Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Microbiol Spectr ; 9(3): e0149721, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34937192

ABSTRACT

Tumors and infectious agents both benefit from an immunosuppressive environment. Cutibacterium acnes (C. acnes) is a bacterium in the normal skin microbiota, which has the ability to survive intracellularly in macrophages and is significantly more common in prostate cancer tissue compared with normal prostate tissue. This study investigated if prostate cancer tissue culture positive for C. acnes has a higher infiltration of regulatory T-cells (Tregs) and if macrophages stimulated with C. acnes induced the expression of immunosuppressive genes that could be linked to an increase of Tregs in prostate cancer. Real-time PCR and enzyme-linked immunosorbent spot assay (ELISA) were used to examine the expression of immunosuppressive genes in human macrophages stimulated in vitro with C. acnes, and associations between the presence of C. acnes and infiltration of Tregs were investigated by statistically analyzing data generated in two previous studies. The in vitro results demonstrated that macrophages stimulated with C. acnes significantly increased their expression of PD-L1, CCL17, and CCL18 mRNA and protein (p <0.05). In the cohort, Tregs in tumor stroma and tumor epithelia were positively associated with the presence of C. acnes (P = 0.0004 and P = 0.046, respectively). Since the macrophages stimulated with C. acnes in vitro increased the expression of immunosuppressive genes, and prostate cancer patients with prostatic C. acnes infection had higher infiltration of Tregs than their noninfected counterparts, we suggest that C. acnes may contribute to an immunosuppressive tumor environment that is vital for prostate cancer progression. IMPORTANCE In an immune suppressive tumor microenvironment constituted by immunosuppressive cells and immunosuppressive mediators, tumors may improve their ability to give rise to a clinically relevant cancer. In the present study, we found that C. acnes might contribute to an immunosuppressive environment by recruiting Tregs and by increasing the expression of immunosuppressive mediators such as PD-L1, CCL17, and CCL18. We believe that our data add support to the hypothesis of a contributing role of C. acnes in prostate cancer development. If established that C. acnes stimulates prostate cancer progression it may open up avenues for targeted prostate cancer treatment.


Subject(s)
Immune Tolerance/immunology , Macrophages/immunology , Propionibacteriaceae/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/microbiology , T-Lymphocytes, Regulatory/immunology , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Chemokine CCL17/biosynthesis , Chemokine CCL17/genetics , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Enzyme-Linked Immunospot Assay , Humans , Immune Tolerance/genetics , Male , Microbiota/immunology , Prostatic Neoplasms/pathology , Tumor Escape/immunology , Tumor Microenvironment/immunology
2.
Int J Mol Sci ; 22(12)2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34208434

ABSTRACT

Skullcapflavone II (SFII), a flavonoid derived from Scutellaria baicalensis, has been reported to have anti-inflammatory properties. However, its therapeutic potential for skin inflammatory diseases and its mechanism are unknown. Therefore, this study aimed to investigate the effect of SFII on TNF-α/IFN-γ-induced atopic dermatitis (AD)-associated cytokines, such as thymus- and activation-regulated chemokine (TARC) and macrophage-derived chemokine (MDC). Co-stimulation with TNF-α/IFN-γ in HaCaT cells is a well-established model for induction of pro-inflammatory cytokines. We treated cells with SFII prior to TNF-α/IFN-γ-stimulation and confirmed that it significantly inhibited TARC and MDC expression at the mRNA and protein levels. Additionally, SFII also inhibited the expression of cathepsin S (CTSS), which is associated with itching in patients with AD. Using specific inhibitors, we demonstrated that STAT1, NF-κB, and p38 MAPK mediate TNF-α/IFN-γ-induced TARC and MDC, as well as CTSS expression. Finally, we confirmed that SFII significantly suppressed TNF-α/IFN-γ-induced phosphorylation of STAT1, NF-κB, and p38 MAPK. Taken together, our study indicates that SFII inhibits TNF-α/IFN-γ-induced TARC, MDC, and CTSS expression by regulating STAT1, NF-κB, and p38 MAPK signaling pathways.


Subject(s)
Cathepsins/biosynthesis , Chemokine CCL17/biosynthesis , Chemokine CCL22/biosynthesis , Flavonoids/pharmacology , Interferon-gamma/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cathepsins/genetics , Cell Survival/drug effects , Chemokine CCL17/genetics , Chemokine CCL22/genetics , Gene Expression Regulation/drug effects , HaCaT Cells , Humans , Keratinocytes/metabolism , NF-kappa B/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Front Immunol ; 11: 1520, 2020.
Article in English | MEDLINE | ID: mdl-32765525

ABSTRACT

Objectives: GM-CSF is a pro-inflammatory cytokine with multiple actions predominantly on myeloid cells. Enhanced GM-CSF expression by lymphocytes from patients with Ankylosing Spondylitis (AS) has recently been described, however, its potential pathogenic role(s) in AS are unknown. Methods: The effects of GM-CSF on TNF, IL-23, and CCL17 production by blood, PBMCs and isolated CD14+ monocytes from AS patients and healthy controls (HCs) were studied using ELISA. Serum CCL17 and GM-CSF and T cell GM-CSF production were studied in AS patients including pre-and on TNFi therapy. Results: GM-CSF markedly increased TNF production by LPS-stimulated whole blood, peripheral blood mononuclear cells (PBMC) and purified monocytes from AS patients, with 2 h GM-CSF exposure sufficient for monocyte "priming." Blocking of GM-CSF significantly reduced the production of TNF by whole blood from AS patients but not HCs. GM-CSF priming increased IL-23 production from LPS-stimulated AS and HC whole blood 5-fold, with baseline and stimulated IL-23 levels being significantly higher in AS whole blood. GM-CSF also stimulated CCL17 production from AS and HC blood and CCL17 levels were elevated in AS plasma. GM-CSF could be detected in plasma from 14/46 (30%) AS patients compared to 3/18 (17%) HC. Conclusion: We provide evidence that GM-CSF primes TNF and IL-23 responses in myeloid cells from AS patients and HC. We also show CCL17 levels, downstream of GM-CSF, were elevated in plasma samples of AS patients. Taken together these observations are supportive of GM-CSF neutralization as a potential novel therapeutic approach for the treatment of AS.


Subject(s)
Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation Mediators/metabolism , Monocytes/immunology , Monocytes/metabolism , Spondylitis, Ankylosing/etiology , Spondylitis, Ankylosing/metabolism , Biomarkers , Case-Control Studies , Chemokine CCL17/biosynthesis , Disease Susceptibility , Gene Expression Regulation , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/immunology , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/pathology , Toll-Like Receptors/agonists , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor Inhibitors/therapeutic use
4.
Cancer Lett ; 453: 184-192, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30953706

ABSTRACT

Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphomas associated with chronic inflammation (DLBCL-CI) develop in patients with chronic inflammation but without any predisposing immunodeficiency. Given the expression of the EBV latent genes, DLBCL-CI should have mechanisms for evasion of host antitumor immunity. EBV-positive pyothorax-associated lymphoma (PAL) is a prototype of DLBCL-CI and may provide a valuable model for the study of immune evasion by DLBCL-CI. This study demonstrates that PAL cell lines express and secrete CCL17 and/or CCL22 chemokines, the ligands of C-C motif chemokine receptor 4 (CCR4), in contrast to EBV-negative DLBCL cell lines. Accordingly, culture supernatants of PAL cell lines efficiently attracted CCR4-positive regulatory T (Treg) cells in human peripheral blood mononuclear cells. PAL cells injected into mice also attracted CCR4-expressing Treg cells. Furthermore, this study confirmed that CCR4-expressing Treg cells were abundantly present in primary PAL tissues. Collectively, these findings provide new insight into the mechanisms of immune evasion by PAL, and further studies are warranted on whether such mechanisms eventually lead to the development of DLBCL-CI.


Subject(s)
Chemokine CCL17/biosynthesis , Chemokine CCL22/biosynthesis , Empyema, Pleural/immunology , Epstein-Barr Virus Infections/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Chemokine CCL17/immunology , Chemokine CCL22/immunology , Empyema, Pleural/pathology , Empyema, Pleural/virology , Epstein-Barr Virus Infections/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/virology , Male , Mice , Mice, Inbred BALB C , Receptors, CCR4/biosynthesis , Receptors, CCR4/immunology
5.
Front Immunol ; 9: 1906, 2018.
Article in English | MEDLINE | ID: mdl-30197642

ABSTRACT

Arachidonate 15-lipoxygenase (ALOX15) and arachidonate 15-lipoxygenase, type B (ALOX15B) catalyze the dioxygenation of polyunsaturated fatty acids and are upregulated in human alternatively activated macrophages (AAMs) induced by Th2 cytokine interleukin-4 (IL-4) and/or interleukin-13. Known primarily for roles in bioactive lipid mediator synthesis, 15-lipoxygenases (15-LOXs) have been implicated in various macrophage functions including efferocytosis and ferroptosis. Using a combination of inhibitors and siRNAs to suppress 15-LOX isoforms, we studied the role of 15-LOXs in cellular cholesterol homeostasis and immune function in naïve and AAMs. Silencing or inhibiting the 15-LOX isoforms impaired sterol regulatory element binding protein (SREBP)-2 signaling by inhibiting SREBP-2 processing into mature transcription factor and reduced SREBP-2 binding to sterol regulatory elements and subsequent target gene expression. Silencing ALOX15B reduced cellular cholesterol and the cholesterol intermediates desmosterol, lanosterol, 24,25-dihydrolanosterol, and lathosterol as well as oxysterols in IL-4-stimulated macrophages. In addition, attenuating both 15-LOX isoforms did not generally affect IL-4 gene expression but rather uniquely impacted IL-4-induced CCL17 production in an SREBP-2-dependent manner resulting in reduced T cell migration to macrophage conditioned media. In conclusion, we identified a novel role for ALOX15B, and to a lesser extent ALOX15, in cholesterol homeostasis and CCL17 production in human macrophages.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Chemokine CCL17/biosynthesis , Cholesterol/metabolism , Homeostasis , Macrophages/immunology , Macrophages/metabolism , Arachidonate 15-Lipoxygenase/genetics , Cell Movement/genetics , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Humans , Lipid Metabolism , Protein Binding , RNA, Small Interfering/genetics , Serum Response Element , Sterol Regulatory Element Binding Protein 2/metabolism
6.
Benef Microbes ; 9(4): 643-652, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29798706

ABSTRACT

Lactobacillus strains, a major group of lactic acid bacteria, are representative food microorganisms that have many potential beneficial effects via their interactions with immune and intestinal epithelial cells. However, little is known about the effect of Lactobacillus strains on atopic dermatitis via keratinocytes, which comprise the physical barrier of the skin. In this study, we report that Lactobacillus strains have a significant suppressive effect on tumour necrosis factor (TNF)-α-induced expression and production of thymus and activation-regulated chemokine (TARC), a T helper 2 cell chemokine responsible for atopic dermatitis, in human keratinocytes. An RNA interference study showed that the effect of Lactobacillus reuteri strain Japan Collection of Microorganisms (JCM) 1112, the most suppressive strain, depended on the presence of Toll-like receptor 2 and the induction of A20 (also known as TNF-α-induced protein 3) and cylindromatosis in HaCaT cells. Topical application of a water-soluble extract of homogenised JCM 1112 cells significantly suppressed the development of house dust mite-induced atopic skin lesions and TARC expression at the lesion sites in NC/Nga mice. Our study provides new insights into the use of Lactobacillus strains as suppressive agents against keratinocyte-involved atopic inflammation of the skin.


Subject(s)
Chemokine CCL17/genetics , Dermatitis, Atopic/therapy , Keratinocytes/drug effects , Lactobacillus , Probiotics/pharmacology , Animals , Cell Line , Chemokine CCL17/biosynthesis , Dermatitis, Atopic/pathology , Deubiquitinating Enzyme CYLD/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Interferon-gamma/pharmacology , Keratinocytes/metabolism , Male , Mice , Probiotics/therapeutic use , Signal Transduction/drug effects , Skin/drug effects , Skin/pathology , Toll-Like Receptor 2/antagonists & inhibitors , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor-alpha/pharmacology
7.
Exp Lung Res ; 44(7): 332-343, 2018 09.
Article in English | MEDLINE | ID: mdl-30676129

ABSTRACT

AIM OF THE STUDY: In patients with asthma, chronic inflammatory processes and the subsequent remodeling of the airways contribute to the symptoms and the pathophysiological changes. Epithelial-mesenchymal transition (EMT) is thought to play an important role in tissue remodeling. Previous reports show that tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a cytokine of the TNF superfamily, exerts pro-inflammatory effects, and enhances transforming growth factor (TGF)-ß-induced EMT in bronchial epithelial cells. In this study, we investigated the TWEAK-induced cytokine and chemokine production in the human bronchial epithelial cell line BEAS-2B during EMT. MATERIALS AND METHODS: Quantitative real-time RT-PCR, enzyme-linked immunosorbent assays, western blotting, and immunohistochemistry were used to define the production of cytokines and chemokines. RESULTS: We found that TWEAK increases mRNA and protein levels of thymic stromal lymphopoietin (TSLP), monocyte chemoattractant protein -1 (MCP-1), regulated upon activation normal T cell express sequence (RANTES), and IL-8 in BEAS-2B bronchial epithelial cells. Moreover, co-treatment with TWEAK and TGF-ß1 induces not only features of EMT but also enhances the production of TSLP and RANTES. Thymus- and activation-regulated chemokines (TARC) production is induced by the co-treatment of TWEAK and TGF-ß1 but not by TWEAK or TGF-ß1 stimulation alone. Furthermore, the increased mRNA expression of TSLP and RANTES after co-treatment with TWEAK and TGF-ß1 is prevented by inhibitors of Smad-independent signaling pathways. CONCLUSIONS: In the present study, we have revealed a novel mechanism for the production of asthma-related cytokines and chemokines in EMT driven by the co-stimulation with TWEAK and TGF-ß1. We conclude that cellular EMT processes caused by TWEAK and TGF-ß1 may contribute to chronic airway inflammation and remodeling.


Subject(s)
Chemokine CCL17/biosynthesis , Chemokine CCL5/biosynthesis , Cytokine TWEAK/pharmacology , Cytokines/biosynthesis , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta1/pharmacology , Airway Remodeling , Asthma/metabolism , Bronchi/cytology , Cell Line , Humans , Thymic Stromal Lymphopoietin
8.
Allergol Immunopathol (Madr) ; 45(3): 220-226, 2017.
Article in English | MEDLINE | ID: mdl-28238403

ABSTRACT

BACKGROUND: Thymus and activation-regulated chemokine (TARC), a member of the CC chemokine family, plays a crucial role in Th2-specific inflammation. We aimed to determine the concentration of sputum TARC in children with asthma and eosinophilic bronchitis (EB) and its relation with eosinophilic inflammation, pulmonary function, and bronchial hyper-responsiveness. METHODS: In total, 90 children with asthma, 38 with EB, and 45 control subjects were enrolled. TARC levels were measured in sputum supernatants using an ELISA. We performed pulmonary function tests and measured exhaled fractional nitric oxide, eosinophil counts in blood, and sputum and serum levels of total IgE in all children. RESULTS: Sputum TARC levels were significantly higher in children with asthma than in either children with EB (p=0.004) or the control subjects (p=0.014). Among patients with asthma, sputum TARC concentration was higher in children with sputum eosinophilia than in those without sputum eosinophilia (p=0.035). Sputum TARC levels positively correlated with eosinophil counts in sputum, serum total IgE levels, exhaled fractional nitric, and the bronchodilator response. Negative significant correlations were found between sputum TARC and FEV1/FVC (the ratio of forced expiratory volume in one second and forced expiratory vital capacity) or PC20 (the provocative concentration of methacholine causing a 20% decrease in the FEV1). CONCLUSION: Elevated TARC levels in sputum were detected in children with asthma but not in children with EB. Sputum TARC could be a supportive marker for discrimination of asthma from EB in children showing characteristics of eosinophilic airway inflammation.


Subject(s)
Asthma/diagnosis , Bronchitis/diagnosis , Chemokine CCL17/biosynthesis , Pulmonary Eosinophilia/diagnosis , Asthma/immunology , Asthma/metabolism , Biomarkers/analysis , Bronchitis/immunology , Bronchitis/metabolism , Child , Female , Humans , Male , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Sputum/chemistry
9.
Eur J Immunol ; 46(4): 981-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26694221

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory disease controlled by the innate and adaptive immune system. To elucidate the impact of innate immune signaling in AD, we analyzed MyD88-deficient mice in a murine model of AD-like dermatitis by epicutaneous sensitization with ovalbumin (OVA). Global MyD88 deficiency led to reduced epidermal thickening and diminished accumulation of macrophages within the inflamed skin. In addition, we observed impaired emigration of Langerhans cells (LCs) out of the epidermis of MyD88-deficient mice. These findings indicate that MyD88 deficiency affects various skin-resident cell types in the AD model. Moreover, production of IFN-g, IL-17, and CCL17 was reduced in skin draining lymph node cells and OVA-specific immunoglobulin levels were lower in MyD88-deficient mice. We further investigated the role of MyD88 in keratinocytes, as keratinocytes contribute to AD pathology. Exclusive expression of MyD88 in epidermal keratinocytes partially restored LC emigration after AD induction and expression of CCL17 in skin draining lymph nodes (LNs), but did not promote epidermal thickening nor production of IL-17. Altogether, these data demonstrate that MyD88 signaling in keratinocytes is able to restore LC migration in an otherwise MyD88-deficient background, and significantly contributes to the development of AD-like dermatitis.


Subject(s)
Dermatitis, Atopic/immunology , Inflammation/immunology , Keratinocytes/metabolism , Langerhans Cells/metabolism , Myeloid Differentiation Factor 88/genetics , Animals , Antibodies/blood , Cell Movement/genetics , Chemokine CCL17/biosynthesis , Dermatitis, Atopic/genetics , Disease Models, Animal , Female , Inflammation/genetics , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Lymph Nodes/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Skin/pathology
10.
Nanotoxicology ; 9(7): 825-34, 2015.
Article in English | MEDLINE | ID: mdl-26356541

ABSTRACT

Superparamagnetic iron oxide nanoparticles (SPIONs) have attracted special attention as novel nanoprobes capable of improving both the therapy and diagnosis of lung diseases. For safe prospective clinical applications, their biocompatibility has to be assessed after intrapulmonary administration. This study was therefore conducted to understand the biological impact of SPIONs and their further surface-functionalization with polyethylene glycol (PEG) having either negative (i.e. carboxyl) or positive (i.e. amine) terminal in a 1-month longitudinal study following acute and sub-acute exposures. Noninvasive free-breathing MR imaging protocols were first optimized to validate SPIONs detection in the lung and investigate possible subsequent systemic translocation to abdominal organs. Pulmonary Magnetic Resonance Imaging (MRI) allowed successful in vivo detection of SPIONs in the lung using ultra-short echo time sequence. Following high-dose lung administration, MR imaging performed on abdominal organs detected transient accumulation of SPIONs in the liver. Iron quantification using Inductive coupled plasma - Mass mass spectroscopy (ICP-MS) confirmed MRI readouts. Oxidative stress induction and genotoxicity were then conducted to evaluate the biocompatibility of SPIONs with their different formulations in a mouse model. A significant increase in lipid peroxidation was observed in both acute and sub-acute sets and found to regress in a time-dependent manner. PEG functionalized SPIONs revealed a lower effect with no difference between both terminal modifications. Genotoxicity assessments revealed an increase in DNA damage and gene expression of CCL-17 and IL-10 biomarkers following SPIONs administration, which was significantly higher than surface-modified nanoparticles and decreased in a time-dependent manner. However, SPIONs with carboxyl terminal showed a slightly prominent effect compared to amine modification.


Subject(s)
Lung/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/toxicity , Materials Testing/methods , Polyethylene Glycols/pharmacology , Animals , Chemokine CCL17/biosynthesis , DNA Damage/drug effects , Gene Expression/drug effects , Interleukin-10/biosynthesis , Lung/diagnostic imaging , Lung/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/ultrastructure , Mice , Polyethylene Glycols/chemistry , Surface Properties
11.
Mol Med Rep ; 12(3): 4773-4781, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26081168

ABSTRACT

Gleditsia sinensis thorns have traditionally been used to treat edema and carbuncles and drain abscesses. In the present study, a simultaneous analysis of four flavonoids [(+)­catechin, (­)­epicatechin, eriodictyol and quercetin] and two phenolic compounds (caffeic acid and ethyl gallate), obtained from a 70% ethanol extract of G. sinensis, was performed using high­performance liquid chromatography­photodiode array techniques. In addition, the inhibitory activities of the solvent fractions from a G. sinensis extract and its major constituents on the lipopolysaccharide­stimulated production of inflammatory mediators by macrophage RAW 264.7 cells and the tumor necrosis factor (TNF)­α and interferon (IFN)­Î³ (TI)­stimulated production of chemokines by HaCaT keratinocyte cells were investigated. The established analytical method showed high linearity, with a correlation coefficient of ≥0.9998. The limits of detection and quantification of the six compounds were 0.037­0.425 and 0.124­1.418 µg/ml, respectively. The ethyl acetate fraction inhibited nitric oxide and prostaglandin E2 production in RAW 264.7 cells and the production of thymus­ and activation­regulated chemokine (TARC) in HaCaT cells more than did the other fractions. Furthermore, the six compounds reduced the production of TARC, macrophage­derived chemokine and regulated on activation normal T­cell expressed and secreted in TI­stimulated HaCaT cells; in particular, ethyl gallate and quercetin exhibited a significant dose­dependent inhibition. Further elucidation of the signaling pathways involved in the T­helper cell 2 chemokine inhibition by G. sinensis is necessary to facilitate the design of therapeutic agents for the inflammatory response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gleditsia/chemistry , Keratinocytes/drug effects , Macrophages/drug effects , Plant Extracts/pharmacology , Animals , Cell Line , Chemokine CCL17/biosynthesis , Dinoprostone/biosynthesis , Drug Evaluation, Preclinical , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Limit of Detection , Macrophages/immunology , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Plant Stems/chemistry
12.
Mol Med Rep ; 12(2): 2511-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25936350

ABSTRACT

Mesenchymal stem cells (MSCs) have been investigated in the treatment of numerous autoimmune diseases. However, the immune properties of MSCs on the development of asthma have remained to be fully elucidated. Airway dendritic cells (DCs) have an important role in the pathogenesis of allergic asthma, and disrupting their function may be a novel therapeutic approach. The present study used a mouse model of asthma to demonstrate that transplantation of MSCs suppressed features of asthma by targeting the function of lung myeloid DCs. MSCs suppressed the maturation and migration of lung DCs to the mediastinal lymph nodes, and thereby reducing the allergen-specific T helper type 2 (Th2) response in the nodes. In addition, MSC-treated DCs were less potent in activating naive and effector Th2 cells and the capacity of producing chemokine (C-C motif) ligand 17 (CCL17) and CCL22, which are chemokines attracting Th2 cells, to the airways was reduced. These results supported that MSCs may be used as a potential treatment for asthma.


Subject(s)
Asthma/therapy , Dendritic Cells/immunology , Lung/immunology , Mesenchymal Stem Cell Transplantation , Animals , Antibodies, Neutralizing/administration & dosage , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Cell Differentiation , Cell Movement , Chemokine CCL17/antagonists & inhibitors , Chemokine CCL17/biosynthesis , Chemokine CCL17/immunology , Chemokine CCL22/antagonists & inhibitors , Chemokine CCL22/biosynthesis , Chemokine CCL22/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Disease Models, Animal , Female , Humans , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Th2 Cells/immunology , Th2 Cells/pathology
13.
Eur J Immunol ; 45(7): 2042-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824485

ABSTRACT

Basophil-derived IL-4 is involved in the alternative activation of mouse monocytes, as recently shown in vivo. Whether this applies to human basophils and monocytes has not been established yet. Here, we sought to characterise the interaction between basophils and monocytes and identify the molecular determinants. A basophil-monocyte co-culture model revealed that IL-3 and basophil-derived IL-4 and IL-13 induced monocyte production of CCL17, a marker of alternative activation. Critically, IL-3 and IL-4 acted directly on monocytes to induce CCL17 production through histone H3 acetylation, but did not increase the recruitment of STAT5 or STAT6. Although freshly isolated monocytes did not express the IL-3 receptor α chain (CD123), and did not respond to IL-3 (as assessed by STAT5 phosphorylation), the overnight incubation with IL-4 (especially if associated with IL-3) upregulated CD123 expression. IL-3-activated JAK2-STAT5 pathway inhibitors reduced the CCL17 production in response to IL-3 and IL-4, but not to IL-4 alone. Interestingly, monocytes isolated from allergen-sensitised asthmatic patients exhibited a higher expression of CD123. Taken together, our data show that the JAK2-STAT5 pathway modulates both basophil and monocyte effector responses. The coordinated activation of STAT5 and STAT6 may have a major impact on monocyte alternative activation in vitro and in vivo.


Subject(s)
Basophils/immunology , Interleukin-13/immunology , Interleukin-3/immunology , Interleukin-4/immunology , Monocytes/immunology , Signal Transduction/immunology , Blotting, Western , Chemokine CCL17/biosynthesis , Chromatin Immunoprecipitation , Coculture Techniques/methods , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Real-Time Polymerase Chain Reaction
14.
Am J Respir Cell Mol Biol ; 53(5): 676-88, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25870903

ABSTRACT

Macrophages are dynamic cells that mature under the influence of signals from the local microenvironment into either classically (M1) or alternatively (M2) activated macrophages with specific functional and phenotypic properties. Although the phenotypic identification of M1 and M2 macrophages is well established in mice, this is less clear for human macrophages. In addition, the persistence and reversibility of polarized human phenotypes is not well established. Human peripheral blood monocytes were differentiated into uncommitted macrophages (M0) and then polarized to M1 and M2 phenotypes using LPS/IFN-γ and IL-4/IL-13, respectively. M1 and M2 were identified as CD64(+)CD80(+) and CD11b(+)CD209(+), respectively, by flow cytometry. Polarized M1 cells secreted IP-10, IFN-γ, IL-8, TNF-α, IL-1ß, and RANTES, whereas M2 cells secreted IL-13, CCL17, and CCL18. Functionally, M2 cells were highly endocytic. In cytokine-deficient medium, the polarized macrophages reverted back to the M0 state within 12 days. If previously polarized macrophages were given the alternative polarizing stimulus after 6 days of resting in cytokine-deficient medium, a switch in polarization was seen (i.e., M1 macrophages switched to M2 and expressed CD11b(+)CD209(+) and vice versa). In summary, we report phenotypic identification of human M1 and M2 macrophages, their functional characteristics, and their ability to be reprogrammed given the appropriate stimuli.


Subject(s)
Macrophage Activation/drug effects , Macrophages/drug effects , Monocytes/drug effects , Cell Differentiation , Chemokine CCL17/biosynthesis , Chemokine CCL17/metabolism , Chemokine CCL5/biosynthesis , Chemokine CCL5/metabolism , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/metabolism , Chemokines, CC/biosynthesis , Chemokines, CC/metabolism , Endocytosis/drug effects , Endocytosis/immunology , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immunophenotyping , Interferon-gamma/pharmacology , Interleukin-13/pharmacology , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Interleukin-4/pharmacology , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Monocytes/cytology , Monocytes/immunology , Primary Cell Culture , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
15.
Biol Pharm Bull ; 38(2): 317-20, 2015.
Article in English | MEDLINE | ID: mdl-25747991

ABSTRACT

Patients with atopic dermatitis (AD) show increased numbers of Th2 cells in their acute skin lesions and superficial skin colonization by Staphylococcus aureus. The purpose of this study was to clarify the effect of S. aureus cell wall components on Th2 chemokine production by murine Langerhans cells (LCs). Murine LCs were stimulated with peptidoglycan (PEG) and/or muramyldipeptide (MDP) for 24 or 48 h, and Th1 and Th2 chemokine production was investigated by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). PEG-stimulation of LCs induced production of the Th2 chemokine CCL17 and this was enhanced in the presence of interleukin (IL)-4. A low-molecular weight PEG fragment, MDP, did not induce CCL17 production by LCs. However, when LCs were stimulated with PEG in combination with MDP, PEG-induced CCL17 production was synergistically enhanced by MDP. Furthermore, PEG- and MDP-induced CCL17 production was enhanced to a greater extent in the presence of IL-4. These results suggest that S. aureus colonization in AD patients may enhance the Th2-prone immune response through upregulation of CCL17 production by LCs, which would occur as a result of simultaneous stimulation with PEG and MDP from S. aureus in a Th2 environment.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Chemokine CCL17/biosynthesis , Langerhans Cells/drug effects , Peptidoglycan/pharmacology , Staphylococcus aureus , Animals , Cell Wall , Cells, Cultured , Female , Langerhans Cells/metabolism , Mice, Inbred BALB C
16.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L710-8, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25659898

ABSTRACT

Pulmonary fibrosis is a progressive and fatal disease that involves the remodeling of the distal airspace and the lung parenchyma, which results in compromised gas exchange. The median survival time once diagnosed is less than three years. Interleukin (IL)-13 has been shown to play a role in a number of inflammatory and fibrotic diseases. IL-13 modulates its effector functions via a complex receptor system that includes the IL-4 receptor (R) α, IL-13Rα1, and the IL-13Rα2. IL-13Rα1 binds IL-13 with low affinity, yet, when it forms a complex with IL-4α, it binds with much higher affinity, inducing the effector functions of IL-13. IL-13Rα2 binds IL-13 with high affinity but has a short cytoplasmic tail and has been shown to act as a nonsignaling decoy receptor. Transfection of fibroblasts and epithelial cells with IL-13Rα2 inhibited the IL-13 induction of soluble collagen, TGF-ß, and CCL17. Adenoviral overexpression of IL-13Rα2 in the lung reduced bleomycin-induced fibrosis. Our work shows that overexpression of IL-13Rα2 inhibits the IL-13 induction of fibrotic markers in vitro and inhibits bleomycin-induced pulmonary fibrosis. In summary our study highlights the antifibrotic nature of IL-13Ra2.


Subject(s)
Interleukin-13 Receptor alpha2 Subunit/physiology , Pulmonary Fibrosis/metabolism , Animals , Bleomycin , Chemokine CCL17/biosynthesis , Collagen/biosynthesis , HEK293 Cells , Humans , Interleukin-13/physiology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta/biosynthesis
18.
J Dermatol Sci ; 76(2): 90-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25219597

ABSTRACT

BACKGROUND: Serum levels of thymus and activation-regulated chemokine (TARC/CCL17) have served as a reliable biomarker of disease progression of atopic dermatitis (AD). However, it remains to be scientifically explained why serum TARC levels correlate well with the degree of AD progression. OBJECTIVE: We hypothesized that dermal cells, but not epidermal keratinocytes, are major cellular sources of TARC and thus responsible for subclinical skin inflammation. This study aimed to identify the skin cells that can produce TARC protein. METHODS: Primary normal human epidermal keratinocytes (NHEK), dermal microvascular endothelial cells (HMVEC-dBl) and dermal fibroblasts (NHDF) were stimulated with TNF-α and IL-4, alone and in combination. TARC mRNA and protein levels were quantified by qPCR and ELISA, respectively. We also investigated the effects of such immunosuppressants as a corticosteroid (dexamethasone) and tacrolimus (FK506) on TARC production, and used various signaling inhibitors to evaluate the signaling pathways involved in TARC expression. RESULTS: Although neither TNF-α nor IL-4 alone induced TARC production by any of the tested cell types, together they induced expression of TARC mRNA and appreciable amounts of TARC protein by HMVEC-dBl and NHDF, but not by NHEK. TARC production by those dermal cells was not inhibited by dexamethasone or FK506. TARC production by HMVEC-dBl was completely inhibited by NF-κB and p38 MAPK inhibitors, but not by an ERK inhibitor. CONCLUSION: Dermal cells, but not epidermal keratinocytes, may be important cellular sources of TARC in AD skin. Therefore, even if epidermal eczematous lesions seem to be improved, complete inhibition of inflammation in the dermis is thought to be particularly important for suppressing both the TARC blood level and progression of AD. However, immunosuppressants did not directly inhibit TARC production by the dermal cells. Anti-inflammatory therapy may decrease TARC blood levels in AD patients indirectly, via its inhibitory effects on TNF-α- and/or IL-4-producing cells in the dermis.


Subject(s)
Chemokine CCL17/biosynthesis , Keratinocytes/metabolism , Skin/metabolism , Cells, Cultured , Chemokine CCL17/blood , Dermatitis, Atopic/metabolism , Humans , Immunosuppressive Agents/pharmacology , Interleukin-4/pharmacology , NF-kappa B/physiology , Skin/cytology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology
19.
PLoS One ; 9(7): e102613, 2014.
Article in English | MEDLINE | ID: mdl-25058417

ABSTRACT

Mycoplasma pneumoniae causes a range of airway and extrapulmonary pathologies in humans. Clinically, M. pneumoniae is associated with acute exacerbations of human asthma and a worsening of experimentally induced asthma in mice. Recently, we demonstrated that Community Acquired Respiratory Distress Syndrome (CARDS) toxin, an ADP-ribosylating and vacuolating toxin synthesized by M. pneumoniae, is sufficient to induce an asthma-like disease in BALB/cJ mice. To test the potential of CARDS toxin to exacerbate preexisting asthma, we examined inflammatory responses to recombinant CARDS toxin in an ovalbumin (OVA) murine model of asthma. Differences in pulmonary inflammatory responses between treatment groups were analyzed by histology, cell differentials and changes in cytokine and chemokine concentrations. Additionally, assessments of airway hyperreactivity were evaluated through direct pulmonary function measurements. Analysis of histology revealed exaggerated cellular inflammation with a strong eosinophilic component in the CARDS toxin-treated group. Heightened T-helper type-2 inflammatory responses were evidenced by increased expression of IL-4, IL-13, CCL17 and CCL22 corresponding with increased airway hyperreactivity in the CARDS toxin-treated mice. These data demonstrate that CARDS toxin can be a causal factor in the worsening of experimental allergic asthma, highlighting the potential importance of CARDS toxin in the etiology and exacerbation of human asthma.


Subject(s)
Asthma/pathology , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Bronchial Hyperreactivity/pathology , Respiratory System/drug effects , Animals , Asthma/chemically induced , Asthma/immunology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CCL17/biosynthesis , Chemokine CCL17/immunology , Chemokine CCL22/biosynthesis , Chemokine CCL22/immunology , Eosinophils/immunology , Eosinophils/pathology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-13/biosynthesis , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Recombinant Proteins/toxicity , Respiratory System/immunology , Respiratory System/pathology , Th2 Cells/immunology , Th2 Cells/pathology
20.
Biochem Biophys Res Commun ; 447(2): 278-84, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24704449

ABSTRACT

Chemokines are important mediators of cell migration, and thymus and activation-regulated chemokine (TARC/CCL17) and macrophage-derived chemokine (MDC/CCL22) are well-known typical inflammatory chemokines involved in atopic dermatitis (AD). (+)-Nootkatone is the major component of Cyperus rotundus. (+)-Nootkatone has antiallergic, anti-inflammatory, and antiplatelet activities. The purpose of this study was to investigate the effect of (+)-nootkatone on tumor necrosis factor α (TNF-α)/interferon γ (IFN-γ)-induced expression of Th2 chemokines in HaCaT cells. We found that (+)-nootkatone inhibited the TNF-α/IFN-γ-induced expression of TARC/CCL17 and MDC/CCL22 mRNA in HaCaT cells. It also significantly inhibited TNF-α/IFN-γ-induced activation of nuclear factor kappa B (NF-κB), p38 mitogen-activated protein kinase (MAPK), and protein kinase Cζ (PKCζ). Furthermore, we showed that PKCζ and p38 MAPK contributed to the inhibition of TNF-α/IFN-γ-induced TARC/CCL17 and MDC/CCL22 expression by blocking IκBα degradation in HaCaT cells. Taken together, these results suggest that (+)-nootkatone may suppress TNF-α/IFN-γ-induced TARC/CCL17 and MDC/CCL22 expression in HaCaT cells by inhibiting of PKCζ and p38 MAPK signaling pathways that lead to activation of NF-κB. We propose that (+)-nootkatone may be a useful therapeutic candidate for inflammatory skin diseases such as AD.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokine CCL17/antagonists & inhibitors , Chemokine CCL22/antagonists & inhibitors , Sesquiterpenes/pharmacology , Th2 Cells/drug effects , Cell Line , Chemokine CCL17/biosynthesis , Chemokine CCL17/genetics , Chemokine CCL22/biosynthesis , Chemokine CCL22/genetics , Dermatitis, Atopic/immunology , Humans , Interferon-gamma/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Polycyclic Sesquiterpenes , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...