1
|
Isono T, Kojima A, Nishida T, Kobayashi Y, Ishiguro T, Takaku Y, Kagiyama N, Kurashima K. Serum TARC Level as a Predictive Marker of Severe Disease in COVID-19 during the Omicron Variant Period of the Pandemic. Intern Med 2025; 64:367-374. [PMID: 39566990 PMCID: PMC11867742 DOI: 10.2169/internalmedicine.4276-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024] Open
Abstract
Objective Thymus and activation-regulated chemokine (TARC) can predict severe disease in patients with coronavirus disease 2019 (COVID-19). However, no reports have addressed the predictive value of TARC with the widespread use of vaccines and medications for COVID-19 during the Omicron variant period of the pandemic. Methods This single-center prospective cohort study enrolled COVID-19 patients admitted to our institution between December 1, 2021, and August 15, 2022. Patients with respiratory failure due to diseases other than COVID-19 were also excluded. We measured the serum TARC levels of patients at admission. Results We enrolled 157 patients, with 89 in the severe group and 68 in the non-severe group. The severe group was more likely than the non-severe group to include older patients, those with no or one dose of vaccine, and those with interstitial lung disease. The cutoff level of TARC derived from a receiver operator characteristic curve analysis to predict severe disease was 174.0 pg/mL. The sensitivity, specificity, positive predictive value, and negative predictive value were 72.1%, 69.7%, 64.5%, and 76.5%, respectively. The area under the curve was 0.722 (95% confidence interval: 0.635-0.809). A multivariate analysis showed that 2 vaccination doses were associated with non-severe disease, and TARC ≤174 pg/mL was associated with severe disease. Conclusion TARC was a predictive factor for severe disease, but its cutoff value was higher and its predictive accuracy lower than those in previous reports. We surmised that during the Omicron variant period of the pandemic, the widespread use of vaccines and medications for COVID-19 decreased the predictive accuracy of TARC.
Collapse
Affiliation(s)
- Taisuke Isono
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Ayaka Kojima
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Takashi Nishida
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Yoichi Kobayashi
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Takashi Ishiguro
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Yotaro Takaku
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Naho Kagiyama
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Kazuyoshi Kurashima
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| |
Collapse
|
2
|
Gu X, Chen B, Zhang S, Zhai X, Hu Y, Ye H. The expression of CCL17 and potential prognostic value on tumor immunity in thyroid carcinoma based on bioinformatics analysis. Sci Rep 2024; 14:31580. [PMID: 39738081 PMCID: PMC11686015 DOI: 10.1038/s41598-024-75750-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/08/2024] [Indexed: 01/01/2025] Open
Abstract
Although CCL17 has been reported to exert a vital role in many cancers, the related studies in the thyroid carcinoma have never reported. As a chemokine, CCL17 plays a positive role by promoting the infiltration of immune cells into the tumor microenviroment (TME) to influence tumor invasion and metastasis. Therefore, this study is aimed to investigate the association of CCL17 level with potential prognostic value on tumor immunity in the thyroid carcinoma (THCA) based on the bioinformatics analysis. GEPIA database was applied to analyze CCL17 mRNA expression in THCA data from TCGA database. Through the collection of the data, totally 500 tumor and 57 normal tissue samples were taken for the study. According to survival status and survival time in 500 tumor samples and CCL17 expression from RNA-seq data, all patients were categorized as high- expression (n = 64) and low-expression (n = 436) groups using X-tile program. Next, the association of CCL17 with survival in the thyroid carcinoma patients was examined by using the Kaplan-Meier plotter database. Then, weighted gene co-expression network (WGCNA) was employed to analyze the 1424 DEGs to classify 9 modules. Besides, STRING database was used to obtain the hub genes. GO and KEGG database were employed to explore blue module genes enrichment situations. In addition, TISIDB was used to analyze the relationship of CCL17 expression with tumor-infiltrating lymphocytes proportion, immunostimulators, and major histocompatibility complexes in THCA. The correlation of CCL17 with 22 TIIC subtypes was evaluated by ESTIMATE and CIBERSORT databases. The association of CCL17 level with gene marker of immune cells in THCA was analyzed by GEPIA and TIMER databases. Finally, immunohistochemistry was applied to validate CCL17 expression in 21 tumor and para-carcinoma tissue samples. CCL17 expression in tumors was significantly up-regulated relative to non-carcinoma samples. Patients from CCL17 high-expression group had significantly decreased overall survival compared with low-expression group, which has a significantly importantly potential prognostic value. Moreover, CCL17 and clinical characteristics were analyzed, suggesting that CCL17 expression significantly increased among patients of advanced stage, with advanced T classification, advanced N classification, and higher CCR4 expression. Based on WGCNA, expression of 1424 DEGs in blue module with 258 genes was negatively related to dismal survival and clinical lymph node metastasis in THCA patients. Moreover, CCR4 and CCL17 genes were identified as hub genes within blue module. CCL17 high-expression had greater ImmuneScore, StromalScore and ESTIMATEScore, while lower TumorPurity compared to the CCL17 low-expression. Then, GO and KEGG database were used to analyze blue module genes enrichment situations. The result showed that genes in blue module were associated with cytokine-cytokine receptor interaction, chemokine, and PI3K - Akt pathways. The results of tumor-infiltrating lymphocytes proportion, immunostimulators, and major histocompatibility complexes were significantly positive in CCL17 high-expression. Our findings showed that B cells naïve, T cells CD4 memory resting, T cells CD8, T cells regulatory (Tregs), and dendritic cells resting were the main immune components of THCA tumor microenvironment (TME). CCL17 high-expression in TC was significantly positively related to expression of immune cell gene markers. The result of immunohistochemistry demonstrated that CCL17 expression in tumor tissues significantly increased compared with para-carcinoma tissues. CCL17 high-expression was significantly positively associated with age and advanced N classification, suggesting that CCL17 could accelerate tumor progression by promoting the lymph node metastasis. CCL17 high-expression in THCA tumor microenvironment (TME) accelerates local infiltration of immune cells and enhances anticancer immunity, resulting in worse survival of patients and exerting potential prognostic value on tumor immunity in THCA.
Collapse
Affiliation(s)
- Xue Gu
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Bingfeng Chen
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Shengcan Zhang
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinyu Zhai
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Hu
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hui Ye
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
3
|
Stephan A, Suhrmann JH, Skowron MA, Che Y, Poschmann G, Petzsch P, Kresbach C, Wruck W, Pongratanakul P, Adjaye J, Stühler K, Köhrer K, Schüller U, Nettersheim D. Molecular and epigenetic ex vivo profiling of testis cancer-associated fibroblasts and their interaction with germ cell tumor cells and macrophages. Matrix Biol 2024; 132:10-23. [PMID: 38851302 DOI: 10.1016/j.matbio.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Germ cell tumors (GCT) are the most common solid tumors in young men of age 15 - 40. In previous studies, we profiled the interaction of GCT cells with cells of the tumor microenvironment (TM), which showed that especially the 3D interaction of fibroblasts (FB) or macrophages with GCT cells influenced the growth behavior and cisplatin response as well as the transcriptome and secretome of the tumor cells, suggesting that the crosstalk of these cells with GCT cells is crucial for tumor progression and therapy outcome. In this study, we shed light on the mechanisms of activation of cancer-associated fibroblasts (CAF) in the GCT setting and their effects on GCT cells lines and the monocyte cell line THP-1. Ex vivo cultures of GCT-derived CAF were established and characterized molecularly and epigenetically by performing DNA methylation arrays, RNA sequencing, and mass spectrometry-based secretome analysis. We demonstrated that the activation state of CAF is influenced by their former prevailing tumor environment in which they have resided. Hereby, we postulate that seminoma (SE) and embryonal carcinoma (EC) activate CAF, while teratoma (TER) play only a minor role in CAF formation. In turn, CAF influence proliferation and the expression of cisplatin sensitivity-related factors in GCT cells lines as well as polarization of in vitro-induced macrophages by the identified effector molecules IGFBP1, LGALS3BP, LYVE1, and PTX3. Our data suggests that the vital interaction of CAF with GCT cells and with macrophages has a huge influence on shaping the extracellular matrix as well as on recruitment of immune cells to the TM. In conclusion, therapeutically interfering with CAF and / or macrophages in addition to the standard therapy might slow-down progression of GCT and re-shaping of the TM to a tumor-promoting environment. Significance: The interaction of CAF with GCT and macrophages considerably influences the microenvironment. Thus, therapeutically interfering with CAF might slow-down progression of GCT and re-shaping of the microenvironment to a tumor-promoting environment.
Collapse
Affiliation(s)
- Alexa Stephan
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jan-Henrik Suhrmann
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Margaretha A Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yue Che
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gereon Poschmann
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Catena Kresbach
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Wasco Wruck
- Institute for Stem cell Research and Regenerative Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Pailin Pongratanakul
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem cell Research and Regenerative Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Germany.
| |
Collapse
|
4
|
Xu W, Goreczny GJ, Forsythe I, Brennan G, Stowell T, Brock K, Capella B, Turner CE. Hic-5 regulates extracellular matrix-associated gene expression and cytokine secretion in cancer associated fibroblasts. Exp Cell Res 2024; 435:113930. [PMID: 38237846 PMCID: PMC10923124 DOI: 10.1016/j.yexcr.2024.113930] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gregory J Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Jnana Therapeutics, Boston, MA, USA
| | - Ian Forsythe
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Zymo Research Corp, Huntington Beach, CA, USA
| | - Grant Brennan
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Theresa Stowell
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Katia Brock
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Benjamin Capella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
5
|
Yamashita K, Takebayashi S, Murata W, Hirai N, Ito Y, Mitsui M, Saito M, Sato K, Terada M, Niizeki N, Suzuki A, Ogitani K, Fujikawa T, Komori M, Inoue N, Arai N, Maekawa M. Analytical Performance of a Novel Latex Turbidimetric Immunoassay, "Nanopia TARC", for TARC/CCL17 Measurement: A Retrospective Observational Study. Diagnostics (Basel) 2023; 13:2935. [PMID: 37761302 PMCID: PMC10529481 DOI: 10.3390/diagnostics13182935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Thymus- and activation-regulated chemokine (TARC, also known as CCL17) is used as a biomarker for atopic dermatitis. The methods currently used for its measurement are complex, time-consuming, and require large machinery, warranting the need for a method that is simple, has a quick turnaround time, and requires less complex machinery. We evaluated the analytical performance of a novel latex turbidimetric immunoassay method, "Nanopia TARC", on 174 residual serum samples from patients with skin or allergic diseases. This evaluation included the assessment of the limit of blank/detection/quantification (LOB/D/Q), precision, accuracy, linearity, interference, and commutability between Nanopia TARC and "HISCL TARC", based on the chemiluminescent enzyme immunoassay (CLEIA) method. The LOB/D/Q values were 13, 57, and 141 pg/mL, respectively. The coefficient of variation of the repeatability was 0.9-3.8%, and that of the intermediate precision was 2.1-5.4%. The total error of the accuracy was 1.9-13.4%. The linearity was 141 and 19,804 pg/mL for TARC. The correlation coefficient between Nanopia TARC and HISCL TARC determined using the Passing-Bablok regression analysis was 0.999. Furthermore, the concordance of diagnostic criteria with AD was 92%. Nanopia TARC was confirmed to have the same analytical performance for TARC measurement as the existing CLEIA method.
Collapse
Affiliation(s)
- Keita Yamashita
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Shiori Takebayashi
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Wataru Murata
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Nao Hirai
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Yui Ito
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Mayuka Mitsui
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Mina Saito
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Kei Sato
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Miyuki Terada
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Noriyasu Niizeki
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Akira Suzuki
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Kenya Ogitani
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Toshihiko Fujikawa
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Marie Komori
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Nozomi Inoue
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Norimitsu Arai
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Masato Maekawa
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| |
Collapse
|
6
|
Lupancu TJ, Eivazitork M, Hamilton JA, Achuthan AA, Lee KMC. CCL17/TARC in autoimmunity and inflammation-not just a T-cell chemokine. Immunol Cell Biol 2023; 101:600-609. [PMID: 36975092 DOI: 10.1111/imcb.12644] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023]
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.
Collapse
Affiliation(s)
- Tanya J Lupancu
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Mahtab Eivazitork
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Adrian A Achuthan
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin M-C Lee
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
7
|
Dragan M, Chen Z, Li Y, Le J, Sun P, Haensel D, Sureshchandra S, Pham A, Lu E, Pham KT, Verlande A, Vu R, Gutierrez G, Li W, Jang C, Masri S, Dai X. Ovol1/2 loss-induced epidermal defects elicit skin immune activation and alter global metabolism. EMBO Rep 2023; 24:e56214. [PMID: 37249012 PMCID: PMC10328084 DOI: 10.15252/embr.202256214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Skin epidermis constitutes the outer permeability barrier that protects the body from dehydration, heat loss, and myriad external assaults. Mechanisms that maintain barrier integrity in constantly challenged adult skin and how epidermal dysregulation shapes the local immune microenvironment and whole-body metabolism remain poorly understood. Here, we demonstrate that inducible and simultaneous ablation of transcription factor-encoding Ovol1 and Ovol2 in adult epidermis results in barrier dysregulation through impacting epithelial-mesenchymal plasticity and inflammatory gene expression. We find that aberrant skin immune activation then ensues, featuring Langerhans cell mobilization and T cell responses, and leading to elevated levels of secreted inflammatory factors in circulation. Finally, we identify failure to gain body weight and accumulate body fat as long-term consequences of epidermal-specific Ovol1/2 loss and show that these global metabolic changes along with the skin barrier/immune defects are partially rescued by immunosuppressant dexamethasone. Collectively, our study reveals key regulators of adult barrier maintenance and suggests a causal connection between epidermal dysregulation and whole-body metabolism that is in part mediated through aberrant immune activation.
Collapse
Affiliation(s)
- Morgan Dragan
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Zeyu Chen
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Department of Dermatology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Present address:
Institute of PsoriasisTongji University School of MedicineShanghaiChina
| | - Yumei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Johnny Le
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Peng Sun
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Daniel Haensel
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Program in Epithelial BiologyStanford University School of MedicineStanfordCAUSA
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Anh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Eddie Lu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Katherine Thanh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Amandine Verlande
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Remy Vu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Guadalupe Gutierrez
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Wei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Selma Masri
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Xing Dai
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
- Department of Dermatology, School of MedicineUniversity of CaliforniaIrvineCAUSA
| |
Collapse
|
8
|
Beppu H, Fukuda T, Otsubo N, Kawanishi T, Ogawa T, Abe Y, Endo M, Hanawa T, Sugita C, Kikkawa Y, Hatakeyama S, Yamada T, Wakai S. Serum chemokine CC-motif ligand 17 is a predictive marker of severe COVID-19 in haemodialysis patients: A retrospective observational study. Nephrology (Carlton) 2023; 28:240-248. [PMID: 36806795 DOI: 10.1111/nep.14151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Maintenance haemodialysis (HD) patients are at higher risk for severe coronavirus disease 2019 (COVID-19). Because of a limited number of facilities that can provide inpatient treatment for COVID-19 and HD, it is important to identify HD patients who are at high risk for severe COVID-19. For mild to moderate COVID-19 patients, chemokine CC-motif ligand 17 (CCL17) was reported to be a predictive marker for severe COVID-19; however, the validity of CCL17 among HD patients is unknown. METHODS This retrospective observational study enrolled 107 HD patients with mild or moderate COVID-19 at hospitalization (mean age 70.1 ± 15.1 years; 71.0% male). Receiver operating characteristic and logistic regression analyses were used to examine the predictive validity of indices for severe COVID-19. RESULTS During hospitalization, 32 patients developed severe COVID-19. Serum CCL17 collected at admission exhibited a higher area under the curve value (0.818) compared with that of other indicators including lactate dehydrogenase and C-reactive protein for the prediction of severe COVID-19. The optimal cut-off value for CCL17 was 150.5 pg/mL. A multi-variate logistic analysis revealed that CCL17 (above 150.5 pg/mL) was significantly associated with severe COVID-19 (Odds ratio, 0.063; 95% Confidence interval [CI], 0.017-0.227; p < .001) even after adjustment for covariates. The addition of the CCL17 to a model consisting of vaccination status, albumin, blood urea nitrogen, C-reacting protein and lactate dehydrogenase significantly improved classification performance for severe COVID-19 using the net reclassification (1.16, 95% CI: 0.82-1.50, p < .001) and integrated discrimination (0.18, 95% CI: 0.09-0.26, p < .001) improvement. CONCLUSION CCL17 levels in HD patients with mild or moderate COVID-19 predict risk of developing severe COVID-19.
Collapse
Affiliation(s)
- Hiroko Beppu
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
- Department of Cooperative Graduate School, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tatsuya Fukuda
- Department of Endocrinology and Metabolism, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Naoya Otsubo
- Department of Endocrinology and Metabolism, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Tomoko Kawanishi
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Toshie Ogawa
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Yasutomo Abe
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Mariko Endo
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Tomohide Hanawa
- Department of Pulmonary Medicine, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Chise Sugita
- Department of Pulmonary Medicine, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| | - Yoshiaki Kikkawa
- Department of Cooperative Graduate School, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shuji Hatakeyama
- Department of Pulmonary Medicine, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
- Department of General Internal Medicine/Infectious Diseases, Jichi Medical University Hospital, Tochigi, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachiko Wakai
- Department of Nephrology, Tokyo Metropolitan Okubo Hospital, Tokyo, Japan
| |
Collapse
|
9
|
Banshodani M, Kawanishi H, Hirai T, Kawai Y, Hashimoto S, Shintaku S, Moriishi M, Marubayashi S, Tsuchiya S. The predictive markers of severity and mortality in hospitalized hemodialysis patients with COVID-19 during Omicron epidemic. Ther Apher Dial 2023. [PMID: 36691364 DOI: 10.1111/1744-9987.13970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/05/2022] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Predictive markers and prognosis remain unclear in hospitalized hemodialysis (HD) patients with coronavirus disease 2019 (COVID-19) during the Omicron epidemic. METHODS We evaluated characteristics, laboratory parameters, and outcomes in hospitalized HD patients with COVID-19 (n = 102) at two centers between January and April 2022. RESULTS The 30-day mortality rate was higher in moderate-critical group (n = 43) than mild group (n = 59) (16.3% vs. 1.7%; p = 0.007), and higher in patients with lower CC chemokine ligand 17 (CCL17) levels (<95.0 pg/mL) compared with normal CCL17 levels (19.0% versus 4.9%; p = 0.03). In multivariate analyses, a low CCL17 level (p = 0.003) was associated with moderate-critical conditions, and moderate-critical conditions (p = 0.04) were associated with 30-day mortality, whereas CCL17 was not associated with 30-day mortality. CONCLUSIONS COVID-19 remains a fatal complication, and CCL17 was a predictive marker of severity in hospitalized HD patients during the Omicron epidemic.
Collapse
Affiliation(s)
- Masataka Banshodani
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Hideki Kawanishi
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Takayuki Hirai
- Kidney Disease and Dialysis, Akane-Foundation, Ajina Tsuchiya Hospital, Hatsukaichi, Japan
| | - Yusuke Kawai
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Shinji Hashimoto
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Sadanori Shintaku
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Misaki Moriishi
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Seiji Marubayashi
- Kidney Disease and Dialysis, Akane-Foundation, Ajina Tsuchiya Hospital, Hatsukaichi, Japan
| | - Shinichiro Tsuchiya
- Kidney Disease and Blood Purification Therapy, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| |
Collapse
|
10
|
Weier AK, Homrich M, Ebbinghaus S, Juda P, Miková E, Hauschild R, Zhang L, Quast T, Mass E, Schlitzer A, Kolanus W, Burgdorf S, Gruß OJ, Hons M, Wieser S, Kiermaier E. Multiple centrosomes enhance migration and immune cell effector functions of mature dendritic cells. J Cell Biol 2022; 221:e202107134. [PMID: 36214847 PMCID: PMC9555069 DOI: 10.1083/jcb.202107134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 03/01/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Centrosomes play a crucial role during immune cell interactions and initiation of the immune response. In proliferating cells, centrosome numbers are tightly controlled and generally limited to one in G1 and two prior to mitosis. Defects in regulating centrosome numbers have been associated with cell transformation and tumorigenesis. Here, we report the emergence of extra centrosomes in leukocytes during immune activation. Upon antigen encounter, dendritic cells pass through incomplete mitosis and arrest in the subsequent G1 phase leading to tetraploid cells with accumulated centrosomes. In addition, cell stimulation increases expression of polo-like kinase 2, resulting in diploid cells with two centrosomes in G1-arrested cells. During cell migration, centrosomes tightly cluster and act as functional microtubule-organizing centers allowing for increased persistent locomotion along gradients of chemotactic cues. Moreover, dendritic cells with extra centrosomes display enhanced secretion of inflammatory cytokines and optimized T cell responses. Together, these results demonstrate a previously unappreciated role of extra centrosomes for regular cell and tissue homeostasis.
Collapse
Affiliation(s)
- Ann-Kathrin Weier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Mirka Homrich
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Stephanie Ebbinghaus
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Pavel Juda
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Eliška Miková
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Lili Zhang
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Thomas Quast
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune System, University of Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences Institute, Cellular Immunology, University of Bonn, Bonn, Germany
| | - Oliver J. Gruß
- Institute of Genetics, University of Bonn, Bonn, Germany
| | - Miroslav Hons
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Stefan Wieser
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Castelldefels, Spain
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
Rodríguez-Fernández JL, Criado-García O. A meta-analysis indicates that the regulation of cell motility is a non-intrinsic function of chemoattractant receptors that is governed independently of directional sensing. Front Immunol 2022; 13:1001086. [PMID: 36341452 PMCID: PMC9630654 DOI: 10.3389/fimmu.2022.1001086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Chemoattraction, defined as the migration of a cell toward a source of a chemical gradient, is controlled by chemoattractant receptors. Chemoattraction involves two basic activities, namely, directional sensing, a molecular mechanism that detects the direction of a source of chemoattractant, and actin-based motility, which allows the migration of a cell towards it. Current models assume first, that chemoattractant receptors govern both directional sensing and motility (most commonly inducing an increase in the migratory speed of the cells, i.e. chemokinesis), and, second, that the signaling pathways controlling both activities are intertwined. We performed a meta-analysis to reassess these two points. From this study emerge two main findings. First, although many chemoattractant receptors govern directional sensing, there are also receptors that do not regulate cell motility, suggesting that is the ability to control directional sensing, not motility, that best defines a chemoattractant receptor. Second, multiple experimental data suggest that receptor-controlled directional sensing and motility can be controlled independently. We hypothesize that this independence may be based on the existence of separated signalling modules that selectively govern directional sensing and motility in chemotactic cells. Together, the information gathered can be useful to update current models representing the signalling from chemoattractant receptors. The new models may facilitate the development of strategies for a more effective pharmacological modulation of chemoattractant receptor-controlled chemoattraction in health and disease.
Collapse
|
12
|
Huang S, Thomsson KA, Jin C, Ryberg H, Das N, Struglics A, Rolfson O, Björkman LI, Eisler T, Schmidt TA, Jay GD, Krawetz R, Karlsson NG. Truncated lubricin glycans in osteoarthritis stimulate the synoviocyte secretion of VEGFA, IL-8, and MIP-1 α: Interplay between O-linked glycosylation and inflammatory cytokines. Front Mol Biosci 2022; 9:942406. [PMID: 36213120 PMCID: PMC9532613 DOI: 10.3389/fmolb.2022.942406] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
The primary aim of the study was to identify inflammatory markers relevant for osteoarthritis (OA)-related systemic (plasma) and local (synovial fluid, SF) inflammation. From this, we looked for inflammatory markers that coincided with the increased amount of O-linked Tn antigen (GalNAcα1-Ser/Thr) glycan on SF lubricin. Inflammatory markers in plasma and SF in OA patients and controls were measured using a 44-multiplex immunoassay. We found consistently 29 markers detected in both plasma and SF. The difference in their concentration and the low correlation when comparing SF and plasma suggests an independent inflammatory environment in the two biofluids. Only plasma MCP-4 and TARC increased in our patient cohort compared to control plasma. To address the second task, we concluded that plasma markers were irrelevant for a direct connection with SF glycosylation. Hence, we correlated the SF-inflammatory marker concentrations with the level of altered glycosylation of SF-lubricin. We found that the level of SF-IL-8 and SF-MIP-1α and SF-VEGFA in OA patients displayed a positive correlation with the altered lubricin glycosylation. Furthermore, when exposing fibroblast-like synoviocytes from both controls and OA patients to glycovariants of recombinant lubricin, the secretion of IL-8 and MIP-1α and VEGFA were elevated using lubricin with Tn antigens, while lubricin with sialylated and nonsialylated T antigens had less or no measurable effect. These data suggest that truncated glycans of lubricin, as found in OA, promote synovial proinflammatory cytokine production and exacerbate local synovial inflammation.
Collapse
Affiliation(s)
- Shan Huang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina A. Thomsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Ryberg
- Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nabangshu Das
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ola Rolfson
- Department of Orthopaedics, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena I. Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Eisler
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tannin A. Schmidt
- Biomedical Engineering Department, University of Connecticut Health Centre, Farmington, CT, United States
| | - Gregory D. Jay
- Department of Emergency Medicine, Warren Alpert Medical School and Division of Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Roman Krawetz
- Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Niclas G. Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Pharmacy, Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
13
|
Mastraftsi S, Vrioni G, Bakakis M, Nicolaidou E, Rigopoulos D, Stratigos AJ, Gregoriou S. Atopic Dermatitis: Striving for Reliable Biomarkers. J Clin Med 2022; 11:jcm11164639. [PMID: 36012878 PMCID: PMC9410433 DOI: 10.3390/jcm11164639] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
Atopic dermatitis (AD) is a highly heterogeneous inflammatory disease regarding both its pathophysiology and clinical manifestations. However, it is treated according to the “one-size-fits-all” approach, which may restrict response to treatment. Thus, there is an unmet need for the stratification of patients with AD into distinct endotypes and clinical phenotypes based on biomarkers that will contribute to the development of precision medicine in AD. The development of reliable biomarkers that may distinguish which patients with AD are most likely to benefit from specific targeted therapies is a complex procedure and to date none of the identified candidate biomarkers for AD has been validated for use in routine clinical practice. Reliable biomarkers in AD are expected to improve diagnosis, evaluate disease severity, predict the course of disease, the development of comorbidities, or the therapeutic response, resulting in effective and personalized treatment of AD. Among the studied AD potential biomarkers, thymus and activation-regulated chemokine/C-C motif ligand 17 (TARC/CCL17) has the greatest evidence-based support for becoming a reliable biomarker in AD correlated with disease severity in both children and adults. In this review, we present the most prominent candidate biomarkers in AD and their suggested use.
Collapse
Affiliation(s)
- Styliani Mastraftsi
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
- Correspondence: ; Tel.: +30-6974819341
| | - Georgia Vrioni
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michail Bakakis
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Electra Nicolaidou
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Dimitrios Rigopoulos
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Alexander J. Stratigos
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Stamatios Gregoriou
- 1st Department of Dermatology and Venereology, Andreas Sygros Hospital for Skin and Venereal Diseases, Medical School, National and Kapodistrian University of Athens, 16121 Athens, Greece
| |
Collapse
|
14
|
Zhang Y, Ye Y, Tang X, Wang H, Tanaka T, Tian R, Yang X, Wang L, Xiao Y, Hu X, Jin Y, Pang H, Du T, Liu H, Sun L, Xiao S, Dong R, Ferrucci L, Tian Z, Zhang S. CCL17 acts as a novel therapeutic target in pathological cardiac hypertrophy and heart failure. J Exp Med 2022; 219:213274. [PMID: 35687056 PMCID: PMC9194836 DOI: 10.1084/jem.20200418] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Circulating proteomic signatures of age are closely associated with aging and age-related diseases; however, the utility of changes in secreted proteins in identifying therapeutic targets for diseases remains unclear. Serum proteomic profiling of an age-stratified healthy population and further community-based cohort together with heart failure patients study demonstrated that circulating C-C motif chemokine ligand 17 (CCL17) level increased with age and correlated with cardiac dysfunction. Subsequent animal experiments further revealed that Ccll7-KO significantly repressed aging and angiotensin II (Ang II)–induced cardiac hypertrophy and fibrosis, accompanied by the plasticity and differentiation of T cell subsets. Furthermore, the therapeutic administration of an anti-CCL17 neutralizing antibody inhibited Ang II–induced pathological cardiac remodeling. Our findings reveal that chemokine CCL17 is identifiable as a novel therapeutic target in age-related and Ang II–induced pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicong Ye
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xufei Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lun Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaomin Hu
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Jin
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyu Pang
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian Du
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Honghong Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihong Sun
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Xiao
- Thermo Fisher Scientific (China) Co., Ltd, Changning, Shanghai, China
| | - Ruijia Dong
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Zhuang Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Nguyen HH, Shinkuma S, Hayashi R, Katsumi T, Nishiguchi T, Natsuga K, Fujita Y, Abe R. New insight of itch mediators and proinflammatory cytokines in epidermolysis bullosa. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2022. [DOI: 10.1002/cia2.12230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Hong Ha Nguyen
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Satoru Shinkuma
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
- Department of Dermatology Nara Medical University Kashihara Japan
| | - Ryota Hayashi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Tatsuya Katsumi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Tomoki Nishiguchi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Ken Natsuga
- Department of Dermatology Faculty of Medicine and Graduate School of Medicine Hokkaido University Sapporo Japan
| | - Yasuyuki Fujita
- Department of Dermatology Faculty of Medicine and Graduate School of Medicine Hokkaido University Sapporo Japan
- Department of Dermatology Sapporo City General Hospital Sapporo Japan
| | - Riichiro Abe
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| |
Collapse
|
16
|
Yoshie O. CCR4 as a Therapeutic Target for Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13215542. [PMID: 34771703 PMCID: PMC8583476 DOI: 10.3390/cancers13215542] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CCR4 is a chemokine receptor selectively expressed on normal T cell subsets such as type 2 helper T cells, skin-homing T cells and regulatory T cells, and on skin-associated T cell malignancies such as adult T cell leukemia/lymphoma (ATLL), which is etiologically associated with human T lymphocyte virus type 1 (HTLV-1), and cutaneous T cell lymphomas (CTCLs). Mogamulizumab is a fully humanized and glyco-engineered monoclonal anti-CCR4 antibody used for the treatment of refractory/relapsed ATLL and CTCLs, often resulting in complete remission. The clinical applications of Mogamulizumab are now being extended to solid tumors, exploring the therapeutic effect of regulatory T cell depletion. This review overviews the expression of CCR4 in various T cell subsets, HTLV-1-infected T cells, ATLL and CTCLs, and the clinical applications of Mogamulizumab. Abstract CCR4 is a chemokine receptor mainly expressed by T cells. It is the receptor for two CC chemokine ligands, CCL17 and CCL22. Originally, the expression of CCR4 was described as highly selective for helper T type 2 (Th2) cells. Later, its expression was extended to other T cell subsets such as regulatory T (Treg) cells and Th17 cells. CCR4 has long been regarded as a potential therapeutic target for allergic diseases such as atopic dermatitis and bronchial asthma. Furthermore, the findings showing that CCR4 is strongly expressed by T cell malignancies such as adult T cell leukemia/lymphoma (ATLL) and cutaneous T cell lymphomas (CTCLs) have led to the development and clinical application of the fully humanized and glyco-engineered monoclonal anti-CCR4 Mogamulizumab in refractory/relapsed ATLL and CTCLs with remarkable successes. However, Mogamulizumab often induces severe adverse events in the skin possibly because of its efficient depletion of Treg cells. In particular, treatment with Mogamulizumab prior to allogenic hematopoietic stem cell transplantation (allo-HSCT), the only curative option of these T cell malignancies, often leads to severe glucocorticoid-refractory graft-versus-host diseases. The efficient depletion of Treg cells by Mogamulizumab has also led to its clinical trials in advanced solid tumors singly or in combination with immune checkpoint inhibitors. The main focus of this review is CCR4; its expression on normal and malignant T cells and its significance as a therapeutic target in cancer immunotherapy.
Collapse
Affiliation(s)
- Osamu Yoshie
- Health and Kampo Institute, Sendai 981-3205, Japan;
- Kindai University, Osaka 577-8502, Japan
- Aoinosono-Sendai Izumi Long-Term Health Care Facility, Sendai 981-3126, Japan
| |
Collapse
|
17
|
Petkov S, Chiodi F. Distinct transcriptomic profiles of naïve CD4+ T cells distinguish HIV-1 infected patients initiating antiretroviral therapy at acute or chronic phase of infection. Genomics 2021; 113:3487-3500. [PMID: 34425224 DOI: 10.1016/j.ygeno.2021.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
We analyzed the whole transcriptome characteristics of blood CD4+ T naïve (TN) cells isolated from HIV-1 infected patients starting ART at acute (early ART = EA; n = 13) or chronic (late ART = LA; n = 11) phase of infection and controls (C; n = 15). RNA sequencing revealed 389 differentially expressed genes (DEGs) in EA and 810 in LA group in relation to controls. Comparison of the two groups of patients showed 183 DEGs. We focused on DEGs involved in apoptosis, inflammation and immune response. Clustering showed a poor separation of EA from C suggesting that these two groups present a similar transcriptomic profile of CD4+ TN cells. The comparison of EA and LA patients resulted in a high cluster purity revealing that different biological dysfunctions characterize EA and LA patients. The upregulated expression of several inflammatory chemokine genes distinguished the patient groups from C; CCL2 and CCL7, however, were downregulated in EA compared to LA patients. BCL2, an anti-apoptotic factor pivotal for naïve T cell homeostasis, distinguished both EA and LA from C. The expression of several DEGs involved in different inflammatory processes (TLR4, PTGS2, RAG1, IFNA16) was lower in EA compared LA. We conclude that although the transcriptome of CD4+ TN cells isolated from patients initiating ART at acute infection reveals a more quiescent phenotype, the survival profile of these cells still appears to be affected. Our results show that the detrimental process of inflammation is under more efficient control in EA patients.
Collapse
Affiliation(s)
- Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
18
|
Ono J, Takai M, Kamei A, Azuma Y, Izuhara K. Pathological Roles and Clinical Usefulness of Periostin in Type 2 Inflammation and Pulmonary Fibrosis. Biomolecules 2021; 11:1084. [PMID: 34439751 PMCID: PMC8391913 DOI: 10.3390/biom11081084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Periostin is known to be a useful biomarker for various diseases. In this article, we focus on allergic diseases and pulmonary fibrosis, for which we and others are now developing detection systems for periostin as a biomarker. Biomarker-based precision medicine in the management of type 2 inflammation and fibrotic diseases since heterogeneity is of utmost importance. Periostin expression is induced by type 2 cytokines (interleukin-4/-13) or transforming growth factor-β, and plays a vital role in the pathogenesis of allergic inflammation or interstitial lung disease, respectively, andits serum levels are correlated disease severity, prognosis and responsiveness to the treatment. We first summarise the importance of type 2 biomarker and then describe the pathological role of periostin in the development and progression of type 2 allergic inflammation and pulmonary fibrosis. In addition, then, we summarise the recent development of assay methods for periostin detection, and analyse the diseases in which periostin concentration is elevated in serum and local biological fluids and its usefulness as a biomarker. Furthermore, we describe recent findings of periostin as a biomarker in the use of biologics or anti-fibrotic therapy. Finally, we describe the factors that influence the change in periostin concentration under the healthy conditions.
Collapse
Affiliation(s)
- Junya Ono
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Masayuki Takai
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
- Division of Medical Biochemistry, Department of Biomolecular Science, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan;
| | - Ayami Kamei
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Yoshinori Azuma
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Science, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan;
| |
Collapse
|
19
|
Catherine J, Roufosse F. What does elevated TARC/CCL17 expression tell us about eosinophilic disorders? Semin Immunopathol 2021; 43:439-458. [PMID: 34009399 PMCID: PMC8132044 DOI: 10.1007/s00281-021-00857-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/14/2021] [Indexed: 12/19/2022]
Abstract
Eosinophilic disorders encompass a large spectrum of heterogeneous diseases sharing the presence of elevated numbers of eosinophils in blood and/or tissues. Among these disorders, the role of eosinophils can vary widely, ranging from a modest participation in the disease process to the predominant perpetrator of tissue damage. In many cases, eosinophilic expansion is polyclonal, driven by enhanced production of interleukin-5, mainly by type 2 helper cells (Th2 cells) with a possible contribution of type 2 innate lymphoid cells (ILC2s). Among the key steps implicated in the establishment of type 2 immune responses, leukocyte recruitment toward inflamed tissues is particularly relevant. Herein, the contribution of the chemo-attractant molecule thymus and activation-regulated chemokine (TARC/CCL17) to type 2 immunity will be reviewed. The clinical relevance of this chemokine and its target, C-C chemokine receptor 4 (CCR4), will be illustrated in the setting of various eosinophilic disorders. Special emphasis will be put on the potential diagnostic, prognostic, and therapeutic implications related to activation of the TARC/CCL17-CCR4 axis.
Collapse
Affiliation(s)
- Julien Catherine
- Department of Internal Medicine, Hôpital Erasme, 808 Route de Lennik, 1070, Brussels, Belgium. .,Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Gosselies, Brussels, Belgium.
| | - Florence Roufosse
- Department of Internal Medicine, Hôpital Erasme, 808 Route de Lennik, 1070, Brussels, Belgium.,Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Gosselies, Brussels, Belgium
| |
Collapse
|
20
|
Erazo AB, Wang N, Standke L, Semeniuk AD, Fülle L, Cengiz SC, Thiem M, Weighardt H, Strugnell RA, Förster I. CCL17-expressing dendritic cells in the intestine are preferentially infected by Salmonella but CCL17 plays a redundant role in systemic dissemination. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:891-904. [PMID: 33945673 PMCID: PMC8342217 DOI: 10.1002/iid3.445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Introduction Salmonella spp. are a recognized and global cause of serious health issues from gastroenteritis to invasive disease. The mouse model of human typhoid fever, which uses Salmonella enterica serovar Typhimurium (STM) in susceptible mouse strains, has revealed that the bacteria gain access to extraintestinal tissues from the gastrointestinal tract to cause severe systemic disease. Previous analysis of the immune responses against Salmonella spp. revealed the crucial role played by dendritic cells (DCs) in carrying STM from the intestinal mucosa to the mesenteric lymph nodes (mLNs), a key site for antigen presentation and T cell activation. In this study, we investigated the influence of chemokine CCL17 on the dissemination of STM. Methods WT, CCL17/EGFP reporter, or CCL17‐deficient mice were infected orally with STM (SL1344) or mCherry‐expressing STM for 1–3 days. Colocalization of STM with CCL17‐expressing DCs in Peyer's patches (PP) and mLN was analyzed by fluorescence microscopy. In addition, DCs and myeloid cell populations from naïve and Salmonella‐infected mice were analyzed by flow cytometry. Bacterial load was determined in PP, mLN, spleen, and liver 1 and 3 days after infection. Results Histological analysis revealed that CCL17‐expressing cells are located in close proximity to STM in the dome area of PP. We show that, in mLN, STM were preferentially located within CCL17+ rather than CCL17− DCs, besides other mononuclear phagocytes, and identified the CD103+ CD11b− DC subset as the main STM‐carrying DC population in the intestine. STM infection triggered upregulation of CCL17 expression in specific intestinal DC subsets in a tissue‐specific manner. The dissemination of STM from the gut to the mLN, however, was only moderately influenced by the presence of CCL17. Conclusion CCL17‐expressing DCs were preferentially infected by Salmonella in the intestine in comparison to other DC. Nevertheless, the production of CCL17 was not essential for the early dissemination of Salmonella from the gut to systemic organs.
Collapse
Affiliation(s)
- Anna B Erazo
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lena Standke
- Department for Innate Immunity and Metaflammation, Institute of Innate Immunity, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Adrian D Semeniuk
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lorenz Fülle
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sevgi C Cengiz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manja Thiem
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Richard A Strugnell
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
21
|
Renert-Yuval Y, Thyssen JP, Bissonnette R, Bieber T, Kabashima K, Hijnen D, Guttman-Yassky E. Biomarkers in atopic dermatitis-a review on behalf of the International Eczema Council. J Allergy Clin Immunol 2021; 147:1174-1190.e1. [PMID: 33516871 PMCID: PMC11304440 DOI: 10.1016/j.jaci.2021.01.013] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Yael Renert-Yuval
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Jacob P Thyssen
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Robert Bissonnette
- Department of Dermatology, Innovaderm Research, Montreal, Quebec, Canada
| | - Thomas Bieber
- Department of Dermatology and Allergy, Christine Kühne-Center for Allergy Research and Education, University of Bonn, Bonn, Germany
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University School of Medicine, Kyoto, Japan
| | - DirkJan Hijnen
- Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
22
|
Serum TARC Levels in Patients with Systemic Sclerosis: Clinical Association with Interstitial Lung Disease. J Clin Med 2021; 10:jcm10040660. [PMID: 33572144 PMCID: PMC7915627 DOI: 10.3390/jcm10040660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/27/2022] Open
Abstract
Systemic sclerosis (SSc) is a multisystem fibrotic disorder with autoimmune background. Accumulating evidence has highlighted the importance of T helper (Th) 2 cells in the pathogenesis of SSc and its complications. Because thymus and activation-regulated chemokine (TARC) is a potent chemoattractant for Th2 cells, we measured serum TARC levels in SSc patients and analyzed their correlation with interstitial lung disease (ILD), a major complication of SSc. Serum TARC levels were significantly elevated in patients with SSc, especially in those with the diffuse subtype, compared with healthy controls. In particular, dcSSc patients with SSc-associated ILD (SSc-ILD) showed higher TARC levels than those without SSc-ILD. However, there was no significant correlation between serum TARC levels and pulmonary function in SSc patients. Serum TARC levels did not correlate with serum levels of interleukin-13, an important Th2 cytokine, either. Furthermore, in the longitudinal study, serum TARC levels did not predict the onset or progression of SSc-ILD in patients with SSc. These results were in contrast with those of KL-6 and surfactant protein D, which correlated well with the onset, severity, and progression of SSc-ILD. Overall, these results suggest that serum TARC levels are not suitable for monitoring the disease activity of SSc-ILD.
Collapse
|
23
|
Sugiyama M, Kinoshita N, Ide S, Nomoto H, Nakamoto T, Saito S, Ishikane M, Kutsuna S, Hayakawa K, Hashimoto M, Suzuki M, Izumi S, Hojo M, Tsuchiya K, Gatanaga H, Takasaki J, Usami M, Kano T, Yanai H, Nishida N, Kanto T, Sugiyama H, Ohmagari N, Mizokami M. Serum CCL17 level becomes a predictive marker to distinguish between mild/moderate and severe/critical disease in patients with COVID-19. Gene 2021; 766:145145. [PMID: 32941953 PMCID: PMC7489253 DOI: 10.1016/j.gene.2020.145145] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 01/08/2023]
Abstract
COVID-19, a novel coronavirus-related illness, has spread worldwide. Patients with apparently mild/moderate symptoms can suddenly develop severe pneumonia. Therefore, almost all COVID-19 patients require hospitalization, which can reduce limited medical resources in addition to overwhelming medical facilities. To identify predictive markers for the development of severe pneumonia, a comprehensive analysis of serum chemokines and cytokines was conducted using serial serum samples from COVID-19 patients. The expression profiles were analyzed along the time axis. Serum samples of common diseases were enrolled from a BioBank to confirm the usefulness of predictive markers. Five factors, IFN-λ3, IL-6, IP-10, CXCL9, and CCL17, were identified as predicting the onset of severe/critical symptoms. The factors were classified into two categories. Category A included IFN-λ3, IL-6, IP-10, and CXCL9, and their values surged and decreased rapidly before the onset of severe pneumonia. Category B included CCL17, which provided complete separation between the mild/moderate and the severe/critical groups at an early phase of SARS-CoV-2 infection. The five markers provided a high predictive value (area under the receiver operating characteristic curve (AUROC): 0.9-1.0, p < 0.001). Low expression of CCL17 was specifically observed in pre-severe COVID-19 patients compared with other common diseases, and the predictive ability of CCL17 was confirmed in validation samples of COVID-19. The factors identified could be promising prognostic markers to distinguish between mild/moderate and severe/critical patients, enabling triage at an early phase of infection, thus avoiding overwhelming medical facilities.
Collapse
Affiliation(s)
- Masaya Sugiyama
- Genome Medical Sciences Project, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan.
| | - Noriko Kinoshita
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Satoshi Ide
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hidetoshi Nomoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Takato Nakamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Masahiro Ishikane
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Satoshi Kutsuna
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Kayoko Hayakawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Masao Hashimoto
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Manabu Suzuki
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Shinyu Izumi
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Masayuki Hojo
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Kiyoto Tsuchiya
- AIDS Clinical Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hiroyuki Gatanaga
- AIDS Clinical Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Jin Takasaki
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Masahide Usami
- Department of Child and Adolescent Psychiatry, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| | - Toshikazu Kano
- Department of Rheumatism and Collagen Diseases, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| | - Hidekatsu Yanai
- Department of Internal Medicine, Kohnodai Hospital, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| | - Nao Nishida
- Genome Medical Sciences Project, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| | - Tatsuya Kanto
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1, Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Haruhito Sugiyama
- Department of Respiratory Medicine, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan.
| | - Masashi Mizokami
- Genome Medical Sciences Project, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| |
Collapse
|
24
|
Machida H, Inoue S, Shibata Y, Kimura T, Sato K, Abe K, Murano H, Yang S, Nakano H, Sato M, Nemoto T, Sato C, Nishiwaki M, Yamauchi K, Igarashi A, Tokairin Y, Watanabe M. Thymus and activation-regulated chemokine (TARC/CCL17) predicts decline of pulmonary function in patients with chronic obstructive pulmonary disease. Allergol Int 2021; 70:81-88. [PMID: 32444304 DOI: 10.1016/j.alit.2020.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The deterioration of pulmonary function, such as FEV1-decline, is strongly associated with poor prognosis in patients with chronic obstructive pulmonary disease (COPD). However, few investigations shed light on useful biomarkers for predicting the decline of pulmonary function. We evaluated whether thymus and activation-regulated chemokine (TARC), a Th2 inflammation marker, could predict rapid FEV1-decline in COPD patients. METHODS We recruited 161 patients with stable COPD and performed pulmonary function test once every six months. At the time of registration, blood tests, including serum levels of TARC were performed. We assessed the correlation between changes in parameters of pulmonary function tests and serum levels of TARC. The rapid-decline in pulmonary function was determined using 25th percentile of change in FEV1 or FEV1 percent predicted (%FEV1) per year. RESULTS In the FEV1-rapid-decline group, the frequency of exacerbations, the degree of emphysema, and serum levels of TARC was higher than in the non-rapid-decline group. When using %FEV1 as a classifier instead of FEV1, age, the frequency of exacerbations, the degree of emphysema and serum levels of TARC in the rapid-decline group was significantly greater than those in the non-rapid-decline group. In univariate logistic regression analysis, TARC was the significant predictive factor for rapid-decline group. In multivariate analysis adjusted for emphysema, serum levels of TARC are independently significant predicting factors for the rapid-decline group. CONCLUSIONS TARC is an independent predictive biomarker for the rapid-decline in FEV1. Measuring serum TARC levels may help the management of COPD patients by predicting the risk of FEV1 decline.
Collapse
|
25
|
Role of Cytokines in EGPA and the Possibility of Treatment with an Anti-IL-5 Antibody. J Clin Med 2020; 9:jcm9123890. [PMID: 33265990 PMCID: PMC7760889 DOI: 10.3390/jcm9123890] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 01/22/2023] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a type of systemic vasculitis with eosinophilia in the peripheral blood, which is preceded by bronchial asthma or allergic disease. EGPA is pathologically characterized by microangiopathy granulomatosis vasculitis. Vasculitis can be exacerbated and cause central nervous system and cardiovascular disorders and gastrointestinal perforation. Histological examination reveals eosinophil infiltration and granulomas in lesions in areas such as the lung, nervous system, and skin. Laboratory tests show inflammatory findings such as C-reactive protein (CRP) elevation, increased eosinophils, elevated serum IgE, and elevated myeloperoxidase-anti-neutrophil cytoplasmic antibodies (MPO-ANCA). MPO-ANCA is positive in approximately 40-70% of cases of this disease. EGPA is a necrotizing vasculitis that affects small- and medium-sized blood vessels; however, it differs from other types of ANCA-related vasculitis (such as microscopic polyangiitis and granulomatosis) because it is preceded by bronchial asthma and eosinophilia in the blood and tissues. Treatment with immunosuppressive agents such as steroids or cyclophosphamide depends on the Five Factor Score, which predicts the prognosis and severity of the condition. If the effect of appropriate treatment with steroids is insufficient, the anti-interleukin-5 antibody mepolizumab can be administered. The combination of mepolizumab with standard treatment leads to a significantly longer duration of remission, a higher proportion of patients who achieve sustained remission, and less steroid use than with a placebo.
Collapse
|
26
|
Yamashita M, Miyoshi M, Iwai M, Takeda R, Ono T, Kabuki T. Lactobacillus helveticus SBT2171 Alleviates Perennial Allergic Rhinitis in Japanese Adults by Suppressing Eosinophils: A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2020; 12:E3620. [PMID: 33255731 PMCID: PMC7760906 DOI: 10.3390/nu12123620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
This article examines the effects of fermented milk (FM) containing Lactobacillus helveticus SBT2171 (LH2171) on the subjective symptoms of individuals with mild and moderate perennial allergy. Two hundred subjects were divided into two groups and consumed FM containing LH2171 or placebo FM once per day for 16 weeks. The primary endpoints were defined as per the degree of nasal and ocular symptoms and difficulty in daily life as determined by the Japanese guidelines for allergy rhinitis and the Japanese allergic rhinitis standard quality of life questionnaire, respectively. The secondary endpoints included parameters related to allergic symptoms in the blood and nasal fluids, as well as the mental status. The severity of allergic rhinitis significantly improved in the LH2171 group compared to that in the placebo group. Additionally, the LH2171 group showed a significantly lower degree of "stuffy nose" (as per the diary survey) than the placebo group. Eosinophil counts in the nasal fluids and in the blood were significantly lower in the LH2171 group compared to the placebo group. Thus, the oral administration of FM containing LH2171 cells alleviated perennial allergic rhinitis in individuals with mild and moderate symptoms, possibly via suppression of eosinophils in both the blood and nasal fluids.
Collapse
Affiliation(s)
- Maya Yamashita
- Milk Science Research Institute, MEGMILK SNOW BRAND Co. Ltd., 1-1-2, Minamidai, Kawagoe, Saitama 350-1165, Japan; (M.Y.); (M.M.); (M.I.)
| | - Masaya Miyoshi
- Milk Science Research Institute, MEGMILK SNOW BRAND Co. Ltd., 1-1-2, Minamidai, Kawagoe, Saitama 350-1165, Japan; (M.Y.); (M.M.); (M.I.)
| | - Masayuki Iwai
- Milk Science Research Institute, MEGMILK SNOW BRAND Co. Ltd., 1-1-2, Minamidai, Kawagoe, Saitama 350-1165, Japan; (M.Y.); (M.M.); (M.I.)
| | - Ryuji Takeda
- Department of Nutritional Sciences for Well-Being, Faculty of Health Sciences for Welfare, Kansai University of Welfare Sciences, 3-11-1, Ashigaoka, Kashiwara, Osaka 582-0026, Japan;
| | - Takahiro Ono
- Ueno-Asagao Clinic, 6F Kairaku Building, 2-7-5, Higashiueno, Taito, Tokyo 110-0015, Japan;
| | - Toshihide Kabuki
- Milk Science Research Institute, MEGMILK SNOW BRAND Co. Ltd., 1-1-2, Minamidai, Kawagoe, Saitama 350-1165, Japan; (M.Y.); (M.M.); (M.I.)
| |
Collapse
|
27
|
Poppema S. Lymphocyte predominant Hodgkin lymphoma, antigen-driven after all? J Pathol 2020; 253:1-10. [PMID: 33044742 DOI: 10.1002/path.5567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/22/2020] [Accepted: 10/06/2020] [Indexed: 01/12/2023]
Abstract
Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) was suggested as an entity separate from other types of Hodgkin lymphoma 40 years ago and recognized in the WHO classification in 2001. Based on its relatively benign course with late distant relapses, relation with lymph node hyperplasia with progressively transformed germinal centers, presence of clonal immunoglobulin gene rearrangements with somatic hypermutations and ongoing mutations, and relation with a number of inherited defects affecting the immune system, it has been suspected that NLPHL might be antigen-driven. Recent evidence has shown that cases of IgD-positive NLPHL are associated with infection by Moraxella catarrhalis, a common bacterium in the upper respiratory tract and in lymph nodes. This review summarizes the evidence for NLPHL as a B-cell lymphoma involving follicular T-lymphocytes normally found in germinal centers, its molecular features and relation to inherited immune defects, and its relation and differential diagnosis from similar entities. Finally, it discusses the evidence that in many cases a watch and wait policy might be a viable initial management strategy. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sibrandes Poppema
- School of Medical and Health Sciences, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
28
|
Tashiro Y, Azukizawa H, Asada H, Niihara H, Morita E, Yamauchi T, Mizukawa Y, Kusakabe Y, Numazawa S, Izumi M, Sueki H, Watanabe H. Drug-induced hypersensitivity syndrome/drug reaction with eosinophilia and systemic symptoms due to lamotrigine differs from that due to other drugs. J Dermatol 2019; 46:226-233. [PMID: 30663091 DOI: 10.1111/1346-8138.14776] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/09/2018] [Indexed: 12/17/2022]
Abstract
Drug-induced hypersensitivity syndrome (DIHS), also referred to as drug reaction with eosinophilia and systemic symptoms (DRESS), is a multi-organ systemic drug reaction characterized by hematological abnormalities and reactivation of human herpesvirus-6 (HHV-6). DIHS/DRESS is typically associated with a limited number of drugs, such as the anticonvulsants. Our group has treated 12 patients for DIHS/DRESS due to lamotrigine (LTG), but their presentation differed from that of patients with DIHS/DRESS caused by other drugs. The aim of the present study was to identify significant differences between DIHS/DRESS caused by LTG versus other drugs. We retrospectively reviewed data of 12 patients with DIHS/DRESS caused by LTG and 32 patients with DIHS/DRESS due to other drugs. The increase in alanine aminotransferase level was significantly milder in the LTG group than the DIHS/DRESS group due to other drugs. The percentage of atypical lymphocytes in the blood during DIHS/DRESS was lower in the LTG group. Serum levels of lactate dehydrogenase and thymus and activation-regulated chemokine were also lower in the LTG group. There were fewer DIHS/DRESS patients with HHV-6 reactivation in the LTG group than in the group treated with other drugs. Lymphocyte transformation after DIHS/DRESS onset was faster in the LTG group. The two groups did not differ with respect to the interval from first drug intake to rash, white blood cell count, blood eosinophilia or DRESS score. There were no significant histopathological differences between the two groups. The features of LTG-associated DIHS/DRESS and DIHS/DRESS due to other drugs differ.
Collapse
Affiliation(s)
- Yasuya Tashiro
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Hiroaki Azukizawa
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| | - Hiroyuki Niihara
- Department of Dermatology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Eishin Morita
- Department of Dermatology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Teruo Yamauchi
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Yoshiko Mizukawa
- Department of Dermatology, Kyorin University School of Medicine, Tokyo, Japan
| | | | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University, Tokyo, Japan
| | - Miki Izumi
- Department of Medical Education, Showa University School of Medicine, Tokyo, Japan
| | - Hirohiko Sueki
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Hideaki Watanabe
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Wang Y, Weng X, Wang L, Hao M, Li Y, Hou L, Liang Y, Wu T, Yao M, Lin G, Jiang Y, Fu G, Hou Z, Meng X, Lu J, Wang J. HIC1 deletion promotes breast cancer progression by activating tumor cell/fibroblast crosstalk. J Clin Invest 2018; 128:5235-5250. [PMID: 30204129 DOI: 10.1172/jci99974] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 09/04/2018] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BrCa) is the malignant tumor that most seriously threatens female health; however, the molecular mechanism underlying its progression remains unclear. Here, we found that conditional deletion of hypermethylated in cancer 1 (HIC1) in the mouse mammary gland might contribute to premalignant transformation in the early stage of tumor formation. Moreover, the chemokine (C-X-C motif) ligand 14 (CXCL14) secreted by HIC1-deleted BrCa cells bound to its cognate receptor GPR85 on mammary fibroblasts in the microenvironment and was responsible for activating these fibroblasts via the ERK1/2, Akt, and neddylation pathways, whereas the activated fibroblasts promoted BrCa progression via the induction of epithelial-mesenchymal transition (EMT) by the C-C chemokine ligand 17 (CCL17)/CC chemokine receptor 4 (CCR4) axis. Finally, we confirmed that the HIC1-CXCL14-CCL17 loop was associated with the malignant progression of BrCa. Therefore, the crosstalk between HIC1-deleted BrCa cells and mammary fibroblasts might play a critical role in BrCa development. Exploring the progression of BrCa from the perspective of microenvironment will be beneficial for identifying the potential prognostic markers of breast tumor and providing more effective treatment strategies.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoling Weng
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Luoyang Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, China
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yue Li
- Pathology Center, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidan Hou
- Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Liang
- Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianqi Wu
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Mengfei Yao
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yiwei Jiang
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guohui Fu
- Pathology Center, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoyuan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangjun Meng
- Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinsong Lu
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianhua Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,School of Medicine, Anhui University of Science & Technology, Huainan, Anhui, China
| |
Collapse
|
30
|
Loss of GRHL3 leads to TARC/CCL17-mediated keratinocyte proliferation in the epidermis. Cell Death Dis 2018; 9:1072. [PMID: 30341279 PMCID: PMC6195598 DOI: 10.1038/s41419-018-0901-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 07/20/2018] [Accepted: 07/22/2018] [Indexed: 12/21/2022]
Abstract
Identifying soluble factors that influence epidermal integrity is critical for the development of preventative and therapeutic strategies for disorders such as ichthyosis, psoriasis, dermatitis and epidermal cancers. The transcription factor Grainyhead-like 3 (GRHL3) is essential for maintaining barrier integrity and preventing development of cutaneous squamous cell carcinoma (SCC); however, how loss of this factor, which in the skin is expressed exclusively within suprabasal epidermal layers triggers proliferation of basal keratinocytes, had thus far remained elusive. Our present study identifies thymus and activation-regulated chemokine (TARC) as a novel soluble chemokine mediator of keratinocyte proliferation following loss of GRHL3. Knockdown of GRHL3 in human keratinocytes showed that of 42 cytokines examined, TARC was the only significantly upregulated chemokine. Mouse skin lacking Grhl3 presented an inflammatory response with hallmarks of TARC activation, including heightened induction of blood clotting, increased infiltration of mast cells and pro-inflammatory T cells, increased expression of the pro-proliferative/pro-inflammatory markers CD3 and pSTAT3, and significantly elevated basal keratinocyte proliferation. Treatment of skin cultures lacking Grhl3 with the broad spectrum anti-inflammatory 5-aminosalicylic acid (5ASA) partially restored epidermal differentiation, indicating that abnormal keratinocyte proliferation/differentiation balance is a key driver of barrier dysfunction following loss of Grhl3, and providing a promising therapeutic avenue in the treatment of GRHL3-mediated epidermal disorders.
Collapse
|
31
|
Nguyen CTH, Kambe N, Ueda-Hayakawa I, Kishimoto I, Ly NTM, Mizuno K, Okamoto H. TARC expression in the circulation and cutaneous granulomas correlates with disease severity and indicates Th2-mediated progression in patients with sarcoidosis. Allergol Int 2018; 67:487-495. [PMID: 29598931 DOI: 10.1016/j.alit.2018.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/07/2018] [Accepted: 02/15/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Sarcoidosis is a systemic disorder characterized by the accumulation of lymphocytes and monocyte/macrophage lineage cells that results in the formation of non-caseating granulomas. Thymus- and activation-regulated chemokine (TARC)/CCL17 is an important chemokine in the amplification of Th2 responses, which are achieved by recruiting CCR4-expressing CD4+ T lymphocytes. TARC concentrations are known to increase in the serum of sarcoidosis patients; however, its role in the assessment of severity and prognosis of sarcoidosis remains unknown. The objective of this study is to elucidate the role of TARC in sarcoidosis by investigating its expression in peripheral blood and at inflammatory sites. We also examined its relationship with clinical features. METHODS Serum levels of TARC, soluble interleukin 2 receptor, angiotensin-converting enzyme, and lysozyme were measured in 82 sarcoidosis patients. The Th1 and Th2 balance in circulating CD4+ T cells was evaluated by flow cytometry. The immunohistochemical staining of TARC and CCR4 was performed in order to identify the source of TARC in affected skin tissues. RESULTS TARC serum levels were elevated in 78% of patients and correlated with disease severity. The percentage of CCR4+ cells and the CCR4+/CXCR3+ cell ratios were significantly higher in sarcoidosis patients than in normal subjects (P = 0.002 and P = 0.015, respectively). Moreover, TARC was expressed by monocyte/macrophage lineage cells within granulomas. The abundancy as well as distribution of TARC staining correlated with its serum levels. CONCLUSIONS The present results suggest that elevations in TARC drive an imbalanced Th2- weighted immune reaction and might facilitate prolonged inflammatory reactions in sarcoidosis.
Collapse
Affiliation(s)
- Chuyen Thi Hong Nguyen
- Department of Dermatology, Kansai Medical University, Osaka, Japan; Department of Dermatology and Venereology, University of Medicine and Pharmacy at Ho Chi Minh City, Vietnam
| | - Naotomo Kambe
- Department of Dermatology, Kansai Medical University, Osaka, Japan; Allergy Center, Kansai Medical University, Osaka, Japan.
| | | | - Izumi Kishimoto
- Department of Dermatology, Kansai Medical University, Osaka, Japan; Allergy Center, Kansai Medical University, Osaka, Japan
| | - Nhung Thi My Ly
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | - Kana Mizuno
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | - Hiroyuki Okamoto
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
32
|
Ruytinx P, Proost P, Van Damme J, Struyf S. Chemokine-Induced Macrophage Polarization in Inflammatory Conditions. Front Immunol 2018; 9:1930. [PMID: 30245686 PMCID: PMC6137099 DOI: 10.3389/fimmu.2018.01930] [Citation(s) in RCA: 316] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022] Open
Abstract
Macrophages represent a heterogeneous cell population and are known to display a remarkable plasticity. In response to distinct micro-environmental stimuli, e.g., tumor stroma vs. infected tissue, they polarize into different cell subtypes. Originally, two subpopulations were defined: classically activated macrophages or M1, and alternatively activated macrophages or M2. Nowadays, the M1/M2 classification is considered as an oversimplified approach that does not adequately cover the total spectrum of macrophage phenotypes observed in vivo. Especially in pathological circumstances, macrophages behave as plastic cells modifying their expression and transcription profile along a continuous spectrum with M1 and M2 phenotypes as extremes. Here, we focus on the effect of chemokines on macrophage differentiation and polarization in physiological and pathological conditions. In particular, we discuss chemokine-induced macrophage polarization in inflammatory diseases, including obesity, cancer, and atherosclerosis.
Collapse
Affiliation(s)
- Pieter Ruytinx
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, REGA Institute KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, REGA Institute KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, REGA Institute KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, REGA Institute KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Svensson M, Chen P. Human Organotypic Respiratory Models. Curr Top Microbiol Immunol 2018:29-54. [PMID: 29808337 DOI: 10.1007/82_2018_91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Biomedical research aiming to understand the molecular basis of human lung tissue development, homeostasis and disease, or to develop new therapies for human respiratory diseases, requires models that faithfully recapitulate the human condition. This has stimulated biologists and engineers to develop in vitro organotypic models mimicking human respiratory tissues. In this chapter, we provide examples of different types of model systems ranging from simple unicellular cultures to more complex multicellular systems. The models contain, in varying degree, cell types present in real tissue in combination with different extracellular matrix components that can critically affect cell phenotype and function. We also describe how organotypic respiratory models can be combined with human innate immune cells, to better recapitulate tissue inflammation, a key component in, for example, infectious diseases. These models have the potential to provide new insights into lung physiology, tissue infection and inflammation, disease mechanisms, as well as provide a platform for identification of novel targets and screening of candidate drugs in human lung disorders.
Collapse
Affiliation(s)
- Mattias Svensson
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden.
| | - Puran Chen
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden
| |
Collapse
|
34
|
Lochman I, Švachová V, Mílková Pavlíková K, Medřická H, Novák V, Trilecová L, Pavliska L, Procházka V. Serum Cytokine and Growth Factor Levels in Children with Autism Spectrum Disorder. Med Sci Monit 2018; 24:2639-2646. [PMID: 29705814 PMCID: PMC5946742 DOI: 10.12659/msm.906817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The immune system may have a role in the pathogenesis of autism spectrum disorder (ASD), including typical and atypical autism. The aim of this study was to determine whether a cytokine and growth factor panel could be identified for the diagnosis and prognosis in children with ASD, including typical and atypical autism. MATERIAL AND METHODS This study included 26 children with ASD (typical or atypical) and 11 of their siblings who did not have ASD. A panel of ten serum cytokines and growth factors were investigated using addressable laser bead assay (ALBIA) and enzyme-linked immunosorbent assay (ELISA) kits. Results were correlated with scores using the Childhood Autism Rating Scale (CARS) and Autism Diagnostic Observation Schedule (ADOS) for the children with ASD and compared with the findings from their siblings without ASD. RESULTS There were no statistically significant differences in serum cytokine and growth factor levels between children with ASD and their siblings. The scores using CARS and ADOS were significantly greater in children with typical autism compared with children with atypical autism as part of the ASD spectrum. Serum levels of cytokines and growth factors showed a positive correlation with CARS and ADOS scores but differed between children with typical and atypical autism and their siblings. CONCLUSIONS The findings of this study showed that serum measurement of appropriately selected panels of cytokines and growth factors might have a role in the diagnosis of ASD.
Collapse
Affiliation(s)
- Ivo Lochman
- Department of Immunology and Serology, The SPADIA LAB Laboratory Plc., Ostrava, Czech Republic
| | - Veronika Švachová
- Department of Immunology and Serology, The SPADIA LAB Laboratory Plc., Ostrava, Czech Republic
| | | | - Hana Medřická
- Department of Paediatric Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Vilém Novák
- Department of Paediatric Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Lenka Trilecová
- Department of Immunology and Serology, The SPADIA LAB Laboratory Plc., Ostrava, Czech Republic
| | - Lubomír Pavliska
- Department of the Deputy Director for Science and Research, University Hospital Ostrava, Ostrava, Czech Republic
| | - Václav Procházka
- Department of the Deputy Director for Science and Research, University Hospital Ostrava, Ostrava, Czech Republic
| |
Collapse
|
35
|
Novel Models to Study Stromal Cell-Leukocyte Interactions in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:131-146. [PMID: 30155626 DOI: 10.1007/978-3-319-78127-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To study human immunology in general and stromal immunology in particular, it is highly motivated to move from monolayers to 3D cultures, such as organotypic models, that better mimic the function of living tissue. These models can potentially contain most if not all cell types present in tissues, in combination with different extracellular matrix components that can critically affect cell phenotype. Besides their well-established use in studies of tissue-specific cells, such as epithelial cells, endothelial cells and stromal fibroblasts in combination with extracellular components, these models have also been shown to be valuable to study how tissue participates in the regulation of leukocyte differentiation and function. Organotypic models with leukocytes represent novel powerful tools to study human stromal immunology and mechanisms involved in the regulation of leukocyte functions and inflammatory processes in human health and disease. In particular, these models are robust, long-lived and reproducible and allow monitoring of disease progression in real time, as well as the mixing of cellular constituents from healthy and pathological tissues. These models are also easy to manipulate, either genetically or by adding external stimulants, such as cytokines and pathogens, to mimic pathological conditions. It is thus not surprising that these models are proposed to be useful in toxicology screening assays, evaluating therapeutic efficacy of drugs and antibiotics, as well as in personalized medicine. Within this chapter, the most recent developments in creating organotypic models for the purpose of study of human leukocyte and stromal cell interactions, in health and disease, will be discussed, in particular focusing on live imaging. Special emphasis will be given on an organotypic model resembling human lung and its usefulness in studying the fine control of physiological and pathological processes in human health and disease. Using these models in studies on human stromal cell and leukocyte interactions will likely help identifying novel disease traits and may point out new potential targets to monitor and treat human diseases.
Collapse
|
36
|
Fülle L, Steiner N, Funke M, Gondorf F, Pfeiffer F, Siegl J, Opitz FV, Haßel SK, Erazo AB, Schanz O, Stunden HJ, Blank M, Gröber C, Händler K, Beyer M, Weighardt H, Latz E, Schultze JL, Mayer G, Förster I. RNA Aptamers Recognizing Murine CCL17 Inhibit T Cell Chemotaxis and Reduce Contact Hypersensitivity In Vivo. Mol Ther 2017; 26:95-104. [PMID: 29103909 PMCID: PMC5763148 DOI: 10.1016/j.ymthe.2017.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 01/21/2023] Open
Abstract
The chemokine CCL17, mainly produced by dendritic cells (DCs) in the immune system, is involved in the pathogenesis of various inflammatory diseases. As a ligand of CCR4, CCL17 induces chemotaxis and facilitates T cell-DC interactions. We report the identification of two novel RNA aptamers, which were validated in vitro and in vivo for their capability to neutralize CCL17. Both aptamers efficiently inhibited the directed migration of the CCR4+ lymphoma line BW5147.3 toward CCL17 in a dose-dependent manner. To study the effect of these aptamers in vivo, we used a murine model of contact hypersensitivity. Systemic application of the aptamers significantly prevented ear swelling and T cell infiltration into the ears of sensitized mice after challenge with the contact sensitizer. The results of this proof-of-principle study establish aptamers as potent inhibitors of CCL17-mediated chemotaxis. Potentially, CCL17-specific aptamers may be used therapeutically in humans to treat or prevent allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Lorenz Fülle
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Nancy Steiner
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Markus Funke
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Fabian Gondorf
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Franziska Pfeiffer
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Julia Siegl
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Friederike V Opitz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Silvana K Haßel
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Anna Belen Erazo
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Oliver Schanz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - H James Stunden
- Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Michael Blank
- AptaIT, Am Klopferspitz 19a, 82152 Planegg-Martinsried, Germany
| | - Carsten Gröber
- AptaIT, Am Klopferspitz 19a, 82152 Planegg-Martinsried, Germany
| | - Kristian Händler
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Marc Beyer
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany; Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Günter Mayer
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany.
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| |
Collapse
|
37
|
Dantoft W, Martínez-Vicente P, Jafali J, Pérez-Martínez L, Martin K, Kotzamanis K, Craigon M, Auer M, Young NT, Walsh P, Marchant A, Angulo A, Forster T, Ghazal P. Genomic Programming of Human Neonatal Dendritic Cells in Congenital Systemic and In Vitro Cytomegalovirus Infection Reveal Plastic and Robust Immune Pathway Biology Responses. Front Immunol 2017; 8:1146. [PMID: 28993767 PMCID: PMC5622154 DOI: 10.3389/fimmu.2017.01146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
Neonates and especially premature infants are highly susceptible to infection but still can have a remarkable resilience that is poorly understood. The view that neonates have an incomplete or deficient immune system is changing. Human neonatal studies are challenging, and elucidating host protective responses and underlying cognate pathway biology, in the context of viral infection in early life, remains to be fully explored. In both resource rich and poor settings, human cytomegalovirus (HCMV) is the most common cause of congenital infection. By using unbiased systems analyses of transcriptomic resources for HCMV neonatal infection, we find the systemic response of a preterm congenital HCMV infection, involves a focused IFN regulatory response associated with dendritic cells. Further analysis of transcriptional-programming of neonatal dendritic cells in response to HCMV infection in culture revealed an early dominant IFN-chemokine regulatory subnetworks, and at later times the plasticity of pathways implicated in cell-cycle control and lipid metabolism. Further, we identify previously unknown suppressed networks associated with infection, including a select group of GPCRs. Functional siRNA viral growth screen targeting 516-GPCRs and subsequent validation identified novel GPCR-dependent antiviral (ADORA1) and proviral (GPR146, RGS16, PTAFR, SCTR, GPR84, GPR85, NMUR2, FZ10, RDS, CCL17, and SORT1) roles. By contrast a gene family cluster of protocadherins is significantly differentially induced in neonatal cells, suggestive of possible immunomodulatory roles. Unexpectedly, programming responses of adult and neonatal dendritic cells, upon HCMV infection, demonstrated comparable quantitative and qualitative responses showing that functionally, neonatal dendritic cell are not overly compromised. However, a delay in responses of neonatal cells for IFN subnetworks in comparison with adult-derived cells are notable, suggestive of subtle plasticity differences. These findings support a set-point control mechanism rather than immaturity for explaining not only neonatal susceptibility but also resilience to infection. In summary, our findings show that neonatal HCMV infection leads to a highly plastic and functional robust programming of dendritic cells in vivo and in vitro. In comparison with adults, a minimal number of subtle quantitative and temporal differences may contribute to variability in host susceptibility and resilience, in a context dependent manner.
Collapse
Affiliation(s)
- Widad Dantoft
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Pablo Martínez-Vicente
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - James Jafali
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lara Pérez-Martínez
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Quantitative Proteomics, Institute of Molecular Biology, Mainz, Germany
| | - Kim Martin
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Synexa Life Sciences, Cape Town, South Africa
| | - Konstantinos Kotzamanis
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie Craigon
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manfred Auer
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,SynthSys-Centre for Synthetic and Systems Biology, School of Engineering, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil T Young
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Paul Walsh
- NSilico Life Science and Department of Computing, Institute of Technology, Cork, Ireland
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Thorsten Forster
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Meng X, Gao W, Tang Y, Shen Z, Wang Z. Alterations of Serum IP-10 and TARC in Patients with Early Acute Rejection after Liver Transplantation. Cell Physiol Biochem 2017; 41:1063-1071. [DOI: 10.1159/000464114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/27/2016] [Indexed: 12/26/2022] Open
Abstract
Background/Aims: To analyze alterations of interferon-γ-induced protein 10 (IP-10) and thymus and activation-regulated chemokine (TARC) levels in early acute liver transplantation rejection. Methods: Thirty-six patients with early acute liver transplantation rejection were classified as non-, mild, moderate, and severe rejection groups. The levels of serum IP-10 and TARC were determined on days 3, 2, 1, and 0 before biopsy. Results: The IP-10 activities in all rejection groups were significantly higher (p < 0.05) than those in the non-rejection group at all time points and correlated with the extent of rejection (p < 0.05). The differences in TARC among the three rejection groups were significant (p < 0.05), and its highest level was found in the mild rejection group at all observed time points, whereas its lowest level was detected in the severe rejection group. The analysis of the TARC/IP-10 ratio revealed that the volume was correlated with the rejection degree. This ratio in the moderate and severe rejection groups on days 2, 1, and 0 before biopsy were 20% lower than that before transplantation. Conclusion: Serum IP-10 showed an increasing trend during early acute liver transplantation rejection. IP-10 increase or TARC/IP-10 ratio decrease combining with abnormal hepatic enzymatic alteration could be a valuable and specific sign for early rejection of the transplanted liver.
Collapse
|
39
|
Komatsu-Fujii T, Kaneko S, Chinuki Y, Suyama Y, Ohta M, Niihara H, Morita E. Serum TARC levels are strongly correlated with blood eosinophil count in patients with drug eruptions. Allergol Int 2017; 66:116-122. [PMID: 27497618 DOI: 10.1016/j.alit.2016.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND This study aims to evaluate the relationship between serum thymus and activation-regulated chemokine (TARC) levels with various clinicopathological conditions in patients with drug eruptions. The value of TARC in diagnosing drug-induced hypersensitivity syndrome (DIHS) was also examined. METHODS Study participants included 84 patients who presented with generalized eruptions suspected to be drug-related, including DIHS, Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN), maculopapular exanthema (MPE), erythema multiforme (EM), erythroderma, and toxicoderma. The correlation coefficients between serum TARC levels and clinical parameters in peripheral blood samples were calculated. RESULTS Serum TARC levels in patients with DIHS were higher than those found in patients with SJS/TEN, MPE, EM, and toxicoderma. TARC levels had 100% sensitivity and 92.3% specificity in diagnosing DIHS, with a threshold value of 13,900 pg/mL. Serum TARC levels positively correlated with age, white blood cell (WBC) count, neutrophil count, eosinophil count, monocyte count, atypical lymphocyte (Aty-ly) count, serum blood urea nitrogen (BUN) levels, and creatinine (Cr) levels. It negatively correlated with serum total protein (TP), albumin (Alb), and estimated glomerular filtration rate (eGFR). Among these clinical parameters, blood eosinophil counts were most strongly correlated with serum TARC levels, with a correlation coefficient of 0.53. CONCLUSIONS Serum TARC levels are well correlated with blood eosinophil counts in patients with generalized drug eruptions, indicating that Th2-type immune reactions underlie TARC production. Serum TARC measurements also have potent diagnostic value for DIHS, with high sensitivity and specificity.
Collapse
Affiliation(s)
| | - Sakae Kaneko
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Yuko Chinuki
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Yohji Suyama
- Department of Laboratory Medicine, Shimane University Hospital, Shimane, Japan
| | - Masataka Ohta
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Hiroyuki Niihara
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Eishin Morita
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan.
| |
Collapse
|
40
|
Lim SJ, Kim M, Randy A, Nam EJ, Nho CW. Effects of Hovenia dulcis Thunb. extract and methyl vanillate on atopic dermatitis-like skin lesions and TNF-α/IFN-γ-induced chemokines production in HaCaT cells. ACTA ACUST UNITED AC 2016; 68:1465-1479. [PMID: 27696405 DOI: 10.1111/jphp.12640] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/24/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Here, we hypothesized that Hovenia dulcis branch extract (HDB) and its active constituents ameliorates 2,4-dinitrochlorobenzene-induced atopic dermatitis (AD)-like skin lesions by modulating the T helper Th1/Th2 balance in NC/Nga mice and TNF-α- and IFN-γ-induced production of thymus and activation-regulated chemokine (TARC) and macrophage-derived chemokine (MDC) in HaCaT cells. METHODS HaCaT cells were stimulated by TNF-α/IFN-γ in the presence of HDB and its constituents. TARC and MDC were measured by ELISA and RT-PCR. For the in-vivo study, oral feeding of HDB was performed for 5 weeks with 2,4-dinitrochlorobenzene (DNCB) treatment every other day. The efficacy of HDB on parameters of DNCB-induced AD was evaluated morphologically, physiologically and immunologically. KEY FINDINGS In-vitro studies showed that HDB and its constituents suppressed TNF-α/IFN-γ-induced production of TARC and MDC in HaCaT cells by inhibiting MAPK signalling. In-vivo studies showed that HDB regulated immunoglobulin (Ig) E and immunoglobulin G2a (IgG2a) levels in serum and the expression of mRNA for Th1- and Th2-related mediators in skin lesions. Histopathological analyses revealed reduced epidermal thickness and reduced infiltration of skin lesions by inflammatory cells. CONCLUSION These results suggest that HDB inhibits AD-like skin diseases by regulating Th1 and Th2 responses in NC/Nga mice and in HaCaT cells.
Collapse
Affiliation(s)
- Sue Ji Lim
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Korea.,Convergence Research Center for Smart Farm Solution, Korea Institute of Science and Technology, Gangneung, Korea
| | - Myungsuk Kim
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Korea.,Convergence Research Center for Smart Farm Solution, Korea Institute of Science and Technology, Gangneung, Korea
| | - Ahmad Randy
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Korea.,Department of Biological Chemistry, Korea University of Science and Technology, Daejeon, Korea
| | - Eui Jeong Nam
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Korea.,Department of Biological Chemistry, Korea University of Science and Technology, Daejeon, Korea
| | - Chu Won Nho
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Korea. .,Convergence Research Center for Smart Farm Solution, Korea Institute of Science and Technology, Gangneung, Korea.
| |
Collapse
|
41
|
Petersen PS, Wolf RM, Lei X, Peterson JM, Wong GW. Immunomodulatory roles of CTRP3 in endotoxemia and metabolic stress. Physiol Rep 2016; 4:4/5/e12735. [PMID: 26997632 PMCID: PMC4823594 DOI: 10.14814/phy2.12735] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
C1q/TNF‐related protein 3 (CTRP3) is a secreted hormone that modulates hepatic glucose and lipid metabolism. Its circulating levels are reduced in human and rodent models of obesity, a metabolic state accompanied by chronic low‐grade inflammation. Recent studies have demonstrated an anti‐inflammatory role for recombinant CTRP3 in attenuating LPS‐induced systemic inflammation, and its deficiency markedly exacerbates inflammation in a mouse model of rheumatoid arthritis. We used genetic mouse models to explore the immunomodulatory function of CTRP3 in response to acute (LPS challenge) and chronic (high‐fat diet) inflammatory stimuli. In a sublethal dose of LPS challenge, neither CTRP3 deficiency nor its overexpression in transgenic mice had an impact on IL‐1β, IL‐6, TNF‐α, or MIP‐2 induction at the serum protein or mRNA levels, contrary to previous findings based on recombinant CTRP3 administration. In a metabolic context, we measured 71 serum cytokine levels in wild‐type and CTRP3 transgenic mice fed a high‐fat diet or a matched control low‐fat diet. On a low‐fat diet, CTRP3 transgenic mice had elevated circulating levels of multiple chemokines (CCL11, CXCL9, CXCL10, CCL17, CX3CL1, CCL22 and sCD30). However, when obesity was induced with a high‐fat diet, CTRP3 transgenic mice had lower circulating levels of IL‐5, TNF‐α, sVEGF2, and sVEGFR3, and a higher level of soluble gp130. Contingent upon the metabolic state, CTRP3 overexpression altered chemokine levels in lean mice, and attenuated systemic inflammation in the setting of obesity and insulin resistance. These results highlight a context‐dependent immunomodulatory role for CTRP3.
Collapse
Affiliation(s)
- Pia S Petersen
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Risa M Wolf
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan M Peterson
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
42
|
Asahina R, Maeda S. A review of the roles of keratinocyte-derived cytokines and chemokines in the pathogenesis of atopic dermatitis in humans and dogs. Vet Dermatol 2016; 28:16-e5. [DOI: 10.1111/vde.12351] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Ryota Asahina
- Department of Veterinary Medicine; Faculty of Applied Biological Sciences; Gifu University; 1-1 Yanagido Gifu 501-1193 Japan
| | - Sadatoshi Maeda
- Department of Veterinary Medicine; Faculty of Applied Biological Sciences; Gifu University; 1-1 Yanagido Gifu 501-1193 Japan
| |
Collapse
|
43
|
Murdaca G, Contini P, Negrini S, Ciprandi G, Puppo F. Immunoregulatory Role of HLA-G in Allergic Diseases. J Immunol Res 2016; 2016:6865758. [PMID: 27413762 PMCID: PMC4931064 DOI: 10.1155/2016/6865758] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/25/2016] [Indexed: 01/17/2023] Open
Abstract
Allergic diseases are sustained by a T-helper 2 polarization leading to interleukin-4 secretion, IgE-dependent inflammation, and mast cell and eosinophil activation. HLA-G molecules, both in membrane-bound and in soluble forms, play a central role in modulation of immune responses. Elevated levels of soluble HLA-G (sHLA-G) molecules are detected in serum of patients with allergic rhinitis to seasonal and perennial allergens and correlate with allergen-specific IgE levels, clinical severity, drug consumption, and response to allergen-specific immunotherapy. sHLA-G molecules are also found in airway epithelium of patients with allergic asthma and high levels of sHLA-G molecules are detectable in plasma and bronchoalveolar lavage of asthmatic patients correlating with allergen-specific IgE levels. Finally, HLA-G molecules are expressed by T cells, monocytes-macrophages, and Langerhans cells infiltrating the dermis of atopic dermatitis patients. Collectively, although at present it is difficult to completely define the role of HLA-G molecules in allergic diseases, it may be suggested that they are expressed and secreted by immune cells during the allergic reaction in an attempt to suppress allergic inflammation.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy
| | - Paola Contini
- Department of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy
| | - Simone Negrini
- Department of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy
| | - Giorgio Ciprandi
- Department of Internal Medicine, Respiratory and Allergy Diseases Unit, University of Genova, 16132 Genova, Italy
| | - Francesco Puppo
- Department of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy
| |
Collapse
|
44
|
Zhao X, Delgado L, Weiner R, Laterza OF. Influence of Pre-Analytical Factors on Thymus- and Activation-Regulated Chemokine Quantitation in Plasma. J Circ Biomark 2015; 4:10. [PMID: 28936246 PMCID: PMC5572979 DOI: 10.5772/61749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 10/01/2015] [Indexed: 01/24/2023] Open
Abstract
Thymus- and activation-regulated chemokine (TARC) in serum/plasma associates with the disease activity of atopic dermatitis (AD), and is a promising tool for assessing the response to the treatment of the disease. TARC also exists within platelets, with elevated levels detectable in AD patients. We examined the effects of pre-analytical factors on the quantitation of TARC in human EDTA plasma. TARC levels in platelet-free plasma were significantly lower than those in platelet-containing plasma. After freeze-thaw, TARC levels increased in platelet-containing plasma, but remained unchanged in platelet-free plasma, suggesting TARC was released from the platelets during the freeze-thaw process. In contrast, TARC levels were stable in serum independent of freeze-thaw. These findings underscore the importance of pre-analytical factors to TARC quantitation. Plasma TARC levels should be measured in platelet-free plasma for accurate quantitation. Pre-analytical factors influence the quantitation, interpretation, and implementation of circulating TARC as a biomarker for the development of AD therapeutics.
Collapse
Affiliation(s)
- Xuemei Zhao
- Translational Molecular Biomarkers, Merck Research Laboratories, Rahway, NJ, USA
| | - Liliana Delgado
- Translational Molecular Biomarkers, Merck Research Laboratories, Rahway, NJ, USA
| | - Russell Weiner
- Translational Molecular Biomarkers, Merck Research Laboratories, Rahway, NJ, USA.,Translational Sciences, Biomarkers & Companion Diagnostics, Daiichi Sankyo, Inc., Edison, NJ, USA
| | - Omar F Laterza
- Translational Molecular Biomarkers, Merck Research Laboratories, Rahway, NJ, USA
| |
Collapse
|
45
|
Bogaczewicz J, Malinowska K, Sysa-Jedrzejowska A, Wozniacka A. Medium-dose ultraviolet A1 phototherapy and mRNA expression of TSLP, TARC, IL-5, and IL-13 in acute skin lesions in atopic dermatitis. Int J Dermatol 2015; 55:856-63. [PMID: 26475182 DOI: 10.1111/ijd.12992] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 11/18/2014] [Accepted: 01/25/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND The mechanisms responsible for the efficacy of ultraviolet A1 (UVA1) in the treatment of atopic dermatitis (AD) are not fully understood. OBJECTIVES This study was designed to investigate mRNA expression of thymic stromal lymphopoietin (TSLP), thymus- and activation-regulated chemokine (TARC), interleukin-5 (IL-5), and IL-13 in AD before and after UVA1 therapy, to determine correlations among them, and to examine whether UVA1 influences their expression and whether it is associated with UVA1 efficacy. METHODS Twenty-five patients with AD underwent medium-dose UVA1 phototherapy. Before and after UVA1, biopsies from acute skin lesions were studied using reverse transcription and real-time polymerase chain reaction. RESULTS Levels of mRNA TSLP correlated with those of TARC, IL-5, and IL-13, and levels of TARC correlated with those of IL-5 and IL-13, both before and after UVA1. Expression of IL-5 correlated with that of IL-13 only before UVA1. SCORAD (SCORing of Atopic Dermatitis) indices correlated with levels of TARC and IL-5 before irradiation. After UVA1, no mRNA level correlated with the SCORAD index. Phototherapy with UVA1 improved SCORAD values (P < 0.001) and increased expression of TARC (P < 0.05) but did not affect mRNA expression of TSLP, IL-5, or IL-13. CONCLUSIONS Expression levels of the mediators TSLP, TARC, IL-5, and IL-13 in AD are interrelated. Phototherapy with UVA1 improves SCORAD indices and increases expression of TARC but has no direct effects on the expression of other molecules. It is likely that UVA1 also interferes with or acts via intermediators on the link between IL-5 and IL-13.
Collapse
Affiliation(s)
- Jaroslaw Bogaczewicz
- Department of Dermatology and Venereology, Medical University of Lodz, Lodz, Poland
| | - Karolina Malinowska
- Department of Dermatology and Venereology, Medical University of Lodz, Lodz, Poland
| | | | - Anna Wozniacka
- Department of Dermatology and Venereology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
46
|
Solari R, Pease JE. Targeting chemokine receptors in disease--a case study of CCR4. Eur J Pharmacol 2015; 763:169-77. [PMID: 25981299 PMCID: PMC4784718 DOI: 10.1016/j.ejphar.2015.05.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/17/2015] [Accepted: 05/12/2015] [Indexed: 01/14/2023]
Abstract
Since their early 1990s, the chemokine receptor family of G protein-coupled receptors (GPCRs) has been the source of much pharmacological endeavour. Best known for their key roles in recruiting leukocytes to sites of infection and inflammation, the receptors present themselves as plausible drug targets for therapeutic intervention. In this article, we will focus our attention upon CC Chemokine Receptor Four (CCR4) which has been implicated in diseases as diverse as allergic asthma and lymphoma. We will review the discovery of the receptors and their ligands, their perceived roles in disease and the successful targeting of CCR4 by both small molecule antagonists and monoclonal antibodies. We will also discuss future directions and strategies for drug discovery in this field.
Collapse
Affiliation(s)
- Roberto Solari
- Airway Disease Infection Section, MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom
| | - James E Pease
- Leukocyte Biology Section, MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
47
|
Kataoka Y. Thymus and activation-regulated chemokine as a clinical biomarker in atopic dermatitis. J Dermatol 2015; 41:221-9. [PMID: 24628072 DOI: 10.1111/1346-8138.12440] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 11/28/2022]
Abstract
Thymus and activation-regulated chemokine (TARC/CCL17) is a member of the T-helper 2 chemokine family. In Japan, serum TARC level has been commercially measured since 2008. After years of experience, we realized that TARC is an extremely useful clinical biomarker for atopic dermatitis (AD) treatment. Usually, physicians conduct a visual examination to determine whether their treatment has been successful; however, the visual examination results may not always be accurate; in such cases, serum TARC levels should be measured to eliminate any ambiguity regarding the treatment outcome. When the waning and waxing of eczema and fluctuations in the serum TARC levels were considered, we frequently found that AD does not follow a natural course but follows non-regulated inflammatory floating caused by insufficient intermittent topical treatment. Serum TARC is a promising biomarker for remission and can be used for accurately monitoring proactive treatment for long-term control. Abnormally high serum TARC levels indicate accelerated pathogenesis of cutaneous inflammation. Rapid normalization and maintaining normal serum TARC levels using appropriate topical treatment is a reasonable strategy for alleviating inflammation without upregulating cytokine expression. Observing serum TARC levels during early intervention for severe infantile AD is worthwhile to determine initial disease activity and evaluate treatment efficacy. Appropriate control of severe early-onset infantile AD is important for improving prognosis of eczema and for preventing food allergies. Additionally, this biomarker is useful for improving patient adherence. Dermatologists will be able to make great progress in treating AD by adopting biomarkers such as TARC for accurately assessing non-visible subclinical disorders.
Collapse
Affiliation(s)
- Yoko Kataoka
- Dermatology, Osaka Prefectural Medical Center for Respiratory and Allergic Diseases, Habikino, Osaka, Japan
| |
Collapse
|
48
|
An ultra-sensitive clinical biomarker assay: quantitation of thymus and activation-regulated chemokine in human plasma. Bioanalysis 2015; 6:1069-80. [PMID: 24830891 DOI: 10.4155/bio.14.72] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Thymus and activation-regulated chemokine (TARC) is a Th2 type, pro-allergic secreted chemokine. TARC in plasma/serum has been proposed as a marker for disease activity of atopic dermatitis (AD) and as a pharmacodynamic readout in the clinical development of novel agents for the treatment of AD. RESULTS An ultra-sensitive electrochemiluminescence assay for TARC in human plasma was developed and analytically validated. The assay demonstrated excellent performance characteristics, including precision, sensitivity, dilution linearity, accuracy and specificity. Stability and biological variability of TARC in plasma were also assessed for clinical sample analysis and data interpretation. CONCLUSION The improved sensitivity allowed the measurement of approximately 90% TARC inhibition from baseline levels of healthy subjects and >90% TARC inhibition from baseline levels of AD patients after drug treatment. A validated TARC electrochemiluminescence assay enables pharmacodynamic assessment in the development of AD therapeutics.
Collapse
|
49
|
|
50
|
Hagemann UB, Gunnarsson L, Géraudie S, Scheffler U, Griep RA, Reiersen H, Duncan AR, Kiprijanov SM. Fully human antagonistic antibodies against CCR4 potently inhibit cell signaling and chemotaxis. PLoS One 2014; 9:e103776. [PMID: 25080123 PMCID: PMC4117600 DOI: 10.1371/journal.pone.0103776] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 06/30/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND CC chemokine receptor 4 (CCR4) represents a potentially important target for cancer immunotherapy due to its expression on tumor infiltrating immune cells including regulatory T cells (Tregs) and on tumor cells in several cancer types and its role in metastasis. METHODOLOGY Using phage display, human antibody library, affinity maturation and a cell-based antibody selection strategy, the antibody variants against human CCR4 were generated. These antibodies effectively competed with ligand binding, were able to block ligand-induced signaling and cell migration, and demonstrated efficient killing of CCR4-positive tumor cells via ADCC and phagocytosis. In a mouse model of human T-cell lymphoma, significant survival benefit was demonstrated for animals treated with the newly selected anti-CCR4 antibodies. SIGNIFICANCE For the first time, successful generation of anti- G-protein coupled chemokine receptor (GPCR) antibodies using human non-immune library and phage display on GPCR-expressing cells was demonstrated. The generated anti-CCR4 antibodies possess a dual mode of action (inhibition of ligand-induced signaling and antibody-directed tumor cell killing). The data demonstrate that the anti-tumor activity in vivo is mediated, at least in part, through Fc-receptor dependent effector mechanisms, such as ADCC and phagocytosis. Anti-CC chemokine receptor 4 antibodies inhibiting receptor signaling have potential as immunomodulatory antibodies for cancer.
Collapse
|