1
|
Dou Y, Niu Y, Shen H, Wang L, Lv Y, Liu S, Xie X, Feng A, Liu X. Identification of disease-specific gut microbial markers in vitiligo. Front Microbiol 2025; 16:1499035. [PMID: 39967732 PMCID: PMC11833150 DOI: 10.3389/fmicb.2025.1499035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
There is a potential correlation between vitiligo and gut microbiota, although research in this area is currently limited. The research employed high-throughput sequencing of 16S rRNA to examine the gut microbiome in the stool samples of 49 individuals with vitiligo and 49 without the condition. The study encompassed four comparison groups: (1) DI (disease) group vs. HC (healthy control) group; (2) DI_m group (disease group of minors) vs. HC_m group (healthy control group of minors); (3) DI_a group (adult disease group) vs. HC_a group (adult healthy control group); (4) DI_m group vs. DI_a group. Research findings have indicated the presence of spatial heterogeneity in the gut microbiota composition between individuals with vitiligo and healthy controls. A significant reduction in gut microbiota diversity has been observed in vitiligo patients across both minors and adult groups. However, variations have been noted in the composition of disease-related differential microbial markers among different age groups. Specifically, Bacteroides and Parabacteroides have been identified as specific markers of the intestinal microbiota of vitiligo patients in both minor and adult groups. Correlative analyses have revealed a positive correlation of these two genera with the Vitiligo Area Scoring Index (VASI) and disease duration. It is noteworthy that there are no significant differences in diversity between the DI_m group and the DI_a group, with similarities in microbiota composition and functional characteristics. Nevertheless, correlative analyses suggest a declining trend in Bacteroides and Parabacteroides with increasing age. Individuals with vitiligo exhibit distinct features in their gut microbiome when contrasted with those in the healthy control group. Additionally, the microbial marker genera that show variances between patients and healthy controls vary among different age groups. Disease-specific microbial marker genera (Bacteroides and Parabacteroides) are associated with VASI, duration of the condition, and age. These findings are essential for improving early diagnosis and developing potential treatment strategies for individuals with vitiligo.
Collapse
Affiliation(s)
- Yimin Dou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Niu
- Department of Gastroenterology Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hexiao Shen
- School of Life Science, Hubei University, Wuhan, China
| | - Lan Wang
- School of Life Science, Hubei University, Wuhan, China
| | - Yongling Lv
- School of Life Science, Hubei University, Wuhan, China
| | - Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiafei Xie
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aiping Feng
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinxin Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Morales-Primo AU, Becker I, Pedraza-Zamora CP, Zamora-Chimal J. Th17 Cell and Inflammatory Infiltrate Interactions in Cutaneous Leishmaniasis: Unraveling Immunopathogenic Mechanisms. Immune Netw 2024; 24:e14. [PMID: 38725676 PMCID: PMC11076297 DOI: 10.4110/in.2024.24.e14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 05/12/2024] Open
Abstract
The inflammatory response during cutaneous leishmaniasis (CL) involves immune and non-immune cell cooperation to contain and eliminate Leishmania parasites. The orchestration of these responses is coordinated primarily by CD4+ T cells; however, the disease outcome depends on the Th cell predominant phenotype. Although Th1 and Th2 phenotypes are the most addressed as steers for the resolution or perpetuation of the disease, Th17 cell activities, especially IL-17 release, are recognized to be vital during CL development. Th17 cells perform vital functions during both acute and chronic phases of CL. Overall, Th17 cells induce the migration of phagocytes (neutrophils, macrophages) to the infection site and CD8+ T cells and NK cell activation. They also provoke granzyme and perforin secretion from CD8+ T cells, macrophage differentiation towards an M2 phenotype, and expansion of B and Treg cells. Likewise, immune cells from the inflammatory infiltrate have modulatory activities over Th17 cells involving their differentiation from naive CD4+ T cells and further expansion by generating a microenvironment rich in optimal cytokines such as IL-1β, TGF-β, IL-6, and IL-21. Th17 cell activities and synergies are crucial for the resistance of the infection during the early and acute stages; however, if unchecked, Th17 cells might lead to a chronic stage. This review discusses the synergies between Th17 cells and the inflammatory infiltrate and how these interactions might destine the course of CL.
Collapse
Affiliation(s)
- Abraham U. Morales-Primo
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Ingeborg Becker
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Claudia Patricia Pedraza-Zamora
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, México
| | - Jaime Zamora-Chimal
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| |
Collapse
|
3
|
Miles MA, Liong S, Liong F, Coward-Smith M, Trollope GS, Oseghale O, Erlich JR, Brooks RD, Logan JM, Hickey S, Wang H, Bozinovski S, O’Leary JJ, Brooks DA, Selemidis S. TLR7 promotes chronic airway disease in RSV-infected mice. Front Immunol 2023; 14:1240552. [PMID: 37795093 PMCID: PMC10545951 DOI: 10.3389/fimmu.2023.1240552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) commonly infects the upper respiratory tract (URT) of humans, manifesting with mild cold or flu-like symptoms. However, in infants and the elderly, severe disease of the lower respiratory tract (LRT) often occurs and can develop into chronic airway disease. A better understanding of how an acute RSV infection transitions to a LRT chronic inflammatory disease is critically important to improve patient care and long-term health outcomes. To model acute and chronic phases of the disease, we infected wild-type C57BL/6 and toll-like receptor 7 knockout (TLR7 KO) mice with RSV and temporally assessed nasal, airway and lung inflammation for up to 42 days post-infection. We show that TLR7 reduced viral titers in the URT during acute infection but promoted pronounced pathogenic and chronic airway inflammation and hyperreactivity in the LRT. This study defines a hitherto unappreciated molecular mechanism of lower respiratory pathogenesis to RSV, highlighting the potential of TLR7 modulation to constrain RSV pathology to the URT.
Collapse
Affiliation(s)
- Mark A. Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Felicia Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Madison Coward-Smith
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Gemma S. Trollope
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Osezua Oseghale
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Jonathan R. Erlich
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Robert D. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jessica M. Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Shane Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hao Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Steven Bozinovski
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, Dublin, Ireland
| | - Doug A. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
4
|
Hinshaw DC, Benavides GA, Metge BJ, Swain CA, Kammerud SC, Alsheikh HA, Elhamamsy A, Chen D, Darley-Usmar V, Rathmell JC, Welner RS, Samant RS, Shevde LA. Hedgehog Signaling Regulates Treg to Th17 Conversion Through Metabolic Rewiring in Breast Cancer. Cancer Immunol Res 2023; 11:687-702. [PMID: 37058110 PMCID: PMC10159910 DOI: 10.1158/2326-6066.cir-22-0426] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/18/2022] [Accepted: 02/28/2023] [Indexed: 04/15/2023]
Abstract
The tumor immune microenvironment dynamically evolves to support tumor growth and progression. Immunosuppressive regulatory T cells (Treg) promote tumor growth and metastatic seeding in patients with breast cancer. Deregulation of plasticity between Treg and Th17 cells creates an immune regulatory framework that enables tumor progression. Here, we discovered a functional role for Hedgehog (Hh) signaling in promoting Treg differentiation and immunosuppressive activity, and when Hh activity was inhibited, Tregs adopted a Th17-like phenotype complemented by an enhanced inflammatory profile. Mechanistically, Hh signaling promoted O-GlcNAc modifications of critical Treg and Th17 transcription factors, Foxp3 and STAT3, respectively, that orchestrated this transition. Blocking Hh reprogramed Tregs metabolically, dampened their immunosuppressive activity, and supported their transdifferentiation into inflammatory Th17 cells that enhanced the recruitment of cytotoxic CD8+ T cells into tumors. Our results demonstrate a previously unknown role for Hh signaling in the regulation of Treg differentiation and activity and the switch between Tregs and Th17 cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Dominique C. Hinshaw
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gloria A. Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brandon J. Metge
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney A. Swain
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah C. Kammerud
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Heba A. Alsheikh
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amr Elhamamsy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongquan Chen
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, VUMC, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert S. Welner
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S. Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham VA Medical Center, Birmingham, AL, USA
| | - Lalita A. Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Senior author
| |
Collapse
|
5
|
Bieber K, Hundt JE, Yu X, Ehlers M, Petersen F, Karsten CM, Köhl J, Kridin K, Kalies K, Kasprick A, Goletz S, Humrich JY, Manz RA, Künstner A, Hammers CM, Akbarzadeh R, Busch H, Sadik CD, Lange T, Grasshoff H, Hackel AM, Erdmann J, König I, Raasch W, Becker M, Kerstein-Stähle A, Lamprecht P, Riemekasten G, Schmidt E, Ludwig RJ. Autoimmune pre-disease. Autoimmun Rev 2023; 22:103236. [PMID: 36436750 DOI: 10.1016/j.autrev.2022.103236] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022]
Abstract
Approximately 5% of the world-wide population is affected by autoimmune diseases. Overall, autoimmune diseases are still difficult to treat, impose a high burden on patients, and have a significant economic impact. Like other complex diseases, e.g., cancer, autoimmune diseases develop over several years. Decisive steps in the development of autoimmune diseases are (i) the development of autoantigen-specific lymphocytes and (often) autoantibodies and (ii) potentially clinical disease manifestation at a later stage. However, not all healthy individuals with autoantibodies develop disease manifestations. Identifying autoantibody-positive healthy individuals and monitoring and inhibiting their switch to inflammatory autoimmune disease conditions are currently in their infancy. The switch from harmless to inflammatory autoantigen-specific T and B-cell and autoantibody responses seems to be the hallmark for the decisive factor in inflammatory autoimmune disease conditions. Accordingly, biomarkers allowing us to predict this progression would have a significant impact. Several factors, such as genetics and the environment, especially diet, smoking, exposure to pollutants, infections, stress, and shift work, might influence the progression from harmless to inflammatory autoimmune conditions. To inspire research directed at defining and ultimately targeting autoimmune predisease, here, we review published evidence underlying the progression from health to autoimmune predisease and ultimately to clinically manifest inflammatory autoimmune disease, addressing the following 3 questions: (i) what is the current status, (ii) what is missing, (iii) and what are the future perspectives for defining and modulating autoimmune predisease.
Collapse
Affiliation(s)
- Katja Bieber
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Jennifer E Hundt
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Marc Ehlers
- Institute of Nutritional Medicine, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Khalaf Kridin
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; Unit of Dermatology and Skin Research Laboratory, Baruch Padeh Medical Center, Poriya, Israel
| | - Kathrin Kalies
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Anika Kasprick
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Stephanie Goletz
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Jens Y Humrich
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Axel Künstner
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Christoph M Hammers
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Reza Akbarzadeh
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | | | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Alexander M Hackel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Inke König
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Mareike Becker
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Anja Kerstein-Stähle
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany; Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany.
| |
Collapse
|
6
|
Grandi A, Ferrini E, Mecozzi L, Ciccimarra R, Zoboli M, Leo L, Khalajzeyqami Z, Kleinjan A, Löwik CWGM, Donofrio G, Villetti G, Stellari FF. Indocyanine-enhanced mouse model of bleomycin-induced lung fibrosis with hallmarks of progressive emphysema. Am J Physiol Lung Cell Mol Physiol 2023; 324:L211-L227. [PMID: 36625471 PMCID: PMC9925167 DOI: 10.1152/ajplung.00180.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The development of new drugs for idiopathic pulmonary fibrosis strongly relies on preclinical experimentation, which requires the continuous improvement of animal models and integration with in vivo imaging data. Here, we investigated the lung distribution of bleomycin (BLM) associated with the indocyanine green (ICG) dye by fluorescence imaging. A long-lasting lung retention (up to 21 days) was observed upon oropharyngeal aspiration (OA) of either ICG or BLM + ICG, with significantly more severe pulmonary fibrosis, accompanied by the progressive appearance of emphysema-like features, uniquely associated with the latter combination. More severe and persistent lung fibrosis, together with a progressive air space enlargement uniquely associated with the BLM + ICG group, was confirmed by longitudinal micro-computed tomography (CT) and histological analyses. Multiple inflammation and fibrosis biomarkers were found to be increased in the bronchoalveolar lavage fluid of BLM- and BLM + ICG-treated animals, but with a clear trend toward a much stronger increase in the latter group. Similarly, in vitro assays performed on macrophage and epithelial cell lines revealed a significantly more marked cytotoxicity in the case of BLM + ICG-treated mice. Also unique to this group was the synergistic upregulation of apoptotic markers both in lung sections and cell lines. Although the exact mechanism underlying the more intense lung fibrosis phenotype with emphysema-like features induced by BLM + ICG remains to be elucidated, we believe that this combination treatment, whose overall effects more closely resemble the human disease, represents a valuable alternative model for studying fibrosis development and for the identification of new antifibrotic compounds.
Collapse
Affiliation(s)
- Andrea Grandi
- 1Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy
| | - Erica Ferrini
- 2Department of Veterinary Science, University of Parma, Parma, Italy
| | - Laura Mecozzi
- 3Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Matteo Zoboli
- 2Department of Veterinary Science, University of Parma, Parma, Italy
| | - Ludovica Leo
- 3Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Zahra Khalajzeyqami
- 4Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Alex Kleinjan
- 5Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Clemens W. G. M. Löwik
- 6Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gaetano Donofrio
- 2Department of Veterinary Science, University of Parma, Parma, Italy
| | - Gino Villetti
- 1Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy
| | | |
Collapse
|
7
|
Easwaran M, Martinez JD, Kim JB, Erickson-DiRenzo E. Modulation of mouse laryngeal inflammatory and immune cell responses by low and high doses of mainstream cigarette smoke. Sci Rep 2022; 12:18667. [PMID: 36333510 PMCID: PMC9636197 DOI: 10.1038/s41598-022-23359-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoking is a major risk factor for laryngeal diseases. Despite well-documented cigarette smoke (CS) induced laryngeal histopathological changes, the underlying immunopathological mechanisms remain largely unexplored. The goal of this study was to evaluate inflammatory and immune cell responses in a CS-exposed larynx. Specifically, we used a 4-week subacute whole-body CS inhalation mouse model to assess these responses in the laryngeal mucosa upon exposure to low (LD; 1 h/day) and high dose (HD; 4 h/day) CS. Laryngeal tissues were harvested and evaluated using a 254-plex NanoString inflammation panel and neutrophil/macrophage/T-cell immunohistochemistry (IHC). NanoString global and differential gene expression analysis revealed a unique expression profile only in the HD group, with 26 significant differentially expressed genes (DEGs). StringDB KEGG pathway enrichment analysis revealed the involvement of these DEGs with pro-inflammatory pathways including TNF/TNFα and IL-17. Furthermore, inflammatory responses remained inhibited in conjunction with predicted activated states of anti-inflammatory regulators like PPARγ and NFE2L2 upon Ingenuity Pathway Analysis (IPA). Subglottic T-cell levels remained significantly inhibited as corroborated by IPA predictions. Overall, our key findings are consistent with HD exposures being anti-inflammatory and immunosuppressive. Furthermore, the identification of important regulatory genes and enriched pathways may help improve clinical interventions for CS-induced laryngeal diseases.
Collapse
Affiliation(s)
- Meena Easwaran
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua D Martinez
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Juyong Brian Kim
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth Erickson-DiRenzo
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
8
|
Taucher E, Mykoliuk I, Lindenmann J, Smolle-Juettner FM. Implications of the Immune Landscape in COPD and Lung Cancer: Smoking Versus Other Causes. Front Immunol 2022; 13:846605. [PMID: 35386685 PMCID: PMC8978964 DOI: 10.3389/fimmu.2022.846605] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking is reported in about one third of adults worldwide. A strong relationship between cigarette smoke exposure and chronic obstructive pulmonary disease (COPD) as well as lung cancer has been proven. However, about 15% of lung cancer cases, and between one fourth and one third of COPD cases, occur in never-smokers. The effects of cigarette smoke on the innate as well as the adaptive immune system have been widely investigated. It is assumed that certain immunologic features contribute to lung cancer and COPD development in the absence of smoking as the major risk factor. In this article, we review different immunological aspects of lung cancer and COPD with a special focus on non-smoking related risk factors.
Collapse
Affiliation(s)
- Elisabeth Taucher
- Division of Pulmonology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Iurii Mykoliuk
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | | |
Collapse
|
9
|
Alexandrova Y, Costiniuk CT, Jenabian MA. Pulmonary Immune Dysregulation and Viral Persistence During HIV Infection. Front Immunol 2022; 12:808722. [PMID: 35058937 PMCID: PMC8764194 DOI: 10.3389/fimmu.2021.808722] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the success of antiretroviral therapy (ART), people living with HIV continue to suffer from high burdens of respiratory infections, lung cancers and chronic lung disease at a higher rate than the general population. The lung mucosa, a previously neglected HIV reservoir site, is of particular importance in this phenomenon. Because ART does not eliminate the virus, residual levels of HIV that remain in deep tissues lead to chronic immune activation and pulmonary inflammatory pathologies. In turn, continuous pulmonary and systemic inflammation cause immune cell exhaustion and pulmonary immune dysregulation, creating a pro-inflammatory environment ideal for HIV reservoir persistence. Moreover, smoking, gut and lung dysbiosis and co-infections further fuel the vicious cycle of residual viral replication which, in turn, contributes to inflammation and immune cell proliferation, further maintaining the HIV reservoir. Herein, we discuss the recent evidence supporting the notion that the lungs serve as an HIV viral reservoir. We will explore how smoking, changes in the microbiome, and common co-infections seen in PLWH contribute to HIV persistence, pulmonary immune dysregulation, and high rates of infectious and non-infectious lung disease among these individuals.
Collapse
Affiliation(s)
- Yulia Alexandrova
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC, Canada
| |
Collapse
|
10
|
Lourenço JD, Ito JT, Martins MDA, Tibério IDFLC, Lopes FDTQDS. Th17/Treg Imbalance in Chronic Obstructive Pulmonary Disease: Clinical and Experimental Evidence. Front Immunol 2021; 12:804919. [PMID: 34956243 PMCID: PMC8695876 DOI: 10.3389/fimmu.2021.804919] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
The imbalance between pro- and anti-inflammatory immune responses mediated by Th17 and Treg cells is deeply involved in the development and progression of inflammation in chronic obstructive pulmonary disease (COPD). Several clinical and experimental studies have described the Th17/Treg imbalance in COPD progression. Due to its importance, many studies have also evaluated the effect of different treatments targeting Th17/Treg cells. However, discrepant results have been observed among different lung compartments, different COPD stages or local and systemic markers. Thus, the data must be carefully examined. In this context, this review explores and summarizes the recent outcomes of Th17/Treg imbalance in COPD development and progression in clinical, experimental and in vitro studies.
Collapse
Affiliation(s)
- Juliana Dias Lourenço
- Laboratory of Experimental Therapeutics (LIM-20), Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Juliana Tiyaki Ito
- Laboratory of Experimental Therapeutics (LIM-20), Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Milton de Arruda Martins
- Laboratory of Experimental Therapeutics (LIM-20), Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | | | | |
Collapse
|
11
|
Kim DH, Kim HY, Lee WW. Induction of Unique STAT Heterodimers by IL-21 Provokes IL-1RI Expression on CD8 + T Cells, Resulting in Enhanced IL-1β Dependent Effector Function. Immune Netw 2021; 21:e33. [PMID: 34796037 PMCID: PMC8568912 DOI: 10.4110/in.2021.21.e33] [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: 05/25/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
IL-1β plays critical roles in the priming and effector phases of immune responses such as the differentiation, commitment, and memory formation of T cells. In this context, several reports have suggested that the IL-1β signal is crucial for CTL-mediated immune responses to viral infections and tumors. However, little is known regarding whether IL-1β acts directly on CD8+ T cells and what the molecular mechanisms underlying expression of IL-1 receptors (IL-1Rs) on CD8+ T cells and features of IL-1R+CD8+ T cells are. Here, we provide evidence that the expression of IL-1R type I (IL-1RI), the functional receptor of IL-1β, is preferentially induced by IL-21 on TCR-stimulated CD8+ T cells. Further, IL-1β enhances the effector function of CD8+ T cells expressing IL-21-induced IL-1RI by increasing cytokine production and release of cytotoxic granules containing granzyme B. The IL-21-IL-1RI-IL-1β axis is involved in an augmented effector function through regulation of transcription factors BATF, Blimp-1, and IRF4. Moreover, this axis confers a unique effector function to CD8+ T cells compared to conventional type 1 cytotoxic T cells differentiated with IL-12. Chemical inhibitor and immunoprecipitation assay demonstrated that IL-21 induces a unique pattern of STAT activation with the formation of both STAT1:STAT3 and STAT3:STAT5 heterodimers, which are critical for the induction of IL-1RI on TCR-stimulated CD8+ T cells. Taken together, we propose that induction of a novel subset of IL-1RI-expressing CD8+ T cells by IL-21 may be beneficial to the protective immune response against viral infections and is therefore important to consider for vaccine design.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hee Young Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul 03080, Korea
| |
Collapse
|
12
|
Bovée DM, Cuevas CA, Zietse R, Danser AHJ, Mirabito Colafella KM, Hoorn EJ. Salt-sensitive hypertension in chronic kidney disease: distal tubular mechanisms. Am J Physiol Renal Physiol 2020; 319:F729-F745. [DOI: 10.1152/ajprenal.00407.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) causes salt-sensitive hypertension that is often resistant to treatment and contributes to the progression of kidney injury and cardiovascular disease. A better understanding of the mechanisms contributing to salt-sensitive hypertension in CKD is essential to improve these outcomes. This review critically explores these mechanisms by focusing on how CKD affects distal nephron Na+ reabsorption. CKD causes glomerulotubular imbalance with reduced proximal Na+ reabsorption and increased distal Na+ delivery and reabsorption. Aldosterone secretion further contributes to distal Na+ reabsorption in CKD and is not only mediated by renin and K+ but also by metabolic acidosis, endothelin-1, and vasopressin. CKD also activates the intrarenal renin-angiotensin system, generating intratubular angiotensin II to promote distal Na+ reabsorption. High dietary Na+ intake in CKD contributes to Na+ retention by aldosterone-independent activation of the mineralocorticoid receptor mediated through Rac1. High dietary Na+ also produces an inflammatory response mediated by T helper 17 cells and cytokines increasing distal Na+ transport. CKD is often accompanied by proteinuria, which contains plasmin capable of activating the epithelial Na+ channel. Thus, CKD causes both local and systemic changes that together promote distal nephron Na+ reabsorption and salt-sensitive hypertension. Future studies should address remaining knowledge gaps, including the relative contribution of each mechanism, the influence of sex, differences between stages and etiologies of CKD, and the clinical relevance of experimentally identified mechanisms. Several pathways offer opportunities for intervention, including with dietary Na+ reduction, distal diuretics, renin-angiotensin system inhibitors, mineralocorticoid receptor antagonists, and K+ or H+ binders.
Collapse
Affiliation(s)
- Dominique M. Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catharina A. Cuevas
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katrina M. Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
13
|
An integrative microenvironment approach for follicular lymphoma: roles of inflammatory cell subsets and immune-response polymorphisms on disease clinical course. Oncotarget 2020; 11:3153-3173. [PMID: 32913559 PMCID: PMC7443366 DOI: 10.18632/oncotarget.27698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/14/2020] [Indexed: 01/16/2023] Open
Abstract
The study of the tumor microenvironment (TME) in follicular lymphoma (FL) has produced conflicting results due to assessment of limited TME subpopulations, and because of heterogeneous treatments among different cohorts. Also, important genetic determinants of immune response, such as single-nucleotide polymorphisms (SNPs), remain underexplored in this disease. We performed a detailed study of the TME in 169 FL biopsies using immunohistochemistry, encompassing lymphocytes, macrophages, and cytokines. We also genotyped 16 SNPs within key immune-response genes (IL12A, IL2, IL10, TGFB1, TGFBR1, TGFBR2, IL17A, and IL17F) in 159 patients. We tested associations between SNPs, clinicopathological features and TME composition, and proposed survival models in R-CHOP/R-CVP-treated patients. Presence of the IL12A rs568408 "A" allele associated with the follicular pattern of FOXP3+ cells. The IL12A AA haplotype included rs583911 and rs568408 and was an independent predictor of worse survival, together with the follicular patterns of T-cells (FOXP3+ and CD8+) and high IL-17F tumor levels. The patterns of CD3+, CD4+ and CD8+ cells, displayed as a principal component, also associated with survival. Hierarchical clustering of the TME proteins demonstrated a cluster that was associated with worse prognosis (tumors enriched in IL-17A, IL-17F, CD8, PD1, and Ki-67). The survival of FL patients who were treated in the rituximab era shows a strong dependence on TME signals, especially the T-cell infiltration patterns and IL-17F tumor levels. The presence of the AA haplotype of IL12A in the genome of FL patients is an additional prognostic factor that may modulate the composition of T-reg cells in this disease.
Collapse
|
14
|
Pawlik A, Anisiewicz A, Filip-Psurska B, Klopotowska D, Maciejewska M, Mazur A, Wietrzyk J. Divergent Effect of Tacalcitol (PRI-2191) on Th17 Cells in 4T1 Tumor Bearing Young and Old Ovariectomized Mice. Aging Dis 2020; 11:241-253. [PMID: 32257539 PMCID: PMC7069462 DOI: 10.14336/ad.2019.0618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022] Open
Abstract
Vitamin D and its analogs are known for their role in the development of breast cancer and in immunomodulation. Our previous studies have shown the pro-metastatic effect of calcitriol and tacalcitol (PRI-2191) in young mice bearing 4T1 breast cancer and the anti-metastatic effect in aged ovariectomized (OVX) mice. Therefore, the aim of our work was to characterize Th17 cell population in young and aged OVX mice bearing 4T1 tumors treated with calcitriol and PRI-2191. The expression of genes typical for Th17 cells was examined in splenocytes, as well as splenocytes differentiated with IL-6 and TGF-β to Th17 cells (iTh17). Expression of genes encoding vitamin D receptor (Vdr) and osteopontin (Spp1) as well as the secretion of IL-17A were evaluated in iTh17 cells. PRI-2191 treatment increased the expression of Rora and Rorc transcription factors, Il17a, Il17re and Il21 in iTh17 cells from young mice. In aged OVX mice this effect was not observed. Increased expression was observed in the case of Vdr and Spp1 genes in iTh17 cells from young mice treated with PRI-2191. What is more, in young mice treated with PRI-2191 the secretion of IL-17A to the culture media by iTh17 cells was increased, whereas in aged OVX mice a significant decrease was noted. Increased expression of Spp1 in young mice treated with PRI-2191 may enhance the differentiation of Th17 cells.
Collapse
Affiliation(s)
- Agata Pawlik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dagmara Klopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Magdalena Maciejewska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Andrzej Mazur
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
15
|
Zhang H, Zhou X, Chen X, Lin Y, Qiu S, Zhao Y, Tang Q, Liang Y, Zhong X. Rapamycin attenuates Tc1 and Tc17 cell responses in cigarette smoke-induced emphysema in mice. Inflamm Res 2019; 68:957-968. [PMID: 31468083 DOI: 10.1007/s00011-019-01278-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/02/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE AND DESIGN Chronic exposure to cigarette smoke promotes airway inflammation and emphysema accompanied by enhanced CD8+ interferon (IFN)-γ+ T(Tc1) and CD8+ interleukin (IL)-17+ T(Tc17) cell responses. The mammalian target of rapamycin (mTOR) has been involved in the pathogenesis of emphysema. Inhibiting mTOR by rapamycin has been reported to alleviate emphysema, but the mechanism is not fully understood. We aimed to explore the effect of rapamycin on Tc1 and Tc17 cell responses induced by cigarette smoke exposure. MATERIALS Male C57BL/6 mice were exposed to cigarette smoke or room air for 24 weeks. Half of the smoke-exposed mice received rapamycin in the last 12 weeks. The severity of emphysema in those mice was evaluated by mean linear intercept (MLI), mean alveolar airspace area (MAA) and destructive index (DI). Bronchoalveolar lavage was collected and analyzed. Phosphorylated (p-) mTOR in CD8+ T cells, Tc1 and Tc17 cells were detected by flow cytometry. The relative expression of p-mTOR in lungs was determined by western blot analysis. IFN-γ and IL-17A levels were detected by enzyme-linked immunosorbent assays. IFN-γ, mTOR and RAR-related orphan receptor (ROR)γt mRNA levels were evaluated by the real-time polymerase chain reaction. RESULTS Elevated p-mTOR expression in CD8+ T cells and lung tissue was accompanied by the enhanced Tc1 and Tc17 cell responses in lungs of mice exposed to cigarette smoke. Rapamycin reduced inflammatory cells in BALF and decreased MLI, DI and MAA in lungs. Rapamycin decreased p-mTOR expression, and down-regulation of mTOR and RORγt mRNA levels along with the attenuation of Tc1 and Tc17 cell responses in mice with emphysema. CONCLUSIONS The mTOR was activated in CD8+ T cells accompanied by the enhanced Tc1 and Tc17 cell responses in cigarette smoke-related pulmonary inflammation. Rapamycin ameliorated emphysema and attenuated Tc1 and Tc17 cell responses probably caused by inhibiting mTOR in cigarette smoke-exposed mice.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Xiu Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Xin Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Yuanzhen Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Shilin Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Yun Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Qiya Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Yi Liang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China
| | - Xiaoning Zhong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, China.
| |
Collapse
|
16
|
Activated CD8+ T Cells Cause Long-Term Neurological Impairment after Traumatic Brain Injury in Mice. Cell Rep 2019; 29:1178-1191.e6. [DOI: 10.1016/j.celrep.2019.09.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 08/08/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
|
17
|
Ma WT, Yao XT, Peng Q, Chen DK. The protective and pathogenic roles of IL-17 in viral infections: friend or foe? Open Biol 2019; 9:190109. [PMID: 31337278 PMCID: PMC6685926 DOI: 10.1098/rsob.190109] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Viral infections cause substantial human morbidity and mortality, and are a significant health burden worldwide. Following a viral infection, the host may initiate complex antiviral immune responses to antagonize viral invasion and replication. However, proinflammatory antiviral immune responses pose a great threat to the host if not properly held in check. Interleukin (IL)-17 is a pleiotropic cytokine participating in a variety of physiological and pathophysiological conditions, including tissue integrity maintenance, cancer progression, autoimmune disease development and, more intriguingly, infectious diseases. Abundant evidence suggests that while IL-17 plays a crucial role in enhancing effective antiviral immune responses, it may also promote and exacerbate virus-induced illnesses. Accumulated experimental and clinical evidence has broadened our understanding of the seemingly paradoxical role of IL-17 in viral infections and suggests that IL-17-targeted immunotherapy may be a promising therapeutic option. Herein, we summarize current knowledge regarding the protective and pathogenic roles of IL-17 in viral infections, with emphasis on underlying mechanisms. The various and critical roles of IL-17 in viral infections necessitate the development of therapeutic strategies that are uniquely tailored to both the infectious agent and the infection environment.
Collapse
Affiliation(s)
- Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - Xiao-Ting Yao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - Qun Peng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, People's Republic of China
| |
Collapse
|
18
|
IL-7-induced proliferation of peripheral Th17 cells is impaired in HAART-controlled HIV infection. AIDS 2019; 33:985-991. [PMID: 30946152 DOI: 10.1097/qad.0000000000002164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Th17 cells are key regulators of functional immunity in mucosal tissues, including the gut-associated lymphoid tissue (GALT), an important site of immune impairment in HIV infection. During HIV infection, Th17 cells are lost in large numbers from the GALT. Despite the recovery of peripheral CD4 T cells that accompanies suppression of viral replication with HAART, Th17 cells in GALT are not completely restored. IL-7 is essential for the survival and proliferation of T cells, but its signaling through its receptor IL-7Rα (CD127), is impaired in CD8 T cells and thymocytes during HIV infection. We set out to determine if decreased CD127 expression or impaired CD127 signaling may be the cause of Th17 impairment in HAART-controlled HIV infection. DESIGN Healthy and HIV donors on HAART were selected for this study of Th17 cell function in HIV. METHODS Peripheral CD4 T cells and Th17 cells were isolated using magnetic beads, then stimulated with IL-7. CD127 expression and the phosphorylation of signaling molecules was determined using flow cytometry. Proliferation was determined with a CFSE dilution assay. RESULTS CD127 was not decreased on Th17 cells from HAART-controlled HIV individuals, in fact, the percentage of Th17 cells that express CD127 was increased in treated HIV individuals. Furthermore, Th17 cells from HAART-controlled individuals, have normal IL-7-induced STAT5 and Bcl-2 responses, but vastly decreased proliferative responses. CONCLUSION This reduced IL-7 responsiveness may explain the lack of Th17 cell recovery and ongoing systemic immune activation that persists despite well treated HIV infection.
Collapse
|
19
|
Jin Y, Sun Z, Geng J, Yang L, Song Z, Song H, Wang J, Tang J. IL-21 reinvigorates exhausted natural killer cells in patients with HBV-associated hepatocellular carcinoma in STAT1-depedent pathway. Int Immunopharmacol 2019; 70:1-8. [PMID: 30780004 DOI: 10.1016/j.intimp.2019.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/22/2019] [Accepted: 02/04/2019] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common liver malignancy with dismal prognosis and limited treatment options. Natural killer (NK) cells are critical components of antitumor immunity due to their capacity to eliminate MHC class I-deficient cells. To evaluate the function of NK cells in HCC patients, circulating CD3-CD56+ NK cells were collected from HBV-associated HCC patients and healthy control individuals. Compared to NK cells from healthy controls, NK cells from HCC patients presented functional impairment, characterized by significantly reduced cytotoxicity, degranulation, and cytokine production. Exogenous IL-21 could reinvigorate NK cells from HCC patients, resulting in significantly increased levels of cytotoxicity, degranulation, and cytokine expression. However, IL-21-treated NK cells from HCC patients still presented lower response than IL-21-treated NK cells from healthy controls. IL-21 resulted in increased phosphorylation of both STAT1 and STAT3 in NK cells. Inhibition of STAT1, but not STAT3, significantly reduced IL-21-mediated reinvigoration of NK function. Together, this study demonstrated that NK cells in HBV-associated HCC patients presented functional impairments that could be reverted by IL-21 in a STAT1-mediated mechanism.
Collapse
Affiliation(s)
- Yun Jin
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhiwei Sun
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiawei Geng
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China; Department of Infectious Diseases, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Lei Yang
- Department of Neurosurgery, Kunming Children's Hospital, Kunming, Yunnan, China
| | - Zhenyu Song
- DICAT Biomedical Computation Centre, Vancouver, British Columbia, Canada
| | - Haihan Song
- DICAT Biomedical Computation Centre, Vancouver, British Columbia, Canada.
| | - Junfeng Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| | - Jianzhong Tang
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
20
|
Ladjemi MZ, Martin C, Lecocq M, Detry B, Nana FA, Moulin C, Weynand B, Fregimilicka C, Bouzin C, Thurion P, Carlier F, Serré J, Gayan-Ramirez G, Delos M, Ocak S, Burgel PR, Pilette C. Increased IgA Expression in Lung Lymphoid Follicles in Severe Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2019; 199:592-602. [DOI: 10.1164/rccm.201802-0352oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Maha Zohra Ladjemi
- Pôle de Pneumologie, ORL & Dermatologie
- Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium
| | - Clémence Martin
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Service de Pneumologie, Hôpital Cochin, Paris, France
| | - Marylène Lecocq
- Pôle de Pneumologie, ORL & Dermatologie
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bruno Detry
- Pôle de Pneumologie, ORL & Dermatologie
- Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium
| | | | | | | | - Chantal Fregimilicka
- Institut de Recherche Expérimentale & Clinique Imaging Platform, Institut de Recherche Expérimentale & Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Bouzin
- Institut de Recherche Expérimentale & Clinique Imaging Platform, Institut de Recherche Expérimentale & Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Pascal Thurion
- Service d’anatomopathologie, CHU de Mont-Godinne, Yvoir, Belgium
| | | | - Jef Serré
- KU Leuven, Laboratory of Respiratory Diseases, Leuven, Belgium; and
| | | | - Monique Delos
- Service d’anatomopathologie, CHU de Mont-Godinne, Yvoir, Belgium
| | - Sebahat Ocak
- Pôle de Pneumologie, ORL & Dermatologie
- Service de Pneumologie, CHU Université Catholique de Louvain Namur (Site Godinne), Yvoir, Belgium
| | - Pierre Régis Burgel
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Service de Pneumologie, Hôpital Cochin, Paris, France
| | - Charles Pilette
- Pôle de Pneumologie, ORL & Dermatologie
- Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
21
|
Imam S, Prathibha R, Dar P, Almotah K, Al-Khudhair A, Hasan SAM, Salim N, Jilani TN, Mirmira RG, Jaume JC. eIF5A inhibition influences T cell dynamics in the pancreatic microenvironment of the humanized mouse model of Type 1 Diabetes. Sci Rep 2019; 9:1533. [PMID: 30733517 PMCID: PMC6367423 DOI: 10.1038/s41598-018-38341-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022] Open
Abstract
We have developed a transgenic mouse model of Type 1 Diabetes (T1D) in which human GAD65 is expressed in pancreatic β-cells, and human MHC-II is expressed on antigen presenting cells. Induced GAD65 antigen presentation activates T-cells, which initiates the downstream events leading to diabetes. In our humanized mice, we have shown downregulation of eukaryotic translation initiation factor 5 A (elF5A), expressed only in actively dividing mammalian cells. In-vivo inhibition of elF5A hypusination by deoxyhypusine synthase (DHS) inhibitor "GC7" was studied; DHS inhibitor alters the pathophysiology in our mouse model by catalyzing the crucial hypusination and the rate-limiting step of elF5A activation. In our mouse model, we have shown that inhibition of eIF5A resets the pro-inflammatory bias in the pancreatic microenvironment. There was: (a) reduction of Th1/Th17 response, (b) an increase in Treg numbers, (c) debase in IL17 and IL21 cytokines levels in serum, (d) lowering of anti-GAD65 antibodies, and (e) ablation of the ER stress that improved functionality of the β-cells, but minimal effect on the cytotoxic CD8 T-cell (CTL) mediated response. Conclusively, immune modulation, in the case of T1D, may help to manipulate inflammatory responses, decreasing disease severity, and may help manage T1D in early stages of disease. Our study also demonstrates that without manipulating the CTLs mediated response extensively, it is difficult to treat T1D.
Collapse
Affiliation(s)
- Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| | - R Prathibha
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Pervaiz Dar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Shuhama, Srinagar, 190006, Jammu and Kashmir, India
| | - Khalil Almotah
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ahmed Al-Khudhair
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Syed Abdul-Moiz Hasan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Talha Naser Jilani
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Juan Carlos Jaume
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
22
|
Meng X, Yu X, Dong Q, Xu X, Li J, Xu Q, Ma J, Zhou C. Distribution of circulating follicular helper T cells and expression of interleukin-21 and chemokine C-X-C ligand 13 in gastric cancer. Oncol Lett 2018; 16:3917-3922. [PMID: 30128008 PMCID: PMC6096128 DOI: 10.3892/ol.2018.9112] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/09/2018] [Indexed: 02/07/2023] Open
Abstract
Circulating follicular helper T (cTfh) cells are a novel subset of cluster of differentiation (CD)4+ helper T cells. Interleukin (IL)-21 and C-X-C motif chemokine ligand (CXCL)13 are the principal effectors and chemotactic regulatory factors of Tfh. However, the roles of IL-21 and CXCL13 in gastric cancer have not yet been completely elucidated. The aim of the present study was to investigate the distribution of cTfh cells, and the expression of IL-21 and CXCL13 in patients with gastric cancer was evaluated in order to ascertain the significance and potential mechanisms of these effectors in gastric cancer. A total of 50 patients with gastric cancer were enrolled as the study subjects, with 30 healthy individuals selected as controls. The percentage of cTfh cells (cTfh%) in the peripheral blood was calculated using flow cytometry. They are identified in the present study as CD4+ chemokine C-X-C receptor (CXCR)5+ inducible T cell co-stimulator (ICOS)+ cells. The serum levels of IL-21 and CXCL13 were determined by ELISA. The cTfh% in the peripheral blood and the concentration of IL-21 and CXCL13 in the serum were significantly higher in patients with gastric cancer compared with the control group. cTfh% was significantly higher in patients with lymph node metastasis, Tumor-Node-Metastasis (TNM) stage III-IV and low differentiation. The concentrations of IL-21 and CXCL13 in patients with lymph node metastasis and/or TNM III-IV were significantly higher than in those without lymph node metastasis or with TNM I-II. There was a positive correlation between cTfh%/CXCL13 and IL-21/CXCL13, while there was no correlation between cTfh%/IL-21. cTfh cells and associated factors (IL-21/CXCL13) may be involved in the development and progression of gastric cancer. There may be mutual regulation among cTfh cells, IL-21 and CXCL13.
Collapse
Affiliation(s)
- Xinying Meng
- Department of Health Care, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xinjuan Yu
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Quanjiang Dong
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xiaona Xu
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Jinghua Li
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Qianqian Xu
- Department of Health Care, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Jian Ma
- Department of Health Care, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Changhong Zhou
- Department of Health Care, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
23
|
Yu HS, Pan ZC, Liao JL. Network modeling of inflammatory dynamics induced by biomass smoke leading to chronic obstructive pulmonary disease. CHINESE J CHEM PHYS 2018. [DOI: 10.1063/1674-0068/31/cjcp1801001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Hai-shan Yu
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Zhi-chao Pan
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Jie-lou Liao
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
24
|
Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1033. [PMID: 29883409 PMCID: PMC5982072 DOI: 10.3390/ijerph15051033] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Many studies have been undertaken to reveal how tobacco smoke skews immune responses contributing to the development of chronic obstructive pulmonary disease (COPD) and other lung diseases. Recently, environmental tobacco smoke (ETS) has been linked with asthma and allergic diseases in children. This review presents the most actual knowledge on exact molecular mechanisms responsible for the skewed inflammatory profile that aggravates inflammation, promotes infections, induces tissue damage, and may promote the development of allergy in individuals exposed to ETS. We demonstrate how the imbalance between oxidants and antioxidants resulting from exposure to tobacco smoke leads to oxidative stress, increased mucosal inflammation, and increased expression of inflammatory cytokines (such as interleukin (IL)-8, IL-6 and tumor necrosis factor α ([TNF]-α). Direct cellular effects of ETS on epithelial cells results in increased permeability, mucus overproduction, impaired mucociliary clearance, increased release of proinflammatory cytokines and chemokines, enhanced recruitment of macrophages and neutrophils and disturbed lymphocyte balance towards Th2. The plethora of presented phenomena fully justifies a restrictive policy aiming at limiting the domestic and public exposure to ETS.
Collapse
Affiliation(s)
- Agnieszka Strzelak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksandra Ratajczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksander Adamiec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| |
Collapse
|
25
|
Liang Y, Shen Y, Kuang L, Zhou G, Zhang L, Zhong X, Zhang J, Liu J. Cigarette smoke exposure promotes differentiation of CD4 + T cells toward Th17 cells by CD40-CD40L costimulatory pathway in mice. Int J Chron Obstruct Pulmon Dis 2018; 13:959-968. [PMID: 29606863 PMCID: PMC5868633 DOI: 10.2147/copd.s155754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose This study aimed to investigate the impact of cigarette smoke exposure upon CD40–CD40L ligation between bone marrow-derived dendritic cells (BMDCs)and CD4+T cells, and to examine the effects of cigarette smoke exposure upon differentiation of CD4+T cells toward Th17 cells through blockade of CD40-CD40L pathway in mice. Methods The study was processed in vivo and in vitro. In vivo, Th17 cells, CD40, interleukin (IL)-17A, and IL-27 in the lung tissues were quantified and compared between mice with and without cigarette smoke exposure. In vitro, Th17 cells, IL-17A, and IL-27 yielded by multiple cell cultivations in which BMDCs from mice with or without cigarette smoke exposure were fostered with CD4+ T cells from healthy mice spleens in the presence of antagonistic CD40 antibody and/or cigarette smoke extract (CSE) were quantified and compared. The flow cytometry was used to detect expressions of Th17 cells and CD40, and the liquid chip was used to detect levels of IL-17A and IL-27. Results Both in vivo exposed to cigarette smoke and in vitro to CSE, CD40 expressions noticeably escalated on the surfaces of BMDCs. The presence of Th17 cells, IL-17A, and IL-27 in the lung tissues prominently increased in mice exposed to cigarette smoke. The in vitro culture of CD4+ T cells and BMDCs significantly enhanced the differentiation of CD4+ T cells toward Th17 cells and secretions of IL-17A and IL-27 in the case that BMDCs were produced from mice exposed to cigarette smoke or the culture occurred in the presence of CSE. Usage of antagonistic CD40 antibody evidently reduced the number of Th17 cells, IL-17A, and IL-27 that increased due to cigarette smoke exposure. Conclusion The CD40–CD40L ligation is associated with the quantities of Th17 cells and relevant cytokines in the context of cigarette smoke exposure. Reducing the number of Th17 cells via the usage of antagonistic CD40 antibody can be an inspiration for pursuing a novel therapeutic target for immune inflammation in COPD.
Collapse
Affiliation(s)
- Yi Liang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Ying Shen
- Division of General Practice, General Practice School of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Liangjian Kuang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Guang Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Longju Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Xiaoning Zhong
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Jianquan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Jifeng Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| |
Collapse
|
26
|
IL-23 signaling in Th17 cells is inhibited by HIV infection and is not restored by HAART: Implications for persistent immune activation. PLoS One 2017; 12:e0186823. [PMID: 29091911 PMCID: PMC5665519 DOI: 10.1371/journal.pone.0186823] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES HIV infection causes a profound depletion of gut derived Th17 cells, contributing to loss of mucosal barrier function and an increase in microbial translocation, thus driving systemic immune activation. Despite normalization of circulating CD4+ T cell counts with highly active antiretroviral therapy (HAART), Th17 frequency and function often remain impaired. Given the importance of interleukin (IL)-23 in the generation and stabilization of Th17 cells we hypothesized that impaired IL-23 signaling causes persistent Th17 dysfunction in HIV infection. METHODS The effects of in vitro HIV infection on responses to IL-23 in Th17 cells were examined. These included the production of IL-17, phosphorylated STAT3 (pSTAT3) and the transcription of retinoic acid orphan receptor C (RORC) gene. Blood derived Th17 cells from untreated and HAART-treated HIV-infected individuals were also examined for the IL-23 induced production of phosphorylated STAT3 (pSTAT3) and the expression of the IL-23 receptors. RESULTS In vitro HIV infection significantly inhibited IL-17 production and IL-23 induced pSTAT3 while expression of RORC RNA was unaffected. Th17 cells isolated from untreated and HAART-treated HIV-infected individuals showed complete loss of IL-23 induced pSTAT3 without a decrease in the expression of the IL-23 receptors. CONCLUSIONS This study is the first to demonstrate an effect of HIV on the IL-23 signaling pathway in Th17 cells. We show that in vitro and in vivo HIV infection results in impaired IL-23 signaling which is not reversed by HAART nor is it a result of reduced receptor expression, suggesting that HIV interferes with IL-23-activated signaling pathways. These findings may explain the inability of HAART to restore Th17 frequency and function and the resulting persistent chronic immune activation observed in HIV infected individuals.
Collapse
|
27
|
Duan MC, Zhang JQ, Liang Y, Liu GN, Xiao J, Tang HJ, Liang Y. Infiltration of IL-17-Producing T Cells and Treg Cells in a Mouse Model of Smoke-Induced Emphysema. Inflammation 2017; 39:1334-44. [PMID: 27150336 DOI: 10.1007/s10753-016-0365-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive and irreversible chronic inflammatory disease associated with the accumulation of activated T cells. To date, there is little information concerning the intrinsic association among Th17, Tc17, and regulatory T (Treg) cells in COPD. The objective of this study was to investigate the variation of lungs CD4(+)Foxp3(+) Treg cells and IL-17-producing CD4 and CD8 (Th17 and Tc17) lymphocytes in mice with cigarette-induced emphysema. Groups of mice were exposed to cigarette smoke or room air. At weeks 12 and 24, mice were sacrificed to observe histological changes by HE stain. The frequencies of Th17 (CD4(+)IL-17(+)T), Tc17 (CD8(+)IL-17(+)T), and Treg (CD4(+)Foxp3(+)T) cells in lungs from these mice were analyzed by flow cytometry. The mRNA levels of orphan nuclear receptor ROR γt and Foxp3 were performed by real-time quantitative polymerase chain reaction. The protein levels of interleukin-17 (IL-17), IL-6, IL-10, and transforming growth factor-beta (TGF-β1) were measured by enzyme-linked immunosorbent assay. Cigarette smoke caused substantial enlargement of the air spaces accompanied by the destruction of the normal alveolar architecture and led to emphysema. The frequencies of Th17 and Tc17 cells, as well as the expressions of IL-6, IL-17, TGF-β1, and ROR γt were greater in the lungs of cigarette smoke (CS)-exposed mice, particularly in the 24-week CS-exposed mice. The frequencies of Treg cells and the expressions of IL-10 and Foxp3 were lower in CS-exposed mice compared to control group. More important, the frequencies of Tregs were negatively correlated with Th17 cells and with Tc17 cells. Interestingly, a significant portion of the cells that infiltrate the lungs was skewed towards a Tc17 phenotype. Our findings suggest the contribution of Th17, Tc17, and Treg cells in the pathogenesis of COPD. Rebalance of these cells will be helpful for developing and refining the new immunological therapies for COPD.
Collapse
Affiliation(s)
- Min-Chao Duan
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jian-Quan Zhang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yue Liang
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
| | - Guang-Nan Liu
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jin Xiao
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
| | - Hai-Juan Tang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yi Liang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
28
|
Qiu F, Liang CL, Liu H, Zeng YQ, Hou S, Huang S, Lai X, Dai Z. Impacts of cigarette smoking on immune responsiveness: Up and down or upside down? Oncotarget 2017; 8:268-284. [PMID: 27902485 PMCID: PMC5352117 DOI: 10.18632/oncotarget.13613] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/12/2016] [Indexed: 01/08/2023] Open
Abstract
Cigarette smoking is associated with numerous diseases and poses a serious challenge to the current healthcare system worldwide. Smoking impacts both innate and adaptive immunity and plays dual roles in regulating immunity by either exacerbation of pathogenic immune responses or attenuation of defensive immunity. Adaptive immune cells affected by smoking mainly include T helper cells (Th1/Th2/Th17), CD4+CD25+ regulatory T cells, CD8+ T cells, B cells and memory T/B lymphocytes while innate immune cells impacted by smoking are mostly DCs, macrophages and NK cells. Complex roles of cigarette smoke have resulted in numerous diseases, including cardiovascular, respiratory and autoimmune diseases, allergies, cancers and transplant rejection etc. Although previous reviews have described the effects of smoking on various diseases and regional immunity associated with specific diseases, a comprehensive and updated review is rarely seen to demonstrate impacts of smoking on general immunity and, especially on major components of immune cells. Here, we aim to systematically and objectively review the influence of smoking on major components of both innate and adaptive immune cells, and summarize cellular and molecular mechanisms underlying effects of cigarette smoking on the immune system. The molecular pathways impacted by cigarette smoking involve NFκB, MAP kinases and histone modification. Further investigations are warranted to understand the exact mechanisms responsible for smoking-mediated immunopathology and to answer lingering questions over why cigarette smoking is always harmful rather than beneficial even though it exerts dual effects on immune responses.
Collapse
Affiliation(s)
- Feifei Qiu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chun-Ling Liang
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huazhen Liu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yu-Qun Zeng
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shaozhen Hou
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Song Huang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoping Lai
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhenhua Dai
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Pan Z, Yu H, Liao JL. Probing Cellular and Molecular Mechanisms of Cigarette Smoke-Induced Immune Response in the Progression of Chronic Obstructive Pulmonary Disease Using Multiscale Network Modeling. PLoS One 2016; 11:e0163192. [PMID: 27669518 PMCID: PMC5036797 DOI: 10.1371/journal.pone.0163192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/06/2016] [Indexed: 01/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disorder characterized by progressive destruction of lung tissues and airway obstruction. COPD is currently the third leading cause of death worldwide and there is no curative treatment available so far. Cigarette smoke (CS) is the major risk factor for COPD. Yet, only a relatively small percentage of smokers develop the disease, showing that disease susceptibility varies significantly among smokers. As smoking cessation can prevent the disease in some smokers, quitting smoking cannot halt the progression of COPD in others. Despite extensive research efforts, cellular and molecular mechanisms of COPD remain elusive. In particular, the disease susceptibility and smoking cessation effects are poorly understood. To address these issues in this work, we develop a multiscale network model that consists of nodes, which represent molecular mediators, immune cells and lung tissues, and edges describing the interactions between the nodes. Our model study identifies several positive feedback loops and network elements playing a determinant role in the CS-induced immune response and COPD progression. The results are in agreement with clinic and laboratory measurements, offering novel insight into the cellular and molecular mechanisms of COPD. The study in this work also provides a rationale for targeted therapy and personalized medicine for the disease in future.
Collapse
Affiliation(s)
- Zhichao Pan
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| | - Haishan Yu
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| | - Jie-Lou Liao
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| |
Collapse
|
30
|
Kuang LJ, Deng TT, Wang Q, Qiu SL, Liang Y, He ZY, Zhang JQ, Bai J, Li MH, Deng JM, Liu GN, Liu JF, Zhong XN. Dendritic cells induce Tc1 cell differentiation via the CD40/CD40L pathway in mice after exposure to cigarette smoke. Am J Physiol Lung Cell Mol Physiol 2016; 311:L581-9. [PMID: 27448664 DOI: 10.1152/ajplung.00002.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells and CD8(+) T cells participate in the pathology of chronic obstructive pulmonary disease, including emphysema, but little is known of the involvement of the CD40/CD40L pathway. We investigated the role of the CD40/CD40L pathway in Tc1 cell differentiation induced by dendritic cells in a mouse model of emphysema, and in vitro. C57BL/6J wild-type and CD40(-/-) mice were exposed to cigarette smoke (CS) or not (control), for 24 wk. In vitro experiments involved wild-type and CD40(-/-) dendritic cells treated with CS extract (CSE) or not. Compared with the control groups, the CS mice (both wild type and CD40(-/-)) had a greater percentage of lung dendritic cells and higher levels of major histocompatability complex (MHC) class I molecules and costimulatory molecules CD40 and CD80. Relative to the CS CD40(-/-) mice, the CS wild type showed greater signs of lung damage and Tc1 cell differentiation. In vitro, the CSE-treated wild-type cells evidenced more cytokine release (IL-12/p70) and Tc1 cell differentiation than did the CSE-treated CD40(-/-) cells. Exposure to cigarette smoke increases the percentage of lung dendritic cells and promotes Tc1 cell differentiation via the CD40/CD40L pathway. Blocking the CD40/CD40L pathway may suppress development of emphysema in mice exposed to cigarette smoke.
Collapse
Affiliation(s)
- Liang-Jian Kuang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Ting-Ting Deng
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Qin Wang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Shi-Lin Qiu
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Yi Liang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Zhi-Yi He
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jian-Quan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jing Bai
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Mei-Hua Li
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jing-Min Deng
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Guang-Nan Liu
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Ji-Feng Liu
- Department of Respiratory Medicine, Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou, Guangxi, China
| | - Xiao-Ning Zhong
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| |
Collapse
|
31
|
Busman-Sahay KO, Walrath T, Huber S, O'Connor W. Cytokine crowdsourcing: multicellular production of TH17-associated cytokines. J Leukoc Biol 2015; 97:499-510. [PMID: 25548251 PMCID: PMC5477895 DOI: 10.1189/jlb.3ru0814-386r] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/17/2014] [Accepted: 11/20/2014] [Indexed: 12/14/2022] Open
Abstract
In the 2 decades since its discovery, IL-17A has become appreciated for mounting robust, protective responses against bacterial and fungal pathogens. When improperly regulated, however, IL-17A can play a profoundly pathogenic role in perpetuating inflammation and has been linked to a wide variety of debilitating diseases. IL-17A is often present in a composite milieu that includes cytokines produced by TH17 cells (i.e., IL-17F, IL-21, IL-22, and IL-26) or associated with other T cell lineages (e.g., IFN-γ). These combinatorial effects add mechanistic complexity and more importantly, contribute differentially to disease outcome. Whereas TH17 cells are among the best-understood cell types that secrete IL-17A, they are frequently neither the earliest nor dominant producers. Indeed, non-TH17 cell sources of IL-17A can dramatically alter the course and severity of inflammatory episodes. The dissection of the temporal regulation of TH17-associated cytokines and the resulting net signaling outcomes will be critical toward understanding the increasingly intricate role of IL-17A and TH17-associated cytokines in disease, informing our therapeutic decisions. Herein, we discuss important non-TH17 cell sources of IL-17A and other TH17-associated cytokines relevant to inflammatory events in mucosal tissues.
Collapse
Affiliation(s)
- Kathleen O Busman-Sahay
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Travis Walrath
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - William O'Connor
- *Center for Immunology and Microbial Disease, Albany Medical Center, Albany, New York, USA; and Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
32
|
Zhou H, Hua W, Jin Y, Zhang C, Che L, Xia L, Zhou J, Chen Z, Li W, Shen H. Tc17 cells are associated with cigarette smoke-induced lung inflammation and emphysema. Respirology 2015; 20:426-33. [PMID: 25677967 DOI: 10.1111/resp.12486] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/14/2014] [Accepted: 11/23/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Hongbin Zhou
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Wen Hua
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Yan Jin
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Chao Zhang
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Luanqing Che
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Lixia Xia
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Jiesen Zhou
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Zhihua Chen
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine; Second Hospital of Zhejiang University School of Medicine; Hangzhou Zhejiang China
- State Key Lab of Respiratory Disease; Guangzhou China
| |
Collapse
|
33
|
Wang MH, Fiocchi C, Zhu X, Ripke S, Kamboh MI, Rebert N, Duerr RH, Achkar JP. Gene-gene and gene-environment interactions in ulcerative colitis. Hum Genet 2014; 133:547-58. [PMID: 24241240 PMCID: PMC4064459 DOI: 10.1007/s00439-013-1395-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies (GWAS) have identified at least 133 ulcerative colitis (UC) associated loci. The role of genetic factors in clinical practice is not clearly defined. The relevance of genetic variants to disease pathogenesis is still uncertain because of not characterized gene-gene and gene-environment interactions. We examined the predictive value of combining the 133 UC risk loci with genetic interactions in an ongoing inflammatory bowel disease (IBD) GWAS. The Wellcome Trust Case-Control Consortium (WTCCC) IBD GWAS was used as a replication cohort. We applied logic regression (LR), a novel adaptive regression methodology, to search for high-order interactions. Exploratory genotype correlations with UC sub-phenotypes [extent of disease, need of surgery, age of onset, extra-intestinal manifestations and primary sclerosing cholangitis (PSC)] were conducted. The combination of 133 UC loci yielded good UC risk predictability [area under the curve (AUC) of 0.86]. A higher cumulative allele score predicted higher UC risk. Through LR, several lines of evidence for genetic interactions were identified and successfully replicated in the WTCCC cohort. The genetic interactions combined with the gene-smoking interaction significantly improved predictability in the model (AUC, from 0.86 to 0.89, P = 3.26E-05). Explained UC variance increased from 37 to 42 % after adding the interaction terms. A within case analysis found suggested genetic association with PSC. Our study demonstrates that the LR methodology allows the identification and replication of high-order genetic interactions in UC GWAS datasets. UC risk can be predicted by a 133 loci and improved by adding gene-gene and gene-environment interactions.
Collapse
Affiliation(s)
- Ming-Hsi Wang
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk A31, Cleveland, OH, 44195, USA,
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ruwanpura SM, McLeod L, Brooks GD, Bozinovski S, Vlahos R, Longano A, Bardin PG, Anderson GP, Jenkins BJ. IL-6/Stat3-driven pulmonary inflammation, but not emphysema, is dependent on interleukin-17A in mice. Respirology 2014; 19:419-27. [DOI: 10.1111/resp.12243] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/25/2013] [Accepted: 12/15/2013] [Indexed: 01/12/2023]
Affiliation(s)
- Saleela M. Ruwanpura
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Gavin D. Brooks
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Steven Bozinovski
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Ross Vlahos
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Anthony Longano
- Pathology; Monash Medical Centre; Clayton Victoria Australia
| | - Philip G. Bardin
- Respiratory and Sleep Medicine; Monash Medical Centre; Clayton Victoria Australia
| | - Gary P. Anderson
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Brendan J. Jenkins
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| |
Collapse
|
35
|
Duan M, Huang Y, Zhong X, Tang H. IL-21 is increased in peripheral blood of emphysema mice and promotes Th1/Tc1 cell generation in vitro. Inflammation 2013; 37:745-55. [PMID: 24357415 DOI: 10.1007/s10753-013-9793-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Interleukin-21 (IL-21) has been reported to be involved in many Th1-associated diseases. However, the alteration and immune regulation of IL-21 in emphysema remains unknown. In this study, we tested the levels of IFN-γ and IL-21 and the frequencies of Th1 and Tc1 in peripheral blood from cigarette smoke (CS)-exposed mice and air-exposed mice and explored the effect of IL-21 on generation of Th1 and Tc1 cells in vitro. It was found that the levels of IFN-γ and IL-21 and the frequencies of Th1, Tc1, CD4(+) IL-21(+), CD4(+) IL-21R(+), and CD8(+) IL-21R(+) T cells were much higher in CS-exposed mice. Moreover, the levels of IL-21 were correlated positively with Th1 cells and with Tc1 cells. Finally, the in vitro experiments showed that IL-21 could promote Th1/Tc1 cell generation in CS-exposed mice. These results indirectly provide evidence that IL-21 produced by CD4(+) T cells could promote Th1/Tc1 response, leading to systemic inflammation in emphysema.
Collapse
Affiliation(s)
- Minchao Duan
- Department of Respiratory Medicine, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Avenue, Nanning, Guangxi, 530021, China
| | | | | | | |
Collapse
|