Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Leukoc Biol ; 108(5): 1501-1514, 2020 11.
Article in English | MEDLINE | ID: mdl-32421906

ABSTRACT

ß2 Integrins mediate neutrophil-endothelial adhesion and recruitment of neutrophils to sites of inflammation. The diminished expression of ß2 integrins in patients with mutations in the ITGB2 (CD18) gene (leukocyte adhesion deficiency-Type 1; LAD1) results in few or no neutrophils in peripheral tissues. In the periodontium, neutrophil paucity is associated with up-regulation of IL-23 and IL-17, which drive inflammatory bone loss. Using a relevant mouse model, we investigated whether diminished efferocytosis (owing to neutrophil scarcity) is associated with LAD1 periodontitis pathogenesis and aimed to develop approaches to restore the missing efferocytosis signals. We first showed that CD18-/- mice phenocopied human LAD1 in terms of IL-23/IL-17-driven inflammatory bone loss. Ab-mediated blockade of c-Mer tyrosine kinase (Mer), a major efferocytic receptor, mimicked LAD1-associated up-regulation of gingival IL-23 and IL-17 mRNA expression in wild-type (WT) mice. Consistently, soluble Mer-Fc reversed the inhibitory effect of efferocytosis on IL-23 expression in LPS-activated Mϕs. Adoptive transfer of WT neutrophils to CD18-/- mice down-regulated IL-23 and IL-17 expression to normal levels, but not when CD18-/- mice were treated with blocking anti-Mer Ab. Synthetic agonist-induced activation of liver X receptors (LXR) and peroxisome proliferator-activated receptors (PPAR), which link efferocytosis to generation of homeostatic signals, inhibited the expression of IL-23 and IL-17 and favorably affected the bone levels of CD18-/- mice. Therefore, our data link diminished efferocytosis-associated signaling due to impaired neutrophil recruitment to dysregulation of the IL-23-IL-17 axis and, moreover, suggest LXR and PPAR as potential therapeutic targets for treating LAD1 periodontitis.


Subject(s)
Homeostasis/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Liver X Receptors/immunology , Periodontitis/immunology , Periodontium/immunology , Peroxisome Proliferator-Activated Receptors/immunology , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , Homeostasis/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Liver X Receptors/genetics , Mice , Mice, Knockout , Periodontitis/genetics , Periodontitis/pathology , Periodontium/pathology , Peroxisome Proliferator-Activated Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Up-Regulation/immunology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/immunology
2.
J Agric Food Chem ; 68(15): 4374-4386, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32250610

ABSTRACT

A long-term exposure to lipopolysaccharides results in the gut inflammation and its impaired barrier function, leading to the development of metabolic disorders. In this study, the role of dietary heat killed Lactobacillus pentosus S-PT84 on preventing endotoxemia to maintain metabolic homeostasis was studied. We demonstrated that the treatment of L. pentosus S-PT84 improved the gut integrity by maintaining tight-junction protein expression, in order to suppress the infiltration of endotoxin into plasma. The systemic inflammatory responses were inhibited via reducing the secretion of TNF-α and MCP-1. Furthermore, the blood lipid profile and glucose level as well as adiponectin in both plasma and white adipose tissues (WAT) were preserved by L. pentosus S-PT84 through upregulation of PPAR-γ and IRS-1 expression in WAT. The above findings suggest that the metabolic homeostasis in mice treated with HFD and LPS was sustained by L. pentosus S-PT84, leading to reducing the early risk for progression into metabolic disorders.


Subject(s)
Lactobacillus pentosus/physiology , Metabolic Diseases/drug therapy , Probiotics/administration & dosage , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/immunology , Lipopolysaccharides/adverse effects , Metabolic Diseases/etiology , Metabolic Diseases/immunology , Metabolic Diseases/microbiology , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
Int J Mol Sci ; 19(6)2018 May 25.
Article in English | MEDLINE | ID: mdl-29799467

ABSTRACT

Increasing evidence points towards the existence of a bidirectional interconnection between metabolic disease and neurodegenerative disorders, in which inflammation is linking both together. Activation of members of the peroxisome proliferator-activated receptor (PPAR) family has been shown to have beneficial effects in these interlinked pathologies, and these improvements are often attributed to anti-inflammatory effects of PPAR activation. In this review, we summarize the role of PPARs in immune cell function, with a focus on macrophages and T cells, and how this was shown to contribute to obesity-associated inflammation and insulin resistance, atherosclerosis, and neurodegenerative disorders. We address gender differences as a potential explanation in observed contradictory results, and we highlight PPAR-induced metabolic changes as a potential mechanism of regulation of immune cell function through these nuclear receptors. Together, immune cell-specific activation of PPARs present a promising therapeutic approach to treat both metabolic and neurodegenerative diseases.


Subject(s)
Atherosclerosis/immunology , Macrophages/immunology , Neurodegenerative Diseases/immunology , Obesity/immunology , Peroxisome Proliferator-Activated Receptors/immunology , T-Lymphocytes/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Female , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation , Macrophages/pathology , Male , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Obesity/genetics , Obesity/pathology , Peroxisome Proliferator-Activated Receptors/genetics , Protein Isoforms/genetics , Protein Isoforms/immunology , Sex Factors , Signal Transduction , T-Lymphocytes/pathology
4.
Virus Res ; 244: 147-152, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29162488

ABSTRACT

Avian leukosis virus (ALV) induces multiple avian tumors, growth decrease and immune suppression. Previously, a novel natural recombinant ALV isolate FJ15HT0 was proven to be associated with significant body weight decrease, immune suppression and lymphocytoma in infected SPF chickens. In order to uncover the interaction between virus and host, we compared differences in the transcriptomes of the thymuses from the mock chickens and simulated congenitally infected chickens at 5days (d), 13d and 21d of age by RNA-seq analysis of the thymuses. Signaling pathways including cytokine-cytokine receptor interactions, peroxisome proliferator-activated receptor (PPAR) signaling pathway, Janus tyrosine kinase/signal transducers and activators of transcription (Jak-STAT) signaling pathway and fatty acid degradation were involved in the interaction between FJ15HT0 and SPF chickens. Interestingly, fold change of ciliary neurotrophic factor receptor α (CNTFRα) in infected donor collected from 2d to 21d showed a significant positive correlation with the corresponding expression of the viral gp85 gene in thymuses (r=0.656, P<0.01) and in livers (r=0.525, P<0.05). It will provide new insights for the molecular pathogenesis of ALV infection.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/genetics , Avian Proteins/genetics , Poultry Diseases/genetics , Thymus Gland/virology , Transcription, Genetic , Animals , Avian Leukosis/immunology , Avian Leukosis/pathology , Avian Leukosis/virology , Avian Leukosis Virus/growth & development , Avian Leukosis Virus/metabolism , Avian Proteins/immunology , Body Weight , Chickens , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/immunology , Cytokines/genetics , Cytokines/immunology , Fatty Acids/metabolism , Host-Pathogen Interactions , Janus Kinases/genetics , Janus Kinases/immunology , Lipid Metabolism , Liver/immunology , Liver/virology , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Poultry Diseases/immunology , Poultry Diseases/pathology , Poultry Diseases/virology , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Signal Transduction , Specific Pathogen-Free Organisms , Thymus Gland/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
5.
Exp Mol Med ; 49(8): e364, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28775365

ABSTRACT

The prevalence of autoimmune, infectious and metabolic diseases is different for men and women owing to the respective ability of their immune systems to respond to self and foreign antigens. Although several factors, including hormones and the X-chromosome, have been suggested to contribute to such sex-specific immune responses, the underlying factors remain poorly defined. Recent studies using peroxisome proliferator-activated receptor (PPAR) ligands and knockout mice have identified sex-dimorphic expression of PPARs, and have shown that the inhibitory functions of PPAR in T cells are substantially affected by the sex hormones. In this review, we consider the sex-specific differences in PPARs and summarize the diverse PPAR-mediated, sex-specific properties of effector T-cell responses, such as T-cell activation, survival and differentiation, as well as their involvement in T-cell-related autoimmune diseases, including colitis, graft-versus-host disease and experimental autoimmune encephalomyelitis. Understanding PPAR-mediated sex differences in immune responses will provide more precise insights into the roles of PPARs in effector T cells.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Ligands , Male , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptors/immunology , Sex Factors , T-Lymphocytes/metabolism
6.
Curr Protein Pept Sci ; 18(6): 619-629, 2017.
Article in English | MEDLINE | ID: mdl-27356936

ABSTRACT

Clinical observations support the postulate that chronic low-grade inflammation underlies metabolic diseases and inflammatory mediators can trigger some metabolic diseases. In disorder condition, what is the first one: metabolic diseases cause inflammation or conversely? This "chicken or egg" type question was hard to answer. However, instead of focusing on this difficult issue, we should ask another challenging question: what are the links between inflammation and metabolic diseases? Seizing the key from this chaos may be the best way to solve the problem and break the cycle. To answer this question, we review the regulators (such as NF-κB, PPARs, mTOR, and STAT3) that have important roles in both metabolism and inflammation. These "bridge proteins" that link metabolic diseases and inflammation not only increase our understanding of these two diseases, but also provide potential targets for therapeutics and practical clinical applications.


Subject(s)
Inflammation/immunology , Metabolic Diseases/immunology , Animals , Humans , Inflammation/complications , Inflammation/metabolism , Metabolic Diseases/complications , Metabolic Diseases/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptors/immunology , Peroxisome Proliferator-Activated Receptors/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 310(1): L40-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26519208

ABSTRACT

Impaired vasodilation in persistent pulmonary hypertension of the newborn (PPHN) is characterized by mitochondrial dysfunction. We investigated the hypothesis that a decreased endothelial nitric oxide synthase level leads to impaired mitochondrial biogenesis and function in a lamb model of PPHN induced by prenatal ductus arteriosus constriction. We ventilated PPHN lambs with 100% O2 alone or with inhaled nitric oxide (iNO). We treated pulmonary artery endothelial cells (PAECs) from normal and PPHN lambs with detaNONOate, an NO donor. We observed decreased mitochondrial (mt) DNA copy number, electron transport chain (ETC) complex subunit levels, and ATP levels in PAECs and lung tissue of PPHN fetal lambs at baseline compared with gestation matched controls. Phosphorylation of AMP-activated kinase (AMPK) and levels of peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC-1α) and sirtuin-1, which facilitate mitochondrial biogenesis, were decreased in PPHN. Ventilation with 100% O2 was associated with larger decreases in ETC subunits in the lungs of PPHN lambs compared with unventilated PPHN lambs. iNO administration, which facilitated weaning of FiO2 , partly restored mtDNA copy number, ETC subunit levels, and ATP levels. DetaNONOate increased eNOS phosphorylation and its interaction with heat shock protein 90 (HSP90); increased levels of superoxide dismutase 2 (SOD2) mRNA, protein, and activity; and decreased the mitochondrial superoxide levels in PPHN-PAECs. Knockdown of eNOS decreased ETC protein levels in control PAECs. We conclude that ventilation with 100% O2 amplifies oxidative stress and mitochondrial dysfunction in PPHN, which are partly improved by iNO and weaning of oxygen.


Subject(s)
Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Mitochondria/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Persistent Fetal Circulation Syndrome/metabolism , Animals , Animals, Newborn , Female , Fetus/immunology , Fetus/metabolism , Mitochondria/immunology , Nitric Oxide/metabolism , Peroxisome Proliferator-Activated Receptors/immunology , Pregnancy , Sheep
8.
Cell Mol Immunol ; 13(1): 3-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26189369

ABSTRACT

Severe influenza remains unusual in its virulence for humans. Complications or ultimately death arising from these infections are often associated with hyperinduction of proinflammatory cytokine production, which is also known as 'cytokine storm'. For this disease, it has been proposed that immunomodulatory therapy may improve the outcome, with or without the combination of antiviral agents. Here, we review the current literature on how various effectors of the immune system initiate the cytokine storm and exacerbate pathological damage in hosts. We also review some of the current immunomodulatory strategies for the treatment of cytokine storms in severe influenza, including corticosteroids, peroxisome proliferator-activated receptor agonists, sphingosine-1-phosphate receptor 1 agonists, cyclooxygenase-2 inhibitors, antioxidants, anti-tumour-necrosis factor therapy, intravenous immunoglobulin therapy, statins, arbidol, herbs, and other potential therapeutic strategies.


Subject(s)
Antiviral Agents/therapeutic use , Cytokines/antagonists & inhibitors , Immunologic Factors/therapeutic use , Immunomodulation , Influenza, Human/drug therapy , Orthomyxoviridae/drug effects , Adrenal Cortex Hormones/therapeutic use , Antioxidants/therapeutic use , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/genetics , Cytokines/immunology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Indoles/therapeutic use , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae/immunology , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Plant Preparations/therapeutic use , Receptors, Lysosphingolipid/therapeutic use
9.
Mol Immunol ; 53(4): 421-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23123408

ABSTRACT

Endocrine disrupting chemicals (EDCs) have become of concern for a variety f health issues. Due to their effects on the endocrine system they have been thoroughly examined with regards to sexual dysfunction, malformation, and cancers of reproductive origin. Bisphenol-A (BPA) is a widely studied EDC and has been characterized regarding its estrogenic effects on a variety of cell types. BPA also alters immune responses. In this review, we examine some of the documented effects of EDCs, with a focus on BPA that pertain to modulation of the immune system and various immune cell populations. We highlight the multiple actions of BPA on altering T cell subsets, B cell functions, and dendritic cell and macrophage biology. Finally, we consider that these immunological activities of BPA may be mediated through estrogen receptor signaling, arylhydrocarbon receptor, and the peroxisome proliferator-activated receptor family of nuclear receptors.


Subject(s)
B-Lymphocytes/drug effects , Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Endocrine System/drug effects , Macrophages/drug effects , Phenols/toxicity , T-Lymphocyte Subsets/drug effects , Animals , B-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Endocrine System/immunology , Endocrine System/metabolism , Gene Expression , Humans , Macrophages/immunology , Mice , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Receptors, Estrogen/genetics , Receptors, Estrogen/immunology , Signal Transduction , T-Lymphocyte Subsets/immunology
10.
Lipids ; 48(1): 29-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23129255

ABSTRACT

Conjugated linoleic acid (CLA) is thought to have anti-proliferative and anti-inflammatory properties, but its effect on cancer cachexia is unknown. Two effects were here investigated: that of CLA on inflammatory mediator production in human lung cancer cells, and that of reduced mediators on the myogenic differentiation of murine muscle C2C12 cells. The latter cells were grown in medium conditioned by human lung cancer A427 cells, with or without CLA, to mimic only the effect of molecules released from the tumor "in vivo", excluding the effect of host-produced cachectic factors. The results obtained show that CLA was found to reduce the production of tumor necrosis factor-α, interleukin (IL)-1ß and prostaglandin E2 (PGE2), but had no effect on IL-6 production. The mechanisms underlying the effect of CLA on cytokine or PGE2 release in A427 cells are probably mediated by activation of peroxisome proliferator-activated receptor (PPAR)α, which increased at 24 h CLA treatment. In turn, the reduced content of inflammatory mediators in medium conditioned by A427 cells, in the presence of CLA, allowed muscle cells to proliferate, again by inducing PPAR. The involvement of PPARα was demonstrated by treatment with the antagonist MK-886. The findings demonstrate the anti-inflammatory and myogenic action of CLA and point to its possible application as a novel dietary supplement and therapeutic agent in inflammatory disease states, such as cachexia.


Subject(s)
Anticarcinogenic Agents/pharmacology , Inflammation Mediators/immunology , Linoleic Acids, Conjugated/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Muscle Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Dinoprostone/immunology , Horses , Indoles/pharmacology , Interleukin-1/immunology , Interleukin-6/immunology , Lung/drug effects , Lung/immunology , Mice , Muscle Cells/cytology , Peroxisome Proliferator-Activated Receptors/immunology
12.
J Agric Food Chem ; 60(11): 2815-24, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22352956

ABSTRACT

Ginkgo biloba, which is considered a "living fossil", has been used for medicinal purposes for thousands of years. Currently, extracts of G. biloba are some of the most widely used herbal products and/or dietary supplements in the world. In this study, three new compounds, (2E,4E,1'R,3'S,5'R,8'S)-dihydrophaseic acid 3'-O-ß-D-glucopyranoside (1), 7,8-dihydro-(R)-7-methoxyconiferyl alcohol (2), and (8S)-3-methoxy-8,4'-oxyneolignan-4,9,9'-triol 3'-O-ß-D-glucopyranoside (3), and 13 known compounds (4-16) were isolated from the stem bark of G. biloba. Their structures were determined by extensive spectroscopic methods, including 1D and 2D NMR, MS, and circular dichroism spectra. Four of the compounds (1, 2, 7, and 10) inhibited TNFα-induced NF-κB transcriptional activity significantly in HepG2 cells in a dose-dependent manner, with IC50 values ranging from 6.9 to 9.1 µM. Furthermore, the transcriptional inhibitory function of these compounds was confirmed based on decreases in COX-2 and iNOS gene expression in HepG2 cells. Compounds 1-5, 7, 9, 10, and 12-14 significantly activated the transcriptional activity of PPARs in a dose-dependent manner, with EC50 values ranging from 0.7 to 12.8 µM. Compounds 2, 3, and 12 exhibited dose-dependent PPARα transactivational activity, with EC50 values of 7.0, 3.3, and 10.1 µM, respectively. Compounds 1-3 activated PPARγ transcriptional activity, with EC50 values of 11.9, 11.0, and 15.3 µM, whereas compounds 1 and 3 promoted the transactivational activity of PPARß(δ) with EC50 values of 10.7 and 11.2 µM, respectively. These results provide a scientific support for the use of G. biloba stem bark for the prevention and treatment of inflammatory and metabolic diseases. Moreover, these data provide the rationale for further studies of the potential of G. biloba stem bark in functional foods.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ginkgo biloba/chemistry , Peroxisome Proliferator-Activated Receptors/genetics , Plant Extracts/pharmacology , Plant Stems/chemistry , Transcriptional Activation/drug effects , Hep G2 Cells , Humans , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Peroxisome Proliferator-Activated Receptors/immunology
13.
Curr Top Med Chem ; 12(6): 548-84, 2012.
Article in English | MEDLINE | ID: mdl-22242855

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand dependent transcription factors. The three mammalian PPARs are key regulators of fatty acid and lipoprotein metabolism, glucose homeostasis, cellular proliferation/ differentiation and the immune response. PPARs are therefore important targets in the treatment of metabolic disorders such as insulin resistance and type 2 diabetes mellitus, and are also of interest in relation to chronic inflammatory diseases such as atherosclerosis, arthritis, chronic pulmonary inflammation, pancreatitis, inflammatory bowel disease, and psoriasis. Recent advances have attributed novel functions to PPARs in blood pressure regulation, neuroinflammation, nerve-cell protection, inflammatory pain reduction, and the hypothalamic control of metabolism. The abundant pleiotropic actions of PPARs suggest that PPAR agonists have enormous therapeutic potential. However, current PPAR-based therapies often have undesired side effects due to the concomitant activation of PPARs in non-target cells. There is therefore growing interest in the development of cell-specific PPAR agonists and improvement of the clinical use of PPAR ligands. This review gives an overview of PPAR functions and discusses the current and potential medical implications of PPAR ligands in various pathologies, ranging from metabolic disorders to cardiovascular disease, chronic inflammation, neurodegenerative disorders and cancer.


Subject(s)
Molecular Targeted Therapy , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Humans , Ligands , Peroxisome Proliferator-Activated Receptors/immunology
14.
Exp Toxicol Pathol ; 64(1-2): 127-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20674317

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) have been implicated in regulating the immune response. We determined the relative changes in the transcriptional expression of PPAR isoforms (α, γ1 and γ2) and cytokines involved in the pathogenesis of type 1 diabetes (T1D) in the immune cells of 5 weeks, 10 weeks and diabetic male non-obese diabetic (NOD) mice compared to those of female NOD mice from our previous studies, "normalized" against their respective non-obese diabetic resistant (NOR) mice controls. Overall PPARα was significantly more elevated in the macrophages of female NOD mice of all age groups whereas PPARγ, particularly the PPARγ2 isoform was more depressed in the macrophages and CD4(+) lymphocytes of female NOD mice compared to their male counterparts. The pro-inflammatory cytokines, IL-1 and TNFα, as well as the Th1 cytokines, IL-2 and IFNγ were more elevated in female NOD mice whereas the Th2 cytokine, IL-4, was more depressed in these mice compared to their male counterparts. These findings suggest that the preponderance of T1D in female NOD mice may be influenced by the more pronounced changes in the expression of PPAR isoforms and pathogenic cytokines compared to those in male NOD mice.


Subject(s)
Cytokines/genetics , Diabetes Mellitus, Type 1/immunology , Peroxisome Proliferator-Activated Receptors/genetics , Sex Characteristics , Transcription, Genetic , Animals , Cytokines/immunology , Diabetes Mellitus, Type 1/genetics , Female , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred NOD , Peroxisome Proliferator-Activated Receptors/immunology , Protein Isoforms , Real-Time Polymerase Chain Reaction , Spleen/cytology , Spleen/immunology
15.
Bioorg Med Chem Lett ; 21(20): 6143-7, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21889336

ABSTRACT

Two new oleanane-type triterpene saponins, tarasaponin IV (1) and elatoside L (2), and four known; stipuleanoside R(2) (3), kalopanax-saponin F (4), kalopanax-saponin F methylester (5), and elatoside D (6) were isolated from the bark of Aralia elata. Kalopanax-saponin F methyl ester was isolated from nature for the first time. Their chemical structures were elucidated using the chemical and physical methods as well as good agreement with those of reported in the literature. Oleanane-type triterpene saponins are the main component of A. elata. All compounds were investigated the anti-inflammatory activity. We measured their inhibition of NF-κB and activation of PPARs activities in HepG2 cells using luciferase reporter system. As results, compounds 2 and 4 were found to inhibit NF-κB activation stimulated by TNFα in a dose-dependent manner with IC(50) values of 4.1 and 9.5 µM, respectively, when compared with that of positive control, sulfasalazine (0.9 µM). Compounds 2 and 4 also inhibited TNFα-induced expression of iNOS and COX-2 mRNA. Furthermore, compounds 1-6 were evaluated PPAR activity using PPAR subtype transactivation assays. Among of them, compounds 4-6 significantly increased PPARγ transactivation. However, compounds 4-6 did not activate in any other PPAR subtypes.


Subject(s)
Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Aralia/chemistry , NF-kappa B/antagonists & inhibitors , Saponins/isolation & purification , Saponins/pharmacology , Anti-Infective Agents/chemistry , Hep G2 Cells , Humans , NF-kappa B/immunology , Oleanolic Acid/chemistry , Oleanolic Acid/isolation & purification , Oleanolic Acid/pharmacology , Peroxisome Proliferator-Activated Receptors/immunology , Plant Bark/chemistry , Saponins/chemistry
16.
Article in English | MEDLINE | ID: mdl-20509837

ABSTRACT

It is now clear that several members of the nuclear receptor superfamily are co-expressed by macrophages, lymphocytes and other cell types that are involved in the regulation of inflammatory and immune responses. Peroxisome proliferator-activated receptors (PPAR) and nuclear liver X receptors (LXR) are members of this family known to be activated by lipid derived endogenous ligands (such as fatty acids, eicosanoids and cholesterol) and pharmacological ones. Here we review the biology of these nuclear receptors and highlight recent work that show that their activation can ameliorate inflammatory conditions, especially due to their effect on macrophage functions. The data discussed herein show the potential beneficial effect of targeting these nuclear receptors in order to improve the outcome of septic patients.


Subject(s)
Lipid Metabolism/immunology , Orphan Nuclear Receptors/immunology , Peroxisome Proliferator-Activated Receptors/immunology , Sepsis/immunology , Humans , Liver X Receptors , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Sepsis/metabolism
17.
Immunobiology ; 215(8): 611-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19833407

ABSTRACT

Cannabinoids act via cell surface G protein-coupled receptors (CB(1) and CB(2)) and the ion channel receptor TRPV1. Evidence has now emerged suggesting that an additional target is the peroxisome proliferator-activated receptor (PPAR) family of nuclear receptors. There are three PPAR subtypes alpha, delta (also known as beta) and gamma, which regulate cell differentiation, metabolism and immune function. The major endocannabinoids, anandamide and 2-arachidonoylglycerol, and ajulemic acid, a structural analogue of the phytocannabinoid Delta(9)-tetrahydrocannabinol (THC), have anti-inflammatory properties mediated by PPARgamma. Other cannabinoids which activate PPARgamma include N-arachidonoyl-dopamine, THC, cannabidiol, HU210, WIN55212-2 and CP55940. The endogenous acylethanolamines, oleoylethanolamide and palmitoylethanolamide regulate feeding and body weight, stimulate fat utilization and have neuroprotective effects mediated through PPARalpha. Other endocannabinoids that activate PPARalpha include anandamide, virodhamine and noladin ether. There is, as yet, little direct evidence for interactions of cannabinoids with PPARdelta. There is a convergence of effects of cannabinoids, acting via cell surface and nuclear receptors, on immune cell function which provides promise for the targeted therapy of a variety of immune, particularly neuroinflammatory, diseases.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cannabinoids/metabolism , Inflammation/metabolism , Neuroimmunomodulation/immunology , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Cannabinoid Receptor Modulators/immunology , Cannabinoids/immunology , Humans , Inflammation/immunology , Peroxisome Proliferator-Activated Receptors/immunology
18.
Free Radic Biol Med ; 46(2): 127-37, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18996472

ABSTRACT

It is well recognized that oxidized LDL (OxLDL) plays a crucial role in the initiation and progression of atherosclerosis. Many biological effects of OxLDL are mediated through signaling pathways, especially via the activation of transcription factors, which in turn stimulate the expression of genes involved in the inflammatory and oxidative stress response or in cell cycle regulation. In this review, we will discuss the various transcription factors activated by OxLDL, the studied cell types, the active compounds of the OxLDL particle, and the downstream genes when identified. Identification of the transcription factors and some of the downstream genes regulated by OxLDL has helped us understand the molecular mechanism involved in generation of the atherosclerotic plaque.


Subject(s)
Atherosclerosis/immunology , Lipoproteins, LDL/genetics , Transcriptional Activation/immunology , Animals , Atherosclerosis/genetics , Cell Cycle , Feedback, Physiological , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/immunology , Hypoxia-Inducible Factor 1/metabolism , Inflammation , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Peroxisome Proliferator-Activated Receptors/metabolism , STAT Transcription Factors/immunology , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/immunology , Smad3 Protein/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
19.
Immunology ; 125(3): 289-301, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18445008

ABSTRACT

Dendritic cells (DCs) are highly potent antigen-presenting cells (APCs) and play a vital role in stimulating naïve T cells. Treatment of human blood monocytes with the cytokines granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 stimulates them to develop into immature dendritic cells (iDCs) in vitro. DCs generated by this pathway have a high capacity to prime and activate resting T cells and prominently express CD1 antigen-presenting molecules on the cell surface. The presence of human serum during the differentiation of iDCs from monocytes inhibits the expression of CD1a, CD1b and CD1c, but not CD1d. Correspondingly, T cells that are restricted by CD1c showed poor responses to DCs that were generated in the presence of human serum, while the responses of CD1d-restricted T cells were enhanced. We chemically fractionated human serum to isolate the bioactive factors that modulate surface expression of CD1 proteins during monocyte to DC differentiation. The human serum components that affected CD1 expression partitioned with polar organic soluble fractions. Lysophosphatidic acid and cardiolipin were identified as lipids present in normal human serum that potently modulate CD1 expression. Control of CD1 expression was mediated at the level of gene transcription and correlated with activation of the peroxisome proliferator-activated receptor (PPAR) nuclear hormone receptors. These findings indicate that the ability of human DCs to present lipid antigens to T cells through expression of CD1 molecules is sensitively regulated by lysophosphatidic acid and cardiolipin in serum, which are ligands that can activate PPAR transcription factors.


Subject(s)
Antigens, CD1/metabolism , Dendritic Cells/immunology , Lipids/immunology , Antigen Presentation/immunology , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Gene Expression , Humans , Lymphocyte Activation/immunology , Peroxisome Proliferator-Activated Receptors/biosynthesis , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Serum/immunology , T-Lymphocyte Subsets/immunology
20.
J Invest Dermatol ; 128(5): 1266-72, 2008 May.
Article in English | MEDLINE | ID: mdl-17960176

ABSTRACT

Differentiation of sebocytes is strongly associated with enhanced lipid synthesis and accumulation in the cells. Liver X receptors (LXRs) are members of the nuclear receptor superfamily, which play a critical role in cholesterol homeostasis and lipid metabolism. We examined whether LXRalpha regulated lipid synthesis in the immortalized human sebaceous gland cell line SZ95. When the SZ95 sebocytes were treated with the ligand of LXR such as TO901317 or 22(R)-hydroxycholesterol, lipid droplets were accumulated in the majority of cells when examined by Oil Red O staining. The expression of the known LXR targets, such as fatty acid synthase and sterol regulatory-binding protein-1, was induced by TO901317. TO901317 treatment increased expression of LXRalpha but not that of LXRbeta. Transfection of antisense LXRalpha significantly decreased TO901317-induced target gene expression and lipid droplet accumulation, suggesting a major role of LXRalpha in differentiation of sebocytes. Further, TO901317 decreased the expression of cyclooxygenase-2 and inducible nitric oxide synthase that was induced by lipopolysaccharide treatment. Together, these results indicate that important roles of LXRalpha in differentiation and inflammatory signaling in sebaceous glands. Thus, we suggest that LXR ligands could provide a new class of therapeutic agents for sebaceous gland-associated disorders such as seborrhea and acne.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lipids/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sebaceous Glands/metabolism , Azo Compounds , Cell Differentiation/physiology , Cell Line, Transformed , Coloring Agents , Gene Expression/drug effects , Gene Expression/immunology , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Orphan Nuclear Receptors , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , RNA, Messenger/metabolism , Sebaceous Glands/cytology , Sebum/metabolism , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/genetics , Sulfonamides/pharmacology , Transfection , fas Receptor/genetics , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...