Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Gene ; 806: 145921, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34454033

ABSTRACT

Maoto, a traditional Japanese medicine (Kampo), is widely used to treat upper respiratory tract infections, including influenza virus infection. Although maoto is known to inhibit pro-inflammatory responses in a rodent model of acute inflammation, its underlying mechanism remains to be determined. In this study, we investigated the involvement of immune responses and noradrenergic function in the inhibitory action of maoto. In a mouse model of polyI:C-induced acute inflammation, maoto was administered orally in conjunction with intraperitoneal injection of PolyI:C (6 mg/kg), and blood was collected after 2 h for measurement of plasma cytokines by ELISA. Maoto significantly decreased PolyI:C-induced TNF-α levels and increased IL-10 production. Neither pretreatment with IL-10 neutralizing antibodies nor T-cell deficiency using nude mice modified the inhibitory effect of maoto, indicating that the anti-inflammatory effects of maoto are independent of IL-10 and T cells. Furthermore, the inhibitory effects of maoto on PolyI:C-induced TNF-α production were not observed in ex vivo splenocytes, suggesting that maoto does not act directly on inflammatory cells. Lastly, pretreatment with a ß-adrenergic receptor antagonist partially cancelled the anti-inflammatory effects of maoto. Collectively, these results suggest that maoto mediates its anti-inflammatory effects via ß-adrenergic receptors in vivo.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Interleukin-10/genetics , Plant Extracts/pharmacology , Receptors, Adrenergic, beta/genetics , Administration, Oral , Animals , Disease Models, Animal , Ephedrine/pharmacology , Gene Expression Regulation , Injections, Intraperitoneal , Interleukin-10/agonists , Interleukin-10/immunology , Japan , Male , Medicine, Kampo/methods , Mice, Inbred BALB C , Mice, Nude , Poly I-C/administration & dosage , Poly I-C/antagonists & inhibitors , Receptors, Adrenergic, beta/immunology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
2.
PLoS One ; 16(12): e0260706, 2021.
Article in English | MEDLINE | ID: mdl-34871316

ABSTRACT

Airway epithelial barrier dysfunction is increasingly recognized as a key feature of asthma and other lung diseases. Respiratory viruses are responsible for a large fraction of asthma exacerbations, and are particularly potent at disrupting epithelial barrier function through pattern recognition receptor engagement leading to tight junction dysfunction. Although different mechanisms of barrier dysfunction have been described, relatively little is known about whether barrier integrity can be promoted to limit disease. Here, we tested three classes of drugs commonly prescribed to treat asthma for their ability to promote barrier function using a cell culture model of virus-induced airway epithelial barrier disruption. Specifically, we studied the corticosteroid budesonide, the long acting beta-agonist formoterol, and the leukotriene receptor antagonist montelukast for their ability to promote barrier integrity of a monolayer of human bronchial epithelial cells (16HBE) before exposure to the viral mimetic double-stranded RNA. Of the three, only budesonide treatment limited transepithelial electrical resistance and small molecule permeability (4 kDa FITC-dextran flux). Next, we used a mouse model of acute dsRNA challenge that induces transient epithelial barrier disruption in vivo, and studied the effects budesonide when administered prophylactically or therapeutically. We found that budesonide similarly protected against dsRNA-induced airway barrier disruption in the lung, independently of its effects on airway inflammation. Taken together, these data suggest that an under-appreciated effect of inhaled budesonide is to maintain or promote airway epithelial barrier integrity during respiratory viral infections.


Subject(s)
Asthma/drug therapy , Bronchi/drug effects , Bronchodilator Agents/pharmacology , Budesonide/pharmacology , Cell Membrane Permeability/drug effects , Poly I-C/antagonists & inhibitors , Acetates/pharmacology , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchi/metabolism , Bronchi/pathology , Cell Line , Cyclopropanes/pharmacology , Dextrans/metabolism , Electric Impedance , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Formoterol Fumarate/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Mimicry , Poly I-C/pharmacology , Quinolines/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Double-Stranded/pharmacology , RNA, Viral/antagonists & inhibitors , RNA, Viral/pharmacology , Sulfides/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism
3.
Biomolecules ; 11(8)2021 08 03.
Article in English | MEDLINE | ID: mdl-34439814

ABSTRACT

Vitamin C is well documented to have antiviral functions; however, there is limited information about its effect on airway epithelial cells-the first cells to encounter infections. Here, we examined the effect of vitamin C on human bronchial epithelium transformed with Ad12-SV40 2B (BEAS-2B) cells, and observed that sodium-dependent vitamin C transporter 2 (SVCT2) was the primary vitamin C transporter. Transcriptomic analysis revealed that treating BEAS-2B cells with vitamin C led to a significant upregulation of several metabolic pathways and interferon-stimulated genes (ISGs) along with a downregulation of pathways involved in lung injury and inflammation. Remarkably, vitamin C also enhanced the expression of the viral-sensing receptors retinoic acid-inducible gene 1 (RIG-1) and melanoma differentiation-associated protein 5 (MDA-5), which was confirmed at the protein and functional levels. In addition, the lungs of l-gulono-γ-lactone oxidase knockout (GULO-KO) mice also displayed a marked decrease in these genes compared to wild-type controls. Collectively, our findings indicate that vitamin C acts at multiple levels to exert its antiviral and protective functions in the lungs.


Subject(s)
Antiviral Agents/pharmacology , Ascorbic Acid/pharmacology , Epithelial Cells/drug effects , Interferon-Induced Helicase, IFIH1/genetics , Receptors, Retinoic Acid/genetics , Sodium-Coupled Vitamin C Transporters/genetics , Animals , Biological Transport , Bronchi/drug effects , Bronchi/metabolism , Cell Line, Transformed , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Mice , Mice, Knockout , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Retinoic Acid/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Transcriptome
4.
Biofactors ; 43(3): 388-399, 2017 May 06.
Article in English | MEDLINE | ID: mdl-28139053

ABSTRACT

Ze339, an herbal extract from Petasites hybridus leaves is effective in treatment of allergic rhinitis by inhibition of a local production of IL-8 and eicosanoid LTB4 in allergen-challenged patients. However, the mechanism of action and anti-inflammatory potential in virally induced exacerbation of the upper airways is unknown. This study investigates the anti-inflammatory mechanisms of Ze339 on primary human nasal epithelial cells (HNECs) upon viral, bacterial and pro-inflammatory triggers. To investigate the influence of viral and bacterial infections on the airways, HNECs were stimulated with viral mimics, bacterial toll-like-receptor (TLR)-ligands or cytokines, in presence or absence of Ze339. The study uncovers Ze339 modulated changes in pro-inflammatory mediators and decreased neutrophil chemotaxis as well as a reduction of the nuclear translocation and phosphorylation of STAT molecules. Taken together, this study suggests that phyto drug Ze339 specifically targets STAT-signalling pathways in HNECs and has high potential as a broad anti-inflammatory drug that exceeds current indication. © 2016 BioFactors, 43(3):388-399, 2017.


Subject(s)
Epithelial Cells/drug effects , Petasites/chemistry , Plant Extracts/pharmacology , STAT Transcription Factors/antagonists & inhibitors , Sesquiterpenes/pharmacology , Cell Movement/drug effects , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Epithelial Cells/cytology , Epithelial Cells/metabolism , Flagellin/antagonists & inhibitors , Flagellin/pharmacology , Gene Expression Regulation , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , Interleukin-4/antagonists & inhibitors , Interleukin-4/pharmacology , Lipopeptides/antagonists & inhibitors , Lipopeptides/pharmacology , Nasal Cavity/cytology , Nasal Cavity/drug effects , Nasal Cavity/metabolism , Neutrophils/drug effects , Plant Leaves/chemistry , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Primary Cell Culture , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
5.
BMB Rep ; 50(1): 25-30, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28027722

ABSTRACT

In the central nervous system, viral infection can induce inflammation by up-regulating pro-inflammatory mediators that contribute to enhanced infiltration of immune cells into the central nervous areas. Celastrol is known to exert various regulatory functions, including anti-microbial activities. In this study, we investigated the regulatory effects and the mechanisms of action of celastrol against astrocytes activated with polyinosinic-polycytidylic acid (poly(I:C)), a synthetic dsRNA, as a model of pro-inflammatory mediated responses. Celastrol significantly inhibited poly(I:C)-induced expression of adhesion molecules, such as ICAM-1/VCAM-1, and chemokines, such as CCL2, CXCL8, and CXCL10, in CRT-MG human astroglioma cells. In addition, celastrol significantly suppressed poly(I:C)-induced activation of JNK MAPK and STAT1 signaling pathways. Furthermore, celastrol significantly suppressed poly(I:C)-induced activation of the NF-κB signaling pathway. These results suggest that celastrol may exert its regulatory activity by inhibiting poly(I:C)-induced expression of pro-inflammatory mediators by suppressing activation of JNK MAPK-STAT1/NF-κB in astrocytes. [BMB Reports 2017; 50(1): 25-30].


Subject(s)
Astrocytes/drug effects , Intercellular Adhesion Molecule-1/biosynthesis , MAP Kinase Signaling System/drug effects , Poly I-C/pharmacology , Triterpenes/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Astrocytes/metabolism , Cell Line , Chemokines/biosynthesis , Chemokines/metabolism , Humans , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Pentacyclic Triterpenes , Poly I-C/antagonists & inhibitors , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/metabolism
6.
Life Sci ; 155: 189-94, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27210890

ABSTRACT

AIMS: Keratinocytes are the predominant cells in the epidermis, exerting their primary role of physical barrier through sophisticated differentiation process. In addition, keratinocytes contribute to the activation of innate immunity, providing the surveillant role against external pathogens. It has been known that chronic skin inflammatory disease such as psoriasis can be provoked by viral pathogens including double-stranded RNA. In this study, we demonstrated that rosmarinic acid (RA) has an inhibitory potential on inflammatory reaction induced by double-stranded RNA mimic poly(I:C) in epidermal keratinocytes. MAIN METHODS: We cultured human epidermal keratinocytes and induced inflammatory reaction by poly(I:C) treatment. The effect of RA on inflammatory reaction of keratinocytes was determined by RT-PCR and Western blot. KEY FINDINGS: RA significantly inhibited poly(I:C)-induced expression of inflammatory cytokines including IL-1ß, IL-6, IL-8, CCL20, and TNF-α, and downregulated NF-κB signaling pathway in human keratinocytes. In addition, RA significantly inhibited poly(I:C)-induced inflammasome activation, in terms of secretion of active form of IL-1ß and caspase-1. Furthermore, RA markedly inhibited poly(I:C)-induced NLRP3 and ASC expression. SIGNIFICANCE: These results indicate that RA can inhibit poly(I:C)-induced inflammatory reaction of keratinocytes, and suggest that it may be a potential candidate for the treatment of psoriasis.


Subject(s)
Cinnamates/pharmacology , Depsides/pharmacology , Inflammation/prevention & control , Keratinocytes/drug effects , Poly I-C/pharmacology , Cells, Cultured , Cytokines/metabolism , Humans , Keratinocytes/metabolism , Poly I-C/antagonists & inhibitors , Rosmarinic Acid
7.
Arch Biochem Biophys ; 585: 98-108, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26321373

ABSTRACT

Tyrosine phosphorylation of insulin receptor beta (IRß) in insulin treated HepG2 cells is inversely correlated to ser(51) phosphorylation in the alpha-subunit of eukaryotic initiation factor 2 (eIF2α) that regulates protein synthesis. Insulin stimulates interaction between IRß and PKR, double stranded RNA-dependent protein kinase, also known as EIF2AK2, and phosphorylation of tyrosine residues in PKR, as analyzed by immunoprecipitation and pull down assays using anti-IRß and anti-phosphotyrosine antibodies, recombinant IRß and immunopurified PKR. Further polyIC or synthetic double stranded RNA-induced threonine phosphorylation or activation of immunopurified and cellular PKR is suppressed in the presence of insulin treated purified IRß and cell extracts. Acute, but not chronic, insulin treatment enhances tyrosine phosphorylation of IRß, its interaction with PKR and tyrosine phosphorylation of PKR. In contrast, lipopolysaccharide that stimulates threonine phosphorylation of PKR and eIF2α phosphorylation and AG 1024, an inhibitor of the tyrosine kinase activity of IRß, reduces PKR association with the receptor, IRß in HepG2 cells. These findings therefore may suggest that tyrosine phosphorylated PKR plays a role in the regulation of insulin induced protein synthesis and in maintaining insulin sensitivity, whereas, suppression of polyIC-mediated threonine phosphorylation of PKR by insulin compromises its ability to fight against virus infection in host cells.


Subject(s)
Insulin/pharmacology , Poly I-C/antagonists & inhibitors , Protein Biosynthesis , Threonine/metabolism , Tyrosine/metabolism , eIF-2 Kinase/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Hep G2 Cells , Humans , Lipopolysaccharides/pharmacology , Phosphorylation , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Tyrphostins/pharmacology , eIF-2 Kinase/genetics
8.
Bioorg Med Chem Lett ; 25(16): 3179-83, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26094120

ABSTRACT

Scavenger receptor A (SRA) has been implicated in the processes of tumor invasion and acts as an immunosuppressor during therapeutic cancer vaccination. Pharmacological inhibition of SRA function thus holds a great potential to improve treatment outcome of cancer therapy. Macromolecular natural product sennoside B was recently shown to block SRA function. Here we report the identification and characterization of a small molecule SRA inhibitor rhein. Rhein, a deconstructed analog of sennoside B, reversed the suppressive activity of SRA in dendritic cell-primed T cell activation, indicated by transcription activation of il2 gene and production of IL-2. Rhein also inhibited SRA ligand polyinosinic:polycytidylic acid (poly(I:C)) induced activation of transcriptional factors, including interferon regulatory factor 3 (IRF3) and signal transducer and activator of transcription 1 (STAT1). Additionally, this newly identified lead compound was docked into the homology models of the SRA cysteine rich domain to gain insights into its interaction with the receptor. It was then found that rhein can favorably interact with SRA cysteine rich domain. Collectively, rhein, being the first identified small molecule inhibitors for SRA, warrants further structure-activity relationship studies, which may lead to development of novel pharmacological intervention for cancer therapy.


Subject(s)
Anthraquinones/chemical synthesis , Anthraquinones/pharmacology , Scavenger Receptors, Class A/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Dendritic Cells/drug effects , Drug Design , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Poly I-C/antagonists & inhibitors , Senna Extract/chemistry , Senna Extract/pharmacology , Sennosides , Small Molecule Libraries , Structure-Activity Relationship , T-Lymphocytes/drug effects , Toll-Like Receptors/drug effects , Transcription Factors/drug effects , beta-Galactosidase/antagonists & inhibitors
9.
Eur J Pharmacol ; 754: 11-8, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25704611

ABSTRACT

Inflammation has been implicated in the pathogenesis of various cerebral diseases. Thus, control of brain inflammation is regarded as one of the important therapeutic strategies for the treatment of neurodegenerative diseases such as Alzheimer׳s disease and stroke. Isobavachalcone, a flavonoid from Psoralea corylifolia, is known to possess a wide spectrum of biological activities and is expected to be useful in preventing or treating neurodegenerative diseases. However, very little is known regarding its effects on cerebral inflammation. In this study, we examined the effect of isobavachalcone on leukocyte adhesion and intercellular adhesion molecule-1 (ICAM-1) expression in brain endothelial cells activated with lipopolysaccharide (LPS) and explored the possible mechanisms involved. Isobavachalcone significantly down-regulated LPS-induced ICAM-1 expression and leukocyte-endothelial cell adhesion and suppressed NF-κB activity which is implicated in the expression of ICAM-1. It attenuated ICAM-1 expression as well as NF-κB transcriptional activity induced by macrophage-activating lipopeptide 2-kDa (MALP-2) or polyriboinosinic polyribocytidylic acid (poly[I:C]). Isobavachalcone also down-regulated LPS or poly[I:C]-induced expression of IFN-ß, which can indirectly activate NF-κB. These data imply that isobavachalcone can modulate both MyD88-dependent and TRIF-dependent signaling of toll-like receptor 4 (TLR4). Taken together, our data suggest that isobavachalcone inhibits LPS-induced ICAM-1 expression and leukocyte adhesion to brain endothelial cell by blocking TLR4 signaling and thus, has the potential to ameliorate neuronal injury in brain diseases associated with inflammation.


Subject(s)
Brain/cytology , Chalcones/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Brain/drug effects , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Lipopeptides/antagonists & inhibitors , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology
10.
Brain Behav Immun ; 33: 190-200, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23876745

ABSTRACT

Prenatal maternal infection is an environmental risk factor for neurodevelopmental psychiatric illness and disease-associated cognitive impairments. Modeling this epidemiological link in animals shows that prenatal immune challenge is capable of inducing long-lasting deficits in numerous cognitive domains. Here, we combined a neonatal cross-fostering design with a mouse model of prenatal immune challenge induced by maternal gestational treatment with the viral mimetic poly(I:C) to dissect the relative contribution of prenatal and postnatal maternal effects on the offspring. We show that offspring prenatally exposed to poly(I:C) display significant impairments in spatial matching-to-position working memory and spatial novelty presence regardless of whether they are raised by gestationally immune-challenged or non-challenged control surrogate mothers. Likewise, prenatally immune challenged offspring exhibit reduced glutamic acid decarboxylase 65-kDa (GAD65) and 67-kDa (GAD67) gene expression in the adult medial prefrontal cortex and dorsal hippocampus largely independently of the postnatal rearing conditions. In addition, we confirm that being raised by a gestationally immune-challenged surrogate mother is sufficient to increase the offspring's locomotor response to systemic amphetamine treatment. Our data thus suggest that prenatal infection-induced deficits in spatial short-term memory are mediated by prenatal maternal effects on the offspring. At the same time, our study adds further weight to the notion that being reared by a surrogate mother that experienced immune activation during pregnancy may constitute a risk factor for specific dopaminergic abnormalities.


Subject(s)
Glutamate Decarboxylase/deficiency , Maternal Exposure , Animals , Animals, Newborn/immunology , Down-Regulation/genetics , Down-Regulation/immunology , Female , Glutamate Decarboxylase/genetics , Hippocampus/enzymology , Hippocampus/immunology , Hippocampus/pathology , Male , Maternal Exposure/adverse effects , Memory Disorders/enzymology , Memory Disorders/genetics , Memory Disorders/immunology , Mice , Mice, Inbred C57BL , Poly I-C/antagonists & inhibitors , Poly I-C/toxicity , Postnatal Care , Prefrontal Cortex/enzymology , Prefrontal Cortex/immunology , Prefrontal Cortex/pathology , Pregnancy , Prenatal Diagnosis , Random Allocation , gamma-Aminobutyric Acid/genetics
11.
J Sci Food Agric ; 93(3): 646-51, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23015382

ABSTRACT

BACKGROUND: Propolis is a bee product with various biological properties, including an antiviral activity when taken orally. However, its mechanisms at the cellular and molecular level are not well understood. RESULTS: We investigated the effect of propolis on antiviral signaling in A549 cells transfected with double-stranded RNA (dsRNA), a model for viral infection. Pretreatment of the cells with propolis inhibited poly I:C (synthetic dsRNA)-induced interferon (IFN)-ß expression. Propolis had no effect on the dsRNA-induced expression of RIG-I-like receptors (RLRs), which are known as intracellular viral RNA sensors. As to the effect on antiviral executor genes, propolis enhanced myxovirus resistance 1 (MX1) expression, whereas interferon-inducible gene 6-16 (G1P3) and 2'-5'-oligoadenylate synthetase (OAS) were unaffected. All of these genes belong to the IFN-inducible genes, suggesting that the effect of propolis on antiviral signaling is not necessarily mediated by the autocrine regulation by IFN-ß. Propolis pretreatment inhibited dsRNA-induced interleukin-8 (IL8) and CCL5 expression, and consequently lowered polymorphonuclear leukocyte (PMN) chemotactic activity in the cell-conditioned medium. CONCLUSION: Taken together, these results suggest that propolis may suppress excess inflammatory responses without affecting the innate immunity during viral infection.


Subject(s)
Antiviral Agents/pharmacology , Interferon-beta/genetics , Neutrophils/drug effects , Propolis/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , Adenocarcinoma , Brazil , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Culture Media, Conditioned , Gene Expression/drug effects , Humans , Interferon-beta/antagonists & inhibitors , Lung Neoplasms , Neutrophils/physiology , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , RNA, Double-Stranded/physiology , Transfection
12.
J Immunol ; 189(6): 2768-73, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22904303

ABSTRACT

RNA receptors such as TLR3 and retinoid acid-inducible gene I/melanoma differentiation-associated gene 5 play essential roles in innate immunity to RNA viruses. However, how innate immunity to RNAs is controlled at the molecular level is not well understood. We describe in this study a new regulatory pathway of anti-RNA immunity that is composed of PI3K, its target GTPase Rac, and the newly described immune regulator TNF-α-induced protein 8 like-2 (TIPE2, or TNFAIP8L2). Polyinosinic-polycytidylic acid [Poly (I:C)], a dsRNA receptor ligand, activates Rac via its guanine nucleotide exchange factor Tiam; this leads to the activation of cytokine genes and, paradoxically, downregulation of the Tipe2 gene. TIPE2 is a negative regulator of immunity; its deficiency leads to hyperactivation of the PI3K-Rac pathway as exemplified by enhanced AKT, Rac, P21-activated kinase, and IFN regulatory factor 3 activities. As a consequence, TIPE2 knockout myeloid cells are hyperreactive to Poly (I:C) stimulation, and TIPE2 knockout mice are hypersensitive to Poly (I:C)-induced lethality. These results indicate that TIPE2 controls innate immunity to RNA by targeting the PI3K-Rac pathway. Therefore, manipulating TIPE2 or Rac functions can be effective for controlling RNA viral infections.


Subject(s)
Immunity, Innate , Intracellular Signaling Peptides and Proteins/physiology , Phosphatidylinositol 3-Kinase/physiology , Signal Transduction/immunology , rac GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , L Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoinositide-3 Kinase Inhibitors , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Signal Transduction/genetics , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology
14.
J Immunol ; 187(10): 5357-62, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21964025

ABSTRACT

Excessive inflammation during bacterial and viral infections is destructive to the host and involves elevated production of proinflammatory cytokines. It is especially deleterious in organs with space constraints such as lung and the CNS. Indeed, a number of viruses that infect lungs, such as avian influenza virus, SARS-associated coronavirus, and respiratory syncytial virus, elicit a very high level of proinflammatory cytokines; however, it is unclear what triggers their production. In this study, we show that IL-17 commonly produced during viral infection specifically augments a proinflammatory response by directly synergizing with antiviral signaling. Costimulation of primary human fibroblasts with IL-17 greatly enhanced respiratory syncytial virus-induced or synthetic dsRNA-based viral mimic polyinosinic:polycytidylic acid-induced expression of proinflammatory genes without affecting expression of IFN-ß-stimulated or IFN-stimulated genes. Knockdown of expression of known mediators of the antiviral signaling pathway revealed that the IL-17-poly(I:C) synergy depends on the presence of the transcriptional factors RelA and IFN regulatory factor 3 and IκB kinases. Moreover, this synergy was blocked by an IκB kinase inhibitor, BAY 11-7082. These findings shed light on the molecular mechanisms behind IL-17-dependent immunopathology observed in viral infections.


Subject(s)
Antiviral Agents/pharmacology , Fibroblasts/immunology , Fibroblasts/pathology , Inflammation Mediators/physiology , Interleukin-17/physiology , Respiratory Syncytial Viruses/immunology , Up-Regulation/immunology , Antiviral Agents/metabolism , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/physiology , Fibroblasts/virology , Gene Expression Regulation/immunology , Humans , I-kappa B Kinase/biosynthesis , I-kappa B Kinase/genetics , I-kappa B Kinase/physiology , Inflammation Mediators/metabolism , Interferon Regulatory Factor-3/biosynthesis , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/physiology , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Signal Transduction/genetics , Signal Transduction/immunology , Skin/immunology , Skin/pathology , Skin/virology , Transcription Factor RelA/biosynthesis , Transcription Factor RelA/genetics , Transcription Factor RelA/physiology
15.
J Immunol ; 183(5): 2984-94, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19648269

ABSTRACT

Tolerogenic dendritic cells (TDC) offer a promising therapeutic potential to ameliorate autoimmune diseases. Reported to inhibit adaptive immune responses, little is known about their innate immunity receptor repertoire. In this study, we compared three types of human TDC (IL-10-DC, dexamethasone (DX)-DC, and 1,25(OH)(2)D(3)-DC) by their TLR expression and response to a set of TLR ligands. TDC are endowed with the same TLR set as standard monocyte-derived dendritic cells but respond differentially to the TLR stimuli Pam3CSK4, polyinosinic-polycytidylic acid, LPS, and flagellin. TDC expressed low or no IL-12-related cytokines and remarkably elevated IL-10 levels. Interestingly, only TDC up-regulated the expression of TLR2 upon stimulation. This boosted the tolerogenic potential of these cells, because IL-10 production was up-regulated in TLR2-stimulated, LPS-primed DX-DC, whereas IL-12 and TNF-alpha secretion remained low. When comparing the TDC subsets, DX-DC and 1,25(OH)(2)D(3)-DC up-regulated TLR2 irrespective of the TLR triggered, whereas in IL-10-DC this effect was only mediated by LPS. Likewise, DX-DC and 1,25(OH)(2)D(3)-DC exhibited impaired ability to mature, reduced allostimulatory properties, and hampered capacity to induce Th1 differentiation. Therefore, both DX-DC and 1,25(OH)(2)D(3)-DC display the strongest tolerogenic and anti-inflammatory features and might be most suitable tools for the treatment of autoimmune diseases.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Inflammation Mediators/antagonists & inhibitors , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/blood , Up-Regulation/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/classification , Down-Regulation/immunology , Feedback, Physiological/immunology , Flagellin/antagonists & inhibitors , Flagellin/metabolism , Humans , Inflammation Mediators/blood , Inflammation Mediators/physiology , Ligands , Lipopeptides/antagonists & inhibitors , Lipopeptides/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/metabolism , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Poly I-C/antagonists & inhibitors , Poly I-C/metabolism , Toll-Like Receptor 2/agonists
16.
Biochem Biophys Res Commun ; 374(3): 431-6, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18639521

ABSTRACT

Curcumin, a compound found in the Indian spice turmeric, has anti-inflammatory and immunomodulatory properties, though the mechanism remains unclear. Dendritic cells (DCs) are important to generating an immune response and the effect of curcumin on human DCs has not been explored. The role curcumin in the DC response to bacterial and viral infection was investigated in vitro using LPS and Poly I:C as models of infection. CD14(+) monocytes, isolated from human peripheral blood, were cultured in GM-CSF- and IL-4-supplemented medium to generate immature DCs. Cultures were incubated with curcumin, stimulated with LPS or Poly I:C and functional assays were performed. Curcumin prevents DCs from responding to immunostimulants and inducing CD4(+) T cell proliferation by blocking maturation marker, cytokine and chemokine expression and reducing both migration and endocytosis. These data suggest a therapeutic role for curcumin as an immune suppressant.


Subject(s)
Adjuvants, Immunologic/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Dendritic Cells/drug effects , Immunosuppressive Agents/pharmacology , Adjuvants, Immunologic/pharmacology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Chemokines/metabolism , Dendritic Cells/immunology , Endocytosis/drug effects , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/immunology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Poly I-C/antagonists & inhibitors , Poly I-C/immunology
17.
J Immunol ; 179(6): 4142-52, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785853

ABSTRACT

The present study underlines the importance of PI3K in mediating the anti-inflammatory effect of gemfibrozil, a prescribed lipid-lowering drug for humans, in mouse microglia. Gemfibrozil inhibited LPS-induced expression of inducible NO synthase (iNOS) and proinflammatory cytokines in mouse BV-2 microglial cells and primary microglia. By overexpressing wild-type and dominant-negative constructs of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in microglial cells and isolating primary microglia from PPAR-alpha-/- mice, we have demonstrated that gemfibrozil inhibits the activation of microglia independent of PPAR-alpha. Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Conversely, overexpression of the constitutively active mutant of p110 enhanced the inhibitory effect of gemfibrozil on LPS-induced expression of proinflammatory molecules. Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Inhibition of PI3K also abolished the inhibitory effect of gemfibrozil on Abeta-, PrP-, poly IC-, Tat-, and MPP+-induced microglial expression of iNOS. Involvement of NF-kappaB activation in LPS-, Abeta-, PrP-, poly IC-, Tat-, and MPP+-, but not IFN-gamma-, induced microglial expression of iNOS and stimulation of IkappaBalpha expression and inhibition of NF-kappaB activation by gemfibrozil via the PI3K pathway suggests that gemfibrozil inhibits the activation of NF-kappaB and the expression of proinflammatory molecules in microglia via PI3K-mediated up-regulation of IkappaBalpha.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gemfibrozil/pharmacology , I-kappa B Proteins/biosynthesis , Microglia/drug effects , Microglia/enzymology , Phosphatidylinositol 3-Kinases/physiology , Up-Regulation/drug effects , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/physiology , Androstadienes/pharmacology , Animals , Cell Line , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Enzyme Activation/drug effects , Gemfibrozil/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Microglia/metabolism , NF-KappaB Inhibitor alpha , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , PPAR alpha/physiology , Phosphatidylinositol 3-Kinases/metabolism , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Prions/antagonists & inhibitors , Prions/physiology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Double-Stranded/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/immunology , Wortmannin
18.
Reprod Biol Endocrinol ; 3: 74, 2005 Dec 29.
Article in English | MEDLINE | ID: mdl-16384532

ABSTRACT

BACKGROUND: The human endometrium is an important site for contact between the host and pathogens ascending the reproductive tract, and thus plays an important role in female reproductive tract immunity. Previous work in our laboratory has suggested that Toll-like receptors (TLRs) are involved in endometrial epithelial recognition of pathogens and that ligation of endometrial TLRs results in the production of cytokines and chemokines important for both immune and reproductive functions of the endometrium. We have also demonstrated cyclic regulation of TLR3 mRNA and protein expression in human endometrium, suggesting that steroid hormones might play a role in the expression and function of TLR3. In this study, the effects of 17beta-estradiol (E2) and progesterone (P) on TLR3 expression and function in endometrial cell lines were investigated. METHODS: Endometrial epithelial cell lines were cultured and examined for the presence of TLR3 and hormone receptors by endpoint RT-PCR. For hormonal studies, cells were pre-treated with ethanol vehicle, 10(-8) M E2, and/or 10(-7) M P. For antagonist assays, cells were treated with the ER antagonist, ICI 182, 780, or the PR antagonist, RU486, for two hours prior to treatment with hormones. Following hormone or hormone/antagonist pre-treatment, cells were stimulated with vehicle, the synthetic TLR3 ligand, polyinosinic-polycytidylic acid (Poly I:C), a negative dsDNA control, or a positive control. Cytokine and chemokine production post-stimulation was measured by ELISA. The effects of E2 and P on TLR3 mRNA and protein expression were measured using Real Time RT-PCR and FACS analysis, respectively. RESULTS: Stimulation of TLR3-expressing cells with the synthetic TLR3 ligand, Poly I:C, resulted in the production of cytokines and chemokines important for endometrial function and regulation. Suppression of Poly I:C-induced cytokine and chemokine production by cells treated with 10(-8) M E2, but not cells treated with 10(-7) M P, was observed in endometrial epithelial cell lines expressing TLR3 and estrogen receptor alpha (ERalpha). The effects of E2 were not observed on cells which did not express ERalpha or in cells pre-treated with the ER antagonist, ICI 182, 780. Treatment with E2 did not affect TLR3 mRNA or protein expression. However, treatment with E2 did suppress cytokine and chemokine production resulting from TLR3 stimulation with Poly I:C, suggesting that E2 modulates TLR3 function. CONCLUSION: The data presented in this study are the first indication that E2 can markedly alter the innate immune response to dsRNA, providing a previously unreported process by which E2 can alter immune responses.


Subject(s)
Chemokines/biosynthesis , Cytokines/biosynthesis , Estradiol/pharmacology , Toll-Like Receptor 3/biosynthesis , Cell Line , Cell Line, Tumor , Endometrium/cytology , Epithelial Cells , Estradiol/analogs & derivatives , Female , Fulvestrant , Humans , Mifepristone/pharmacology , Poly I-C/antagonists & inhibitors , Poly I-C/pharmacology , Polydeoxyribonucleotides/pharmacology , Progesterone/pharmacology , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology , Toll-Like Receptor 3/drug effects
19.
Alcohol ; 28(2): 75-81, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12457938

ABSTRACT

Natural killer (NK) cells can be activated in vitro and in vivo by polyinosinic:polycytidylic acid (poly I:C) through induction of type I interferons or other cytokines. Ethanol suppresses in vivo and ex vivo poly I:C activation of NK cell activity in a mouse model for binge drinking, but it is not known whether this effect is mediated by changes in NK cells or in other cell types (e.g., those that produce NK cell-activating cytokines). Splenocytes were obtained from C57BL/6 [NK cell-competent (NKc)] and C57BL/6 perforin knockout [NK cell-incompetent (NKi)] mice 6 h after administration of ethanol (6 g/kg) or vehicle (VH; dH(2)O). Cells were incubated in vitro 18 h with poly I:C (100 micro g/ml), followed by a 4-h 51Cr release assay with the use of YAC-1 target cells. Results of cell-mixing experiments involving all relevant combinations of splenocytes obtained from NKc and NKi mice treated with VH or ethanol strongly supported the suggestion that NK cells, not other cell types, are the primary target of ethanol-induced suppression of NK cell activation. For example, mixing of splenocytes obtained from ethanol-treated NKc and VH-treated NKi mice or from ethanol-treated NKc and ethanol-treated NKi mice yielded similar cytolytic function. However, mixing of splenocytes obtained from ethanol-treated NKc and VH-treated NKi mice yielded significantly less cytolytic activity than that of splenocytes from VH-treated NKc and ethanol-treated NKi mice. In addition, mixing of splenocytes obtained from VH-treated NKc and NKi mice resulted in lower cytolytic activity than when splenocytes from the NKi mice were treated with ethanol instead of with VH, demonstrating that ethanol did not decrease the function of other cell types. A strikingly similar pattern of results was observed when B6C3F1 mice, rendered NK cell deficient by administration of anti-NK 1.1 monoclonal antibody, were used instead of perforin knockout mice. These results indicate that ethanol suppresses activation of NK cells primarily by suppressing the NK cell response to poly I:C, not by acting on another cell type.


Subject(s)
Ethanol/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Poly I-C/pharmacology , Animals , Female , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Perforin , Poly I-C/antagonists & inhibitors , Pore Forming Cytotoxic Proteins
20.
J Leukoc Biol ; 72(5): 953-61, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429717

ABSTRACT

Here, we investigated the influence of cyclosporin A (CsA) on dendritic cell (DC) generation. With this aim, human DC were propagated from monocytes in serum-free medium with granulocyte macrophage-colony stimulating factor and interleukin-4. DC were then exposed to tumor necrosis factor alpha (TNF-alpha) for maturation. Our results show that CsA does not impair commitment of monocytes into DC, as assessed by loss of CD14 and increase of CD40 and CD1a. However, TNF-alpha-induced DC maturation was affected, as CsA-treated DC expressed lower levels of human leukocyte antigen and costimulatory molecules but sustained levels of CD1a, and less DC expressed DC-lysosomal-associated-membrane-protein (LAMP) and CD83. Accordingly, CsA inhibited the allostimulatory and accessory cell functions of DC. Surprisingly, when other maturation stimuli were used, we observed that CsA significantly inhibited maturation induced by lipopolysaccharides but not by polyribocytidylic acid or CD40 ligand, as assessed by DC phenotype and functions. Therefore, our results indicate that CsA may differentially affect DC maturation.


Subject(s)
Cyclosporine/pharmacology , Dendritic Cells/immunology , Immunosuppressive Agents/pharmacology , Antigen Presentation/drug effects , Antigens, CD/analysis , CD40 Ligand/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , HLA-DR Antigens/analysis , Kinetics , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Culture Test, Mixed , Poly I-C/antagonists & inhibitors , RNA, Double-Stranded/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...