receptors, we overlaid the effect of GRα and PPARα crosstalk in mouse primary hepatocytes under LPS-induced inflammatory conditions on a genome-wide level. RNA-seq results revealed lipid metabolism genes that were upregulated and inflammatory genes that were additively downregulated. Validation at the cytokine protein level finally supported a consistent additive anti-inflammatory response in hepatocytes.


Subject(s)
Inflammation/immunology , PPAR alpha/immunology , Receptors, Glucocorticoid/immunology , A549 Cells , Animals , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism/genetics , Lipopolysaccharides , Male , Mice, Inbred C57BL , NF-kappa B/immunology , PPAR alpha/agonists
8.
PLoS One ; 14(8): e0220894, 2019.
Article in English | MEDLINE | ID: mdl-31437187

ABSTRACT

Transcriptional regulations exert a critical control of metabolic homeostasis. In particular, the nuclear receptors (NRs) are involved in regulating numerous pathways of the intermediate metabolism. The purpose of the present study was to explore in liver cells the interconnectedness between three of them, LXR, FXR, and PPARα, all three known to act on lipid and glucose metabolism, and also on inflammation. The human cell line HepaRG was selected for its best proximity to human primary hepatocytes. Global gene expression of differentiated HepaRG cells was assessed after 4 hours and 24 hours of exposure to GW3965 (LXR agonist), GW7647 (PPARα agonist), and GW4064 and CDCA (FXR synthetic and natural agonist, respectively). Our work revealed that, contrary to our expectations, NR specificity is largely present at the level of target genes, with a smaller than expected overlap of the set of genes targeted by the different NRs. It also highlighted the much broader activity of the synthetic FXR ligand compared to CDCA. More importantly, our results revealed that activation of FXR has a pro-proliferative effect and decreases the number of tetraploid (or binucleated) hepatocytes, while LXR inhibits the cell cycle progression, inducing hepatocyte differentiation and an increase in tetraploidism. Conclusion: these results highlight the importance of analyzing the different NR activities in a context allowing a direct confrontation of each receptor outcome, and reveals the opposite role of FXR and LXR in hepatocyte cells division and maturation.


Subject(s)
Liver X Receptors/metabolism , Receptor Cross-Talk/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Benzoates , Benzylamines , Butyrates , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/physiology , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Humans , Isoxazoles , Liver/pathology , Liver X Receptors/immunology , Orphan Nuclear Receptors/metabolism , PPAR alpha/immunology , PPAR alpha/metabolism , Phenylurea Compounds , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/immunology , Systems Analysis
9.
Food Funct ; 10(8): 4861-4867, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31334539

ABSTRACT

This study investigates the acute anti-inflammatory activity of Mangifera indica L. leaf extract and mangiferin in the liver of rats fed a cafeteria diet. This study was a randomized longitudinal experimental study. The animals were divided into three groups - Control: cafeteria diet (CD); Extract: CD + leaf extract (250 mg kg-1); and Mangiferin: CD + mangiferin (40 mg kg-1). Body weight and food intake were measured every week. On day eight, mRNA and protein expression of inflammatory markers were evaluated in the liver. Also, liver weight, SOD activity and malondialdehyde concentration were measured. Treatment for only eight days with mango leaf extract and mangiferin increased SOD activity. Mangiferin intake increased the mRNA expression of PPAR-α and HSP72. The leaf extract treatment enhanced PPAR-α mRNA expression. Mangiferin and leaf extract consumption caused a lower concentration of NFκB (p65) in nuclear extracts, and greater IL-10 mRNA and protein levels. This study highlights the potential of acute treatment with mango leaf extract and mangiferin to prevent liver inflammation caused by fat-rich diets. These results indicate a new use for a product that has low cost, is found in great amounts, and is not routinely used.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Liver Diseases/drug therapy , Mangifera/chemistry , Plant Extracts/administration & dosage , Animals , Diet, High-Fat/adverse effects , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Liver/drug effects , Liver/immunology , Liver Diseases/etiology , Liver Diseases/genetics , Liver Diseases/immunology , Male , Malondialdehyde/immunology , PPAR alpha/genetics , PPAR alpha/immunology , Phytotherapy , Plant Leaves/chemistry , Rats
10.
Fish Shellfish Immunol ; 90: 244-249, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31029776

ABSTRACT

Adipose tissue plays an important role in energy reservation, also be considered as vital immunological organ in animals. Adipocytes are the basic unit of adipose tissue, while little is known about the relationship between lipid metabolism and inflammatory response in fish adipocytes so far. In this study, forskolin was used to induce adipocyte lipolysis, and 5 µM forskolin and 30 µM forskolin both triggered lipolysis by increasing ATGL expression. Consequently, 30 µM Forskolin instead of 5 µM Forskolin induced the expression of NF-κB and its target pro-inflammatory cytokine genes including MCP-1, IL-6 and TNF-α. Further study found that low grade rate of lipolysis activated PPARα gene, and its inhibitory effect on the mRNA expression of NF-κB and its target genes inhibited the adipocyte inflammation. On the contrary, high grade rate of lipolysis increased the expression levels of NF-κB and its target genes, while their expression were attenuated by inhibition of reactive oxygen species (ROS) using α-tocopherol, suggesting that ROS generated due to the PPARα-mediated oxidation of released fatty acids from lipolysis may contribute to adipocyte inflammation. These results indicated that PPARα has dose effect in inflammatory responses to adipocyte lipolysis in grass carp. Taken together, grass carp adipocytes have immune activity. The inflammatory response is linked to the grade rate of adipocyte lipolysis in grass carp adipocytes, and excessive adipocyte lipolysis may promote a dynamic immune response in adipose tissue. This is the first study showing the regulatory effects of lipolysis on immune functions in fish adipocytes.


Subject(s)
Carps/genetics , Carps/immunology , Fish Diseases/immunology , Fish Proteins/genetics , Immunity, Innate/genetics , Lipolysis/immunology , Signal Transduction/immunology , Adipocytes/immunology , Animals , Colforsin/pharmacology , Cytokines/genetics , Cytokines/immunology , Dose-Response Relationship, Drug , Fish Diseases/chemically induced , Fish Diseases/genetics , Fish Proteins/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , NF-kappa B/genetics , NF-kappa B/immunology , PPAR alpha/genetics , PPAR alpha/immunology
11.
Shock ; 51(2): 221-227, 2019 02.
Article in English | MEDLINE | ID: mdl-29547450

ABSTRACT

BACKGROUND: Colonization with Pseudomonas aeruginosa (PA), the most common pathogen isolated mainly in patients with cystic fibrosis, is particularly difficult to eradicate and is associated with acceleration of decline in lung function and with poorer prognosis. PA LPS is recognized by Toll-like receptors-4 (TLR4) and has been shown to induce lung inflammation in vivo. In addition, regulation of this process is essential for proper pathogen clearance and to prevent excessive inflammatory response resulting in tissue damage. One potential regulator of these process is the peroxisome proliferator-activated receptors (PPARs), and in particular PPARα. Thus, the purpose of the present study was to evaluate the effects of the absence of TLR4 and PPARα receptors in the pulmonary innate immunity response to PA and in the consequent inflammatory response and in the activation of the macromolecular complex of the NLRP3 inflammosome. METHODS: To evaluate the involvement of TLR4 and PPARα in a PA infection, we used TLR4 KO and PPARα KO mice that received an intratracheal (i.t.) administration of 50 µL of PA strain (106 CFU), thus evaluating if these mice were profoundly susceptible to PA compared with WT mice. RESULTS: The results of the present study showed that administration of PA worsened the pathophysiology of PA lung disease in TLR4 and PPARα KO mice compared with WT mice. CONCLUSIONS: The present study demonstrated that TLR4 and PPARα receptors would mediate the earliest control of bacterial replication as well as proinflammatory responses to PA infections, and in particular that PPARα receptors are needed to prevent an excessive inflammatory response, as in the control of the inflammasome complex NLP3 activation.


Subject(s)
Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , PPAR alpha/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Toll-Like Receptor 4/immunology , Animals , Inflammasomes/genetics , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , PPAR alpha/genetics , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Pseudomonas Infections/genetics , Pseudomonas Infections/pathology , Toll-Like Receptor 4/genetics
12.
Mol Immunol ; 105: 46-54, 2019 01.
Article in English | MEDLINE | ID: mdl-30496976

ABSTRACT

Increasing evidence has indicated that lncRNAs and miRNAs play important roles in the pathogenesis of myocardial ischemic and reperfusion (I/R) injury. This study investigated the potential roles and underlying molecular mechanisms of lncRNA H19 and H19-derived miR-675 in regulating myocardial I/R injury in vitro and in vivo. The results showed that expression of H19 and H19-derived miR-675 was upregulated in cardiomyocytes exposed to oxygen-glucose deprivation and reperfusion. Knockdown of H19 increased cell viability, reduced cell apoptosis, decreased inflammatory cytokines (IL-1ß, TNF-α and IL-6), inhibited oxidative stress, downregulated p-IκB-α and p-p65, and upregulated expression of Nrf2 and HO-1. All of these effects were partly reversed by overexpression of miR-675. Furthermore, we found that PPARα was a target gene of miR-675 and that H19 negatively regulated PPARα expression via miR-675. By inhibiting PPARα, the biological effects of miR-675 or H19 inhibition on cellular functions (apoptosis, inflammation and oxidative stress) were at least partially reversed. Moreover, knockdown of H19 significantly reduced infarct size, increased left ventricular systolic pressure, and decreased left ventricular end-diastolic pressure in a mouse model of myocardial I/R. Taken together, these data indicate that H19 inhibition protects the heart against myocardial I/R injury, which may be partly attributed to regulation of the miR-675/PPARα axis.


Subject(s)
Gene Expression Regulation/immunology , MicroRNAs/immunology , Myocardial Reperfusion Injury/immunology , Myocytes, Cardiac/immunology , PPAR alpha/immunology , RNA, Long Noncoding/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Gene Knockdown Techniques , Male , Mice , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , PPAR alpha/genetics , RNA, Long Noncoding/genetics
13.
Lipids Health Dis ; 17(1): 147, 2018 Jun 23.
Article in English | MEDLINE | ID: mdl-29935534

ABSTRACT

BACKGROUND: Among the eight stereoisomers of phytanic acid (PA), the 3RS, 7R, 11R-isomer is naturally occurring and is present in foods and the human body. PA is considered to have possible health benefits in the immune system. However, it remains undetermined whether these effects are elicited by the 3RS, 7R, 11R-PA isomer, because previous studies used a commercially available PA whose isomer configuration is unknown. In this study, we synthesized a preparation of 3RS, 7R, 11R-PA, and investigated its in vitro immunomodulatory effects, especially the T-cell production of interferon (IFN)-γ, which is associated with various autoimmune diseases. This study also investigated the effects of 3RS, 7R, 11R-PA on NF-κB activity in order to address the mechanism of its immunomodulatory effects. METHODS: Mouse splenocytes and purified T-cells were stimulated with T-cell mitogens and incubated with 3RS, 7R, 11R-PA, followed by evaluation of IFN-γ production. The effect of 3RS, 7R, 11R-PA on NF-κB activity was also investigated using an A549 cell line with stable expression of an NF-κB-dependent luciferase reporter gene. RESULTS: 3RS, 7R, 11R-PA significantly reduced in vitro IFN-γ production at both the protein and mRNA levels, and was accompanied by decreased expression of T-bet, a key regulator of Th1 cell differentiation. The results indicated that NF-κB-mediated transcriptional activity was significantly decreased by 3RS, 7R, 11R-PA and that GW6471, an antagonist of peroxisome proliferator activated receptor α (PPARα), abrogated the inhibitory effect of 3RS, 7R, 11R-PA on NF-κB activity. CONCLUSIONS: The present study suggests that 3RS, 7R, 11R-PA is a functional and bioactive fatty acid, and has a potentially beneficial effect for amelioration of T-cell mediated autoimmune diseases. This study also indicates that interference in the NF-κB pathway via PPARα activation is a potential mechanism of the immunomodulatory effects of 3RS, 7R, 11R-PA.


Subject(s)
Immunologic Factors/pharmacology , Interferon-gamma/genetics , PPAR alpha/genetics , Phytanic Acid/pharmacology , RNA, Messenger/genetics , T-Lymphocytes/drug effects , A549 Cells , Animals , Cell Differentiation/drug effects , Female , Gene Expression Regulation , Genes, Reporter , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Luciferases/genetics , Luciferases/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/immunology , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR alpha/antagonists & inhibitors , PPAR alpha/immunology , Phytohemagglutinins/antagonists & inhibitors , Phytohemagglutinins/pharmacology , Primary Cell Culture , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/immunology , Signal Transduction , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
14.
Front Immunol ; 9: 202, 2018.
Article in English | MEDLINE | ID: mdl-29503646

ABSTRACT

Metabolism provides substrates for reactive oxygen species (ROS) and nitric oxide (NO) generation, which are a part of the macrophage (Mφ) anti-microbial response. Mφs infected with Trypanosoma cruzi (Tc) produce insufficient levels of oxidative species and lower levels of glycolysis compared to classical Mφs. How Mφs fail to elicit a potent ROS/NO response during infection and its link to glycolysis is unknown. Herein, we evaluated for ROS, NO, and cytokine production in the presence of metabolic modulators of glycolysis and the Krebs cycle. Metabolic status was analyzed by Seahorse Flux Analyzer and mass spectrometry and validated by RNAi. Tc infection of RAW264.7 or bone marrow-derived Mφs elicited a substantial increase in peroxisome proliferator-activated receptor (PPAR)-α expression and pro-inflammatory cytokine release, and moderate levels of ROS/NO by 18 h. Interferon (IFN)-γ addition enhanced the Tc-induced ROS/NO release and shut down mitochondrial respiration to the levels noted in classical Mφs. Inhibition of PPAR-α attenuated the ROS/NO response and was insufficient for complete metabolic shift. Deprivation of glucose and inhibition of pyruvate transport showed that Krebs cycle and glycolysis support ROS/NO generation in Tc + IFN-γ stimulated Mφs. Metabolic profiling and RNAi studies showed that glycolysis-pentose phosphate pathway (PPP) at 6-phosphogluconate dehydrogenase was essential for ROS/NO response and control of parasite replication in Mφ. We conclude that IFN-γ, but not inhibition of PPAR-α, supports metabolic upregulation of glycolytic-PPP for eliciting potent ROS/NO response in Tc-infected Mφs. Chemical analogs enhancing the glucose-PPP will be beneficial in controlling Tc replication and dissemination by Mφs.


Subject(s)
Chagas Cardiomyopathy/immunology , Host-Parasite Interactions/immunology , Macrophages/immunology , Pentose Phosphate Pathway/immunology , Trypanosoma cruzi/immunology , Animals , Chagas Cardiomyopathy/parasitology , Disease Models, Animal , Humans , Interferon-gamma/immunology , Macrophages/parasitology , Mice , Mice, Knockout , Nitric Oxide/immunology , Nitric Oxide/metabolism , PPAR alpha/genetics , PPAR alpha/immunology , Primary Cell Culture , RAW 264.7 Cells , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Up-Regulation
15.
Biol Res ; 51(1): 5, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29448959

ABSTRACT

BACKGROUND: Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in ß oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). METHODS: Wild type (wt) or PPARα-deficient (PPARα-/-) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. RESULTS: Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα-/- mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα-/- splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. CONCLUSION: Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.


Subject(s)
Cytokines/immunology , Galactosylceramides/immunology , Hepatitis, Autoimmune/etiology , Macrolides/toxicity , PPAR alpha/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepatitis, Autoimmune/immunology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
16.
Biochem Biophys Res Commun ; 496(2): 508-514, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29353040

ABSTRACT

Eupatilin (5,7-dihydroxy-3',4',6-trimethoxyflavone) is the main lipophilic flavonoid obtained from the Artemisia species. Eupatilin has been reported to have anti-apoptotic, anti-oxidative and anti-inflammatory activities. Previously, we found that eupatilin increases transcriptional activity and expression of peroxisome proliferator-activated receptor α (PPARα) in a keratinocyte cell line and acts as an agonist of PPARα. PPARα agonists ameliorate atopic dermatitis (AD) and restore the skin barrier function. In this study, we confirmed that the effects of eupatilin improved AD-like symptoms in an oxazolone-induced AD-like mouse model. Furthermore, we found that eupatilin suppressed the levels of serum immunoglobulin E (IgE), interleukin-4 (IL-4), and AD involved cytokines, such as tumor necrosis factor α (TNFα), interferon-γ (IFN-γ), IL-1ß, and thymic stromal lymphopoietin (TSLP), IL-33, IL-25 and increased the levels of filaggrin and loricrin in the oxazolone-induced AD-like mouse model. Taken together, our data suggest that eupatilin is a potential candidate for the treatment of AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatologic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , PPAR alpha/genetics , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Dose-Response Relationship, Drug , Female , Filaggrin Proteins , Gene Expression Regulation , Immunoglobulin E/blood , Immunoglobulin E/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-33/genetics , Interleukin-33/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukins/genetics , Interleukins/immunology , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Oxazolone , PPAR alpha/immunology , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Thymic Stromal Lymphopoietin
17.
Biol. Res ; 51: 5, 2018. tab, graf
Article in English | LILACS | ID: biblio-888430

ABSTRACT

Abstract Background Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in β oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). Methods Wild type (wt) or PPARα-deficient (PPARα−/−) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. Results Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα−/− mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα−/− splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. Conclusion Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.


Subject(s)
Animals , Male , Mice , T-Lymphocytes/immunology , Cytokines/immunology , Macrolides/toxicity , Hepatitis, Autoimmune/etiology , PPAR alpha/immunology , Galactosylceramides/immunology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Hepatitis, Autoimmune/immunology , Disease Models, Animal , Real-Time Polymerase Chain Reaction , Mice, Inbred C57BL
18.
J Immunol ; 198(8): 3283-3295, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28275133

ABSTRACT

The role of peroxisome proliferator-activated receptor α (PPAR-α) in innate host defense is largely unknown. In this study, we show that PPAR-α is essential for antimycobacterial responses via activation of transcription factor EB (TFEB) transcription and inhibition of lipid body formation. PPAR-α deficiency resulted in an increased bacterial load and exaggerated inflammatory responses during mycobacterial infection. PPAR-α agonists promoted autophagy, lysosomal biogenesis, phagosomal maturation, and antimicrobial defense against Mycobacterium tuberculosis or M. bovis bacillus Calmette-Guérin. PPAR-α agonists regulated multiple genes involved in autophagy and lysosomal biogenesis, including Lamp2, Rab7, and Tfeb in bone marrow-derived macrophages. Silencing of TFEB reduced phagosomal maturation and antimicrobial responses, but increased macrophage inflammatory responses during mycobacterial infection. Moreover, PPAR-α activation promoted lipid catabolism and fatty acid ß-oxidation in macrophages during mycobacterial infection. Taken together, our data indicate that PPAR-α mediates antimicrobial responses to mycobacterial infection by inducing TFEB and lipid catabolism.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Immunity, Innate/immunology , Lipid Metabolism/immunology , Mycobacterium Infections/immunology , PPAR alpha/immunology , Animals , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Lipid Droplets/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium , PPAR alpha/metabolism , Polymerase Chain Reaction
19.
Biochim Biophys Acta ; 1862(4): 662-669, 2016 04.
Article in English | MEDLINE | ID: mdl-26850476

ABSTRACT

BACKGROUND AND AIMS: Pretreatment with clofibrate, a peroxisome proliferator-activated receptor alpha (PPARa) agonist, protects mice from acetaminophen (APAP) injury. Protection is not due to alterations in APAP metabolism and is dependent on PPARa expression. Gene array analysis revealed that mice receiving clofibrate have enhanced hepatic Vanin-1 (Vnn1) gene expression, a response that is also PPARa dependent. METHODS: We examined the role of Vnn1 by comparing the responses of Vnn1 knockout and wild-type mice following APAP hepatotoxicity. APAP metabolism, hepatotoxicity, and compensatory hepatocyte proliferation and immune responses were assessed. RESULTS: Vnn1 knockout mice are more susceptible to APAP hepatotoxicity despite no differences in hepatic glutathione content, gene expression of APAP metabolizing enzymes, or hepatic capacity to bioactivate or detoxify APAP ex vivo. Together, these data strongly suggest that the susceptibility of Vnn1 knockout mice is not due to differences in APAP metabolism. Immunochemistry revealed a lack of proliferating cell nuclear antigen-positive hepatocytes and F4/80-positive macrophages in and around areas of centrilobular necrosis in APAP-treated Vnn1 knockouts. Hepatic gene induction of pro-inflammatory cytokines was either significantly reduced or completely blunted in these mice. This was correlated with a reduction in early recruitment of cells positive for granulocyte differentiation antigen 1 or integrin alpha M. Heightened toxicity was also observed in CCl4 and ConA hepatitis models in the absence of Vnn1. CONCLUSIONS: These results indicate that mice lacking Vnn1 have deficiencies in compensatory repair and immune responses following toxic APAP exposure and that these mechanisms may contribute to the enhanced hepatotoxicity seen.


Subject(s)
Acetaminophen/adverse effects , Amidohydrolases/deficiency , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/immunology , Liver/immunology , Acetaminophen/pharmacology , Amidohydrolases/immunology , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Clofibrate/pharmacology , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Liver/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/immunology
20.
Curr Opin Immunol ; 38: 30-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26605965

ABSTRACT

Recent developments have demonstrated that metabolic rewiring imposed by adaptation of tissues to stress leads to the release of various metabolites which directly or indirectly impact innate immune responses and inflammation. Some metabolites can behave as second messengers and leave local cues in tissues. Immune cells which infiltrate stressed tissues reorient their metabolism to cope with these microenvironmental cues while preserving their effector functions in tissues.


Subject(s)
Adaptation, Physiological/immunology , Adenosine Triphosphate/immunology , Immunity, Innate , Metabolome/immunology , Second Messenger Systems/immunology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Adenosine Triphosphate/metabolism , Animals , Citric Acid Cycle/immunology , Cytoprotection , Gene Expression Regulation , Humans , Inflammation , Oxidative Phosphorylation , PPAR alpha/genetics , PPAR alpha/immunology , Plants/immunology , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...