Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046040

ABSTRACT

Inflammatory pain, such as hypersensitivity resulting from surgical tissue injury, occurs as a result of interactions between the immune and nervous systems with the orchestrated recruitment and activation of tissue-resident and circulating immune cells to the site of injury. Our previous studies identified a central role for Ly6Clow myeloid cells in the pathogenesis of postoperative pain. We now show that the chemokines CCL17 and CCL22, with their cognate receptor CCR4, are key mediators of this response. Both chemokines are up-regulated early after tissue injury by skin-resident dendritic and Langerhans cells to act on peripheral sensory neurons that express CCR4. CCL22, and to a lesser extent CCL17, elicit acute mechanical and thermal hypersensitivity when administered subcutaneously; this response abrogated by pharmacological blockade or genetic silencing of CCR4. Electrophysiological assessment of dissociated sensory neurons from naïve and postoperative mice showed that CCL22 was able to directly activate neurons and enhance their excitability after injury. These responses were blocked using C 021 and small interfering RNA (siRNA)-targeting CCR4. Finally, our data show that acute postoperative pain is significantly reduced in mice lacking CCR4, wild-type animals treated with CCR4 antagonist/siRNA, as well as transgenic mice depleted of dendritic cells. Together, these results suggest an essential role for the peripheral CCL17/22:CCR4 axis in the genesis of inflammatory pain via direct communication between skin-resident dendritic cells and sensory neurons, opening therapeutic avenues for its control.


Subject(s)
Langerhans Cells/metabolism , Pain, Postoperative/etiology , Pain, Postoperative/metabolism , Receptors, CCR4/metabolism , Sensory Receptor Cells/metabolism , Action Potentials , Animals , Biomarkers , Chemokine CCL17/genetics , Chemokine CCL17/metabolism , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Langerhans Cells/immunology , Mice , Pain, Postoperative/diagnosis , Signal Transduction
2.
Cytokine ; 182: 156721, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39106576

ABSTRACT

AIMS: Acute lymphoblastic leukemia (ALL) is the most common type of pediatrics cancer. Chemokines exert different roles in leukemia process through leukocyte recruitment and regulation of disease severity. Due to the prominent roles of chemokine/receptor axes, this study aimed to measure the blood expression levels of CCR4 and their ligands in pediatrics with B-cell ALL (B-ALL). We also evaluated the impact of cytotoxic chemotherapy on this axis. MATERIAL AND METHOD: Thirty children suffering from B-ALL were included in the study and followed up for 30 days after completion of a chemotherapy course. The blood sampling was performed before and after chemotherapy. 30 healthy donors have also entered the study as control subjects. The mRNA expression of CCL17, CCL22 and CCR4 genes was determined by quantitative real-time PCR. The frequency of the peripheral blood mononuclear cells expressing CCR4 (CCR4 + PBMCs) was also evaluated by the flow cytometry method. Moreover, we evaluated the association of the CCL17/CCL22-CCR4 axis with some diagnostic, prognostic and predictive biomarkers in ALL patients. RESULTS: There was overexpression of the CCL17/CCL22-CCR4 axis along with lactate dehydrogenase (LDH) in pediatrics with B-ALL compared to healthy controls. After induction of chemotherapy, the blood expression levels of the CCL17/CCL22-CCR4 axis have reached the levels of healthy controls. The findings for the blood expression levels of CCR4 were also confirmed using flow cytometry. CONCLUSION: The CCL17/CCL22-CCR4 axis can be used as a novel predictive and prognostic biomarker in B-ALL.


Subject(s)
Chemokine CCL17 , Chemokine CCL22 , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, CCR4 , Humans , Receptors, CCR4/metabolism , Receptors, CCR4/genetics , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Child , Male , Chemokine CCL17/genetics , Chemokine CCL17/blood , Chemokine CCL17/metabolism , Female , Child, Preschool , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/blood , Leukocytes, Mononuclear/metabolism , Adolescent , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis
3.
Cytokine ; 181: 156669, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38875750

ABSTRACT

OBJECTIVES: Alveolar echinococcosis (AE) represents one of the deadliest helminthic infections, characterized by an insidious onset and high lethality. METHODS: This study utilized the Gene Expression Omnibus (GEO) database, applied Weighted Correlation Network Analysis (WGCNA) and Differential Expression Analysis (DEA), and employed the Matthews Correlation Coefficient (MCC) to identify CCL17 and CCL19 as key genes in AE. Immunohistochemistry and immunofluorescence co-localization techniques were used to examine the expression of CCL17 and CCL19 in liver tissue lesions of AE patients. Additionally, a mouse model of multilocular echinococcus larvae infection was developed to study the temporal expression patterns of these genes, along with liver fibrosis and inflammatory responses. RESULTS: The in vitro model simulating echinococcal larva infection mirrored the hepatic microenvironment post-infection with multilocular echinococcal tapeworms. Quantitative RT-PCR analysis showed that liver fibrosis occurred in AE patients, with proximal activation and increased expression of CCL17 and CCL19 over time post-infection. Notably, expression peaked during the late stages of infection. Similarly, F4/80, a macrophage marker, exhibited corresponding trends in expression. Upon stimulation of normal hepatocytes by vesicular larvae in cellular experiments, there was a significant increase in CCL17 and CCL19 expression at 12 h post-infection, mirroring the upregulation observed with F4/80. CONCLUSION: CCL17 and CCL19 facilitate macrophage aggregation via the chemokine pathway and their increased expression correlates with the progression of infection, suggesting their potential as biomarkers for AE progression.


Subject(s)
Biomarkers , Chemokine CCL17 , Chemokine CCL19 , Disease Progression , Animals , Humans , Mice , Biomarkers/metabolism , Chemokine CCL19/metabolism , Chemokine CCL17/metabolism , Chemokine CCL17/genetics , Echinococcosis/metabolism , Liver Cirrhosis/parasitology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Disease Models, Animal , Liver/parasitology , Liver/metabolism , Liver/pathology , Echinococcosis, Hepatic/metabolism , Echinococcosis, Hepatic/parasitology , Female , Male , Hepatocytes/metabolism , Hepatocytes/parasitology
4.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Article in English | MEDLINE | ID: mdl-34934004

ABSTRACT

Signal tranducer and activator of transcription 5 (STAT5) plays a critical role in mediating cellular responses following cytokine stimulation. STAT proteins critically signal via the formation of dimers, but additionally, STAT tetramers serve key biological roles, and we previously reported their importance in T and natural killer (NK) cell biology. However, the role of STAT5 tetramerization in autoimmune-mediated neuroinflammation has not been investigated. Using the STAT5 tetramer-deficient Stat5a-Stat5b N-domain double knockin (DKI) mouse strain, we report here that STAT5 tetramers promote the pathogenesis of experimental autoimmune encephalomyelitis (EAE). The mild EAE phenotype observed in DKI mice correlates with the impaired extravasation of pathogenic T-helper 17 (Th17) cells and interactions between Th17 cells and monocyte-derived cells (MDCs) in the meninges. We further demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF)-mediated STAT5 tetramerization regulates the production of CCL17 by MDCs. Importantly, CCL17 can partially restore the pathogenicity of DKI Th17 cells, and this is dependent on the activity of the integrin VLA-4. Thus, our study reveals a GM-CSF-STAT5 tetramer-CCL17 pathway in MDCs that promotes autoimmune neuroinflammation.


Subject(s)
Autoimmune Diseases/metabolism , Neuroinflammatory Diseases/metabolism , STAT5 Transcription Factor , Animals , Chemokine CCL17/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Protein Multimerization , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , Th17 Cells/metabolism
5.
Circulation ; 145(10): 765-782, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35113652

ABSTRACT

BACKGROUND: Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS: We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS: We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (ß-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS: These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.


Subject(s)
Heart Failure , Myocardial Infarction , Angiotensin II/pharmacology , Animals , Chemokine CCL17/metabolism , Chemokine CCL17/pharmacology , Diphtheria Toxin/metabolism , Diphtheria Toxin/pharmacology , Heart Failure/genetics , Heart Failure/metabolism , Humans , Inflammation/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylephrine/metabolism , Phenylephrine/pharmacology , T-Lymphocytes, Regulatory/metabolism , Ventricular Remodeling
6.
Immunol Cell Biol ; 101(7): 600-609, 2023 08.
Article in English | MEDLINE | ID: mdl-36975092

ABSTRACT

Chemokine (C-C) ligand 17 (CCL17) was first identified as thymus- and activation-regulated chemokine when it was found to be constitutively expressed in the thymus and identified as a T-cell chemokine. This chemoattractant molecule has subsequently been found at elevated levels in a range of autoimmune and inflammatory diseases, as well as in cancer. CCL17 is a C-C chemokine receptor type 4 (CCR4) ligand, with chemokine (C-C) ligand 22 being the other major ligand and, as CCR4 is highly expressed on helper T cells, CCL17 can play a role in T-cell-driven diseases, usually considered to be via its chemotactic activity on T helper 2 cells; however, given that CCR4 is also expressed by other cell types and there is elevated expression of CCL17 in many diseases, a broader CCL17 biology is suggested. In this review, we summarize the biology of CCL17, its regulation and its potential contribution to the pathogenesis of various preclinical models. Reference is made, for example, to recent literature indicating a role for CCL17 in the control of pain as part of a granulocyte macrophage-colony-stimulating factor/CCL17 pathway in lymphocyte-independent models and thus not as a T-cell chemokine. The review also discusses the potential for CCL17 to be a biomarker and a therapeutic target in human disorders.


Subject(s)
Autoimmunity , Receptors, Chemokine , Humans , Ligands , Receptors, Chemokine/metabolism , Chemokine CCL17/metabolism , Chemokines , Inflammation
7.
Molecules ; 28(8)2023 Apr 16.
Article in English | MEDLINE | ID: mdl-37110740

ABSTRACT

The main pathogenic factor in atopic dermatitis (AD) is Th2 inflammation, and levels of serum CCL17 and CCL22 are related to severity in AD patients. Fulvic acid (FA) is a kind of natural humic acid with anti-inflammatory, antibacterial, and immunomodulatory effects. Our experiments demonstrated the therapeutic effect of FA on AD mice and revealed some potential mechanisms. FA was shown to reduce TARC/CCL17 and MDC/CCL22 expression in HaCaT cells stimulated by TNF-α and IFN-γ. The inhibitors showed that FA inhibits CCL17 and CCL22 production by deactivating the p38 MAPK and JNK pathways. After 2,4-dinitrochlorobenzene (DNCB) induction in mice with atopic dermatitis, FA effectively reduced the symptoms and serum levels of CCL17 and CCL22. In conclusion, topical FA attenuated AD via downregulation of CCL17 and CCL22, via inhibition of P38 MAPK and JNK phosphorylation, and FA is a potential therapeutic agent for AD.


Subject(s)
Dermatitis, Atopic , Animals , Mice , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Keratinocytes , NF-kappa B/metabolism , Chemokine CCL22/metabolism , Chemokine CCL22/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Dinitrochlorobenzene/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chemokine CCL17/metabolism , Chemokine CCL17/pharmacology , Chemokine CCL17/therapeutic use
8.
Anticancer Drugs ; 33(2): 149-157, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34657098

ABSTRACT

Recently, cytokine-induced killer (CIK) cells have been shown to possess effective cytotoxic activity against some tumor cells both in vitro and in clinical research. Furthermore, dendritic cell-activated CIK (DC-CIK) cells display significantly increased antitumor activity compared to unstimulated CIK cells. Study findings indicate DC cells can secrete chemokine C-C motif ligand 17 (CCL17) and chemokine C-C motif ligand 22 (CCL22) with a common receptor molecule, C-C chemokine receptor type-4(CCR4). CCL17 and CCL22 levels were measured by ELISA from CIK cell culture supernatants and the expression of CCR4 on CIK and DC-CIK cells was analyzed by flow cytometry. Through Migration and Killing assays, further analyzed the effects of the altered expression levels of CCR4 on the chemotactic ability and the tumor-killing efficiency of CIK cells. We found markedly increased CCL17 and CCL22 in supernatants of DC-CIK co-cultures. Similarly, the expression of CCR4 was also increased on CIK cells in these co-cultures. Further, the stimulation of CCL17 and CCL22 increased expression of the CCR4 and enhanced the migratory capacity and antitumor efficacy of CIK cells. Simultaneously, similar effects had achieved by transfecting the CCR4 gene into CIK cells. DC cells may promote the expression of CCR4 on CIK cells by secreting CCL17 and CCL22, thereby promoting infiltration of DC-CIK cells into the tumor microenvironment, and exerting stronger antitumor activity than CIK cells.


Subject(s)
Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , Cytokine-Induced Killer Cells/metabolism , Receptors, CCR4/biosynthesis , Cell Movement/physiology , Dendritic Cells , Humans , Ligands
9.
J Immunol ; 205(1): 213-222, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32461237

ABSTRACT

It has been reported that a GM-CSF→CCL17 pathway, originally identified in vitro in macrophage lineage populations, is implicated in the control of inflammatory pain, as well as arthritic pain and disease. We explore, in this study and in various inflammation models, the cellular CCL17 expression and its GM-CSF dependence as well as the function of CCL17 in inflammation and pain. This study used models allowing the convenient cell isolation from Ccl17E/+ reporter mice; it also exploited both CCL17-dependent and unique CCL17-driven inflammatory pain and arthritis models, the latter permitting a radiation chimera approach to help identify the CCL17 responding cell type(s) and the mediators downstream of CCL17 in the control of inflammation and pain. We present evidence that 1) in the particular inflammation models studied, CCL17 expression is predominantly in macrophage lineage populations and is GM-CSF dependent, 2) for its action in arthritic pain and disease development, CCL17 acts on CCR4+ non-bone marrow-derived cells, and 3) for inflammatory pain development in which a GM-CSF→CCL17 pathway appears critical, nerve growth factor, CGRP, and substance P all appear to be required.


Subject(s)
Arthritis, Experimental/immunology , Chemokine CCL17/metabolism , Pain/immunology , Peritonitis/immunology , Pneumonia/immunology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/pathology , Calcitonin Gene-Related Peptide/metabolism , Chemokine CCL17/genetics , Genes, Reporter/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Mice, Transgenic , Nerve Growth Factor/metabolism , Pain/diagnosis , Pain/pathology , Pain Measurement , Peritonitis/complications , Peritonitis/pathology , Pneumonia/complications , Pneumonia/pathology , Signal Transduction/immunology , Substance P/metabolism
10.
Cell Mol Biol (Noisy-le-grand) ; 68(2): 54-59, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35869715

ABSTRACT

This study was developed to investigate the expression of TOLL2, TARC and MDC in placenta tissue of pregnant patients infected with syphilis and their clinical significance. For this aim, placenta samples were collected from five pregnant patients co-infected with syphilis and five undergoing full-term delivery before RT-PCR was performed to detect the mRNA expression of TLR2, TARC and MDC genes. The protein expression of TLR2, TARC and MDC genes was examined by Western Blotting. Results showed that TLR2, TARC and MDC were expressed in placental syncytiotrophoblast cells of patients with pregnancy-associated syphilis infection. TLR2 level was found significantly higher in placenta tissue of patients with pregnancy-associated syphilis infection compared with normal placenta tissue (P<0.05), so were TARC (P<0.05) and MDC genes (P<0.05). It is concluded that TOLL2, TARC and MDC levels significantly increased in the placenta tissue of pregnant patients infected with syphilis, suggesting that the three genes were involved in the molecular pathology of the patients.


Subject(s)
Chemokine CCL17 , Syphilis , Chemokine CCL17/metabolism , Chemokine CCL22 , Female , Humans , Placenta/metabolism , Pregnancy , Toll-Like Receptor 2/genetics
11.
Mar Drugs ; 20(9)2022 Aug 31.
Article in English | MEDLINE | ID: mdl-36135752

ABSTRACT

Polysiphonia morrowii is a well-known red alga that has promising pharmacological characteristics. The current study evaluates the protective effect of 3-bromo-4,5-dihydroxybenzaldehyde (BDB) isolated from P. morrowii on tumor necrosis factor (TNF)-α/interferon (IFN)-γ-stimulated inflammation and skin barrier deterioration in HaCaT keratinocytes. The anti-inflammatory effect of BDB in TNF-α/IFN-γ-stimulated HaCaT keratinocytes is evaluated by investigating nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, inflammatory cytokines, and chemokines. Further, the interaction between BDB and the skin barrier functions in stimulated HaCaT keratinocytes is investigated. The findings of the study reveal that BDB dose-dependently increases cell viability while decreasing intracellular reactive oxygen species (ROS) production. BDB downregulates the expression of inflammatory cytokines, interleukin (IL)-6, -8, -13, IFN-γ, TNF-α, and chemokines, Eotaxin, macrophage-derived chemokine (MDC), regulated on activation, normal T cells expressed and secreted (RANTES), and thymus and activation-regulated chemokine (TARC) by modulating the MAPK and NF-κB signaling pathways in TNF-α/IFN-γ-stimulated HaCaT keratinocytes. Furthermore, BDB increases the production of skin hydration proteins and tight junction proteins in stimulated HaCaT keratinocytes by preserving skin moisturization and tight junction stability. These findings imply that BDB exhibits a protective ability against inflammation and deterioration of skin barrier via suppressing the expression of inflammatory signaling in TNF-α/IFN-γ-stimulated HaCaT keratinocytes.


Subject(s)
Benzaldehydes , Keratinocytes , Rhodophyta , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Benzaldehydes/pharmacology , Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , Chemokine CCL5/metabolism , Chemokines/metabolism , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukins/metabolism , Keratinocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Rhodophyta/chemistry , STAT1 Transcription Factor/metabolism , Tight Junction Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Proc Natl Acad Sci U S A ; 116(51): 25816-25827, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31796596

ABSTRACT

Mounting an effective immune response relies critically on the coordinated interactions between adaptive and innate compartments. How and where immune cells from these different compartments interact is still poorly understood. Here, we demonstrate that the cross-talk between invariant natural killer T cells (iNKT) and CD8+ T cells in the spleen, essential for initiating productive immune responses, is biphasic and occurs at 2 distinct sites. Codelivery of antigen and adjuvant to antigen-presenting cells results in: 1) initial short-lived interactions (0 to 6 h), between CD8+ T cells, dendritic cells (DCs), and iNKT cells recruited outside the white pulp; 2) followed by long-lasting contacts (12 to 24 h) between iNKT cells, DCs, and CD8+ T cells occurring in a 3-way interaction profile within the white pulp. Both CXCR3 and CCR4 are essential to orchestrate this highly dynamic process and play nonredundant in T cell memory generation. While CXCR3 promotes memory T cells, CCR4 supports short-lived effector cell generation. We believe our work provides insights into the initiation of T cell responses in the spleen and their consequences for T cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CCL17/metabolism , Interleukin-4/metabolism , Natural Killer T-Cells/immunology , Spleen/immunology , Animals , Cell Differentiation , Chemokine CXCL9/metabolism , Dendritic Cells/immunology , Homeodomain Proteins/genetics , Immunologic Memory/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/immunology , Receptor Cross-Talk/physiology , Receptors, CCR4/metabolism , Receptors, CXCR3/metabolism , Spleen/cytology
13.
J Allergy Clin Immunol ; 147(4): 1174-1190.e1, 2021 04.
Article in English | MEDLINE | ID: mdl-33516871

ABSTRACT

Atopic dermatitis (AD) is a common yet complex skin disease, posing a therapeutic challenge with increasingly recognized different phenotypes among variable patient populations. Because therapeutic response may vary on the basis of heterogeneous clinical and molecular phenotypes, a shift toward precision medicine approaches may improve AD management. Herein, we will consider biomarkers as potential instruments in the toolbox of precision medicine in AD and will review the process of biomarker development and validation, the opinion of AD experts on the use of biomarkers, types of biomarkers, encompassing biomarkers that may improve AD diagnosis, biomarkers reflecting disease severity, and those potentially predicting AD development, concomitant atopic diseases, or therapeutic response, and current practice of biomarkers in AD. We found that chemokine C-C motif ligand 17/thymus and activation-regulated chemokine, a chemoattractant of TH2 cells, has currently the greatest evidence for robust correlation with AD clinical severity, at both baseline and during therapy, by using the recommendations, assessment, development, and evaluation approach. Although the potential of biomarkers in AD is yet to be fully elucidated, due to the complexity of the disease, a comprehensive approach taking into account both clinical and reliable, AD-specific biomarker evaluations would further facilitate AD research and improve patient management.


Subject(s)
Biomarkers/metabolism , Chemokine CCL17/metabolism , Dermatitis, Atopic/diagnosis , Eosinophils/immunology , Th2 Cells/immunology , Animals , Humans , Immunoglobulin E/metabolism , International Cooperation , Precision Medicine
14.
Hell J Nucl Med ; 25(2): 125-131, 2022.
Article in English | MEDLINE | ID: mdl-35913858

ABSTRACT

OBJECTIVE: Although the majority of patients with Hodgkin lymphoma (HL) has recently become long-term survivors, 20%-30% of HL patients have primary refractory disease or relapse. It is essential to identify patients at risk of treatment failure during first-line therapy. To objective of the present study was to investigate the combined prognostic role of fluorine-18-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) imaging and thymus and activation-regulated chemokine (TARC) levels in Hodgkin lymphoma. SUBJECTS AND METHODS: Between 01/01/2013 and 01/03/2019 77 HL patients were enrolled in this study where serum TARC levels were measured by an immunoassay and 18F-FDG PET/CT scans were performed at baseline, after the second cycle of ABVD treatment (interim) and at the end of first-line therapy. RESULTS: Twenty-six patients (34%) had early-stage HL, while 51 patients presented with advanced-stage disease. Fifteen patients had primary refractory HL, while 1 patient relapsed after first-line therapy. Optimal TARC cut-off value for progression-free survival (PFS) was 700pg/mL based on receiver operating characteristic (ROC) curve analysis. With Cox regression analysis, 18F-FDG PET/CT with Deauville scores of 3, 4, or 5 and TARC levels above 700pg/mL predicted treatment failure at interim assessment. Inclusion of HL patients with a Deauville score of 3 to the high-risk population resulted in a 7-fold increase in the estimated risk of relapse compared to patients with Deauville score 4-5 with TARC levels above 700pg/mL. Patients with interim 18F-FDG PET/CT Deauville scores 3-5 had a significant survival benefit if their TARC levels were 700pg/mL. Positive predictive value (PPV) of interim 18F-FDG PET/CT scans with a Deauville score 3-5 was 47.8%, while combined PPV of a similar 18F-FDGPET/CT assessment and elevated TARC levels was 88.8%. CONCLUSION: Interim 18F-FDG PET/CT and TARC analyzed together accurately identify HL patients who do not respond sufficiently to treatment and who need an early change of therapy.


Subject(s)
Chemokine CCL17/metabolism , Fluorodeoxyglucose F18 , Hodgkin Disease , Antineoplastic Combined Chemotherapy Protocols , Bleomycin , Dacarbazine , Doxorubicin , Humans , Neoplasm Recurrence, Local , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography , Prognosis , Vinblastine
15.
Am J Respir Cell Mol Biol ; 64(3): 344-356, 2021 03.
Article in English | MEDLINE | ID: mdl-33264064

ABSTRACT

The interplay of type-2 inflammation and antiviral immunity underpins asthma exacerbation pathogenesis. Virus infection induces type-2 inflammation-promoting chemokines CCL17 and CCL22 in asthma; however, mechanisms regulating induction are poorly understood. By using a human rhinovirus (RV) challenge model in human airway epithelial cells in vitro and mice in vivo, we assessed mechanisms regulating CCL17 and CCL22 expression. Subjects with mild to moderate asthma and healthy volunteers were experimentally infected with RV and airway CCL17 and CCL22 protein quantified. In vitro airway epithelial cell- and mouse-RV infection models were then used to define STAT6- and NF-κB-mediated regulation of CCL17 and CCL22 expression. Following RV infection, CCL17 and CCL22 expression was higher in asthma, which differentially correlated with clinical and immunological parameters. Air-liquid interface-differentiated primary epithelial cells from donors with asthma also expressed higher levels of RV-induced CCL22. RV infection boosted type-2 cytokine-induced STAT6 activation. In epithelial cells, type-2 cytokines and STAT6 activation had differential effects on chemokine expression, increasing CCL17 and suppressing CCL22, whereas NF-κB promoted expression of both chemokines. In mice, RV infection activated pulmonary STAT6, which was required for CCL17 but not CCL22 expression. STAT6-knockout mice infected with RV expressed increased levels of NF-κB-regulated chemokines, which was associated with rapid viral clearance. Therefore, RV-induced upregulation of CCL17 and CCL22 was mediated by NF-κB activation, whereas expression was differentially regulated by STAT6. Together, these findings suggest that therapeutic targeting of type-2 STAT6 activation alone will not block all inflammatory pathways during RV infection in asthma.


Subject(s)
Asthma/pathology , Asthma/virology , Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , Disease Progression , Rhinovirus/physiology , STAT6 Transcription Factor/metabolism , A549 Cells , Adolescent , Adult , Animals , Biomarkers/metabolism , Chemokines/metabolism , Epithelial Cells/metabolism , Female , Humans , Kinetics , Lung/pathology , Lung/virology , Male , Mice, Inbred BALB C , Middle Aged , NF-kappa B/metabolism , Tissue Donors , Young Adult
16.
Cancer Sci ; 112(11): 4604-4616, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34449935

ABSTRACT

An inhibitor of the histone methyltransferase enhancer of zeste homologue 2 (EZH2), tazemetostat, has been developed for the treatment of B-cell lymphoma, but its mechanisms of action are not fully elucidated. We screened for genes targeted by tazemetostat in eleven B-cell lymphoma cell lines and found that tazemetostat significantly increased the expression of chemokine (C-C motif) ligand 17 (CCL17)/thymus- and activation-regulated chemokine (TARC) in all, which codes for a chemokine that is a hallmark of Hodgkin/Reed-Sternberg (H/RS) cells in Hodgkin lymphoma. Notably, gene set enrichment analysis demonstrated a positive correlation between the genes upregulated by tazemetostat in five follicular lymphoma (FL) cell lines and those reported to be overexpressed in H/RS cells. The CCL17 promoter region was enriched in repressive histone modification H3K27me3, and tazemetostat induced H3K27 demethylation and activated gene transcription. CCL17 protein secretion was also induced by EZH2 inhibition, which was further enhanced by concurrent CpG stimulation. In vitro transwell migration assay demonstrated that CCL17 produced by tazemetostat-treated B cells enhanced the recruitment of T cells, which had the potential to exert antilymphoma response. Analysis of publicly available human lymphoma databases showed that CCL17 gene expression was inversely correlated with the EZH2 activation signature and significantly paralleled the CD4+ and CD8+ T-cell-rich signature in FL and germinal center B-cell-like diffuse large B-cell lymphoma. Our findings indicate that tazemetostat can potentially activate antilymphoma response by upregulating CCL17 expression in B-cell lymphoma cells and promote T-cell recruitment, which provides a rationale for its combination with immunotherapy.


Subject(s)
Benzamides/pharmacology , Biphenyl Compounds/pharmacology , Chemokine CCL17/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating , Lymphoma, Large B-Cell, Diffuse/metabolism , Morpholines/pharmacology , Pyridones/pharmacology , Cell Line, Tumor , Cell Movement , Chemokine CCL17/genetics , Databases, Factual , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Promoter Regions, Genetic , Reed-Sternberg Cells , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Up-Regulation
17.
J Neuroinflammation ; 18(1): 62, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33648537

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. METHODS: Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. RESULTS: Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1ß, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. CONCLUSIONS: rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH.


Subject(s)
Cerebral Hemorrhage/pathology , Chemokine CCL17/metabolism , Neurons/pathology , Receptors, CCR4/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Brain/metabolism , Brain/pathology , Cerebral Hemorrhage/metabolism , Forkhead Box Protein O1/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins
18.
Rheumatology (Oxford) ; 60(4): 1724-1733, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33067627

ABSTRACT

OBJECTIVES: To characterize the expression profiles of two nuclear-encoded mitochondrial genes previously associated with chronic pain, the translocator protein (TSPO) and family with sequence similarity 173B (FAM173B), in different knee compartments from patients with painful knee OA. Also, to examine their association with the joint expression of inflammatory cytokines/chemokines and clinical symptoms. METHODS: The study was performed on 40 knee OA patients and 19 postmortem (PM) controls from which we collected the knee tissues: articular cartilage (AC), synovial membrane (SM) and subchondral bone (SB). Quantitative real-time polymerase chain reaction was used to determine the relative mRNA levels of TSPO, FAM173B, and inflammatory mediators IL6, IL8, IL10, IL12, MCP1, CCL11 and CCL17. OA patients rated their pain intensity (visual analogue scale), severity of knee-related outcomes (KOOS) and pain sensitivity assessed by pressure algometry. RESULTS: The gene expression of TSPO in SM was elevated in OA patients compared with control subjects while there were no group differences in AC and SB. Expression of FAM173B was reduced in SM but elevated in SB in OA patients compared with controls. The expression of TSPO and FAM173B in SM and SB was associated with the expression of inflammatory substances, but not in AC. Synovial expression of TSPO correlated with lower pain intensity and FAM173B with increased pressure pain sensitivity in OA. CONCLUSION: Our results suggest that altered expression of TSPO and FAM173B is associated with joint expression of inflammatory mediators and with clinical symptoms indicating the relevance for the pathophysiology of knee OA.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Osteoarthritis, Knee/genetics , RNA, Messenger/metabolism , Receptors, GABA/genetics , Adult , Aged , Arthralgia/etiology , Cartilage, Articular/metabolism , Case-Control Studies , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Chemokine CCL17/genetics , Chemokine CCL17/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Female , Gene Expression , Histone-Lysine N-Methyltransferase/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , Knee Joint/metabolism , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Real-Time Polymerase Chain Reaction , Receptors, GABA/metabolism , Synovial Membrane/metabolism , Visual Analog Scale
19.
J Pathol ; 250(1): 55-66, 2020 01.
Article in English | MEDLINE | ID: mdl-31579932

ABSTRACT

Peritoneal fibrosis remains a problem in kidney failure patients treated with peritoneal dialysis. Severe peritoneal fibrosis with encapsulation or encapsulating peritoneal sclerosis is devastating and life-threatening. Although submesothelial fibroblasts as the major precursor of scar-producing myofibroblasts in animal models and M2 macrophage (Mϕ)-derived chemokines in peritoneal effluents of patients before diagnosis of encapsulating peritoneal sclerosis have been identified, attenuation of peritoneal fibrosis is an unmet medical need partly because the mechanism for cross talk between Mϕs and fibroblasts remains unclear. We use a sodium hypochlorite-induced mouse model akin to clinical encapsulated peritoneal sclerosis to study how the peritoneal Mϕs activate fibroblasts and fibrosis. Sodium hypochlorite induces the disappearance of CD11bhigh F4/80high resident Mϕs but accumulation of CD11bint F4/80int inflammatory Mϕs (InfMϕs) through recruiting blood monocytes and activating local cell proliferation. InfMϕs switch to express chemokine (C-C motif) ligand 17 (CCL17), CCL22, and arginase-1 from day 2 after hypochlorite injury. More than 75% of InfMϕs undergo genetic recombination by Csf1r-driven Cre recombinase, providing the possibility to reduce myofibroblasts and fibrosis by diphtheria toxin-induced Mϕ ablation from day 2 after injury. Furthermore, administration of antibody against CCL17 can reduce Mϕs, myofibroblasts, fibrosis, and improve peritoneal function after injury. Mechanistically, CCL17 stimulates migration and collagen production of submesothelial fibroblasts in culture. By breeding mice that are induced to express red fluorescent protein in Mϕs and green fluorescence protein (GFP) in Col1a1-expressing cells, we confirmed that Mϕs do not produce collagen in peritoneum before and after injury. However, small numbers of fibrocytes are found in fibrotic peritoneum of chimeric mice with bone marrow from Col1a1-GFP reporter mice, but they do not contribute to myofibroblasts. These data demonstrate that InfMϕs switch to pro-fibrotic phenotype and activate peritoneal fibroblasts through CCL17 after injury. CCL17 blockade in patients with peritoneal fibrosis may provide a novel therapy. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Chemokine CCL17/metabolism , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Macrophage Activation , Macrophages, Peritoneal/metabolism , Paracrine Communication , Peritoneal Fibrosis/metabolism , Peritoneum/metabolism , Animals , Cell Proliferation , Chemokine CCL17/genetics , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Fibroblasts/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Macrophages, Peritoneal/pathology , Mice, Inbred C57BL , Mice, Transgenic , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/genetics , Peritoneal Fibrosis/pathology , Peritoneum/pathology , Phenotype , Promoter Regions, Genetic , Signal Transduction , Sodium Hypochlorite
20.
Allergy Asthma Proc ; 42(1): e30-e39, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33404399

ABSTRACT

Background: Serum thymus and activation-regulated chemokine (TARC) and periostin are reliable biomarkers in eosinophilic asthma. Objective: This study was carried out to determine the use of periostin and TARC as biomarkers in asthma and to compare the superiority of one over the other, especially in asthma with an eosinophilic phenotype. Methods: The study was conducted with 87 patients with asthma and 42 healthy control subjects. Patients with asthma were also divided into eosinophilic and non-eosinophilic phenotypes. A pulmonary function test was performed in all the participants, and serum and induced sputum TARC, periostin concentrations, eosinophils, and total immunoglobulin E values were examined. Results: TARC and periostin levels were significantly higher in the asthma group than in the control group (p < 0.001). The serum TARC level in the eosinophilic group was significantly higher than in the non-eosinophilic and control groups (p < 0.001). The induced sputum TARC level was significantly higher in the non-eosinophilic group than in the control group (p < 0.001). The TARC and periostin levels of the patients were evaluated by using receiver operator characteristic analysis. The cutoff value for TARC was determined to be 1415.39 ng/L; likewise, the cutoff value for periostin was 107.60 ng/L. The present study detected that serum levels of TARC correlated to serum levels of periostin (r = 0.54; p = 0.032). Furthermore, when evaluating correlations between serum and sputum levels, there was a correlation detected between TARC and periostin in serum, whereas this correlation was stronger in sputum: r = 0.66, p = 0.020; and r = 0.62, p = 0.028, respectively. Conclusion: Serum and sputum TARC and periostin may contribute for monitoring the improvement of patients, particularly those with asthma. Furthermore, TARC was a more reliable biomarker than periostin for patients with eosinophilic asthma.


Subject(s)
Asthma/diagnosis , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Chemokine CCL17/metabolism , Eosinophilia/diagnosis , Eosinophils/immunology , Adult , Female , Humans , Immunoglobulin E/blood , Male , Middle Aged , Phenotype , Prospective Studies , Respiratory Function Tests , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL