1
|
Morali K, Giacomello G, Vuono M, Gregori S. Leveraging current insights on IL-10-producing dendritic cells for developing effective immunotherapeutic approaches. FEBS Lett 2024. [PMID: 39266465 DOI: 10.1002/1873-3468.15017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells involved in promoting and controlling immune responses. Different subsets of DC, named tolerogenic (tol)DC, play a critical role in the maintenance of tissue homeostasis and in fostering tolerance. These unique skills make tolDC especially attractive for strategies aimed at re-establishing/inducing tolerance in immune-mediated conditions. The generation of potent tolDC in vitro from peripheral blood monocytes has seen remarkable advancements. TolDC modulate T cell dynamics by favoring regulatory T cells (Tregs) and curbing effector/pathogenic T cells. Among the several methods developed for in vitro tolDC generation, IL-10 conditioning has been proven to be the most efficient, as IL-10-modulated tolDC were demonstrated to promote Tregs with the strongest suppressive activities. Investigating the molecular, metabolic, and functional profiles of tolDC uncovers essential pathways that facilitate their immunoregulatory functions. This Review provides an overview of current knowledge on the role of tolDC in health and disease, focusing on IL-10 production, functional characterization of in vitro generated tolDC, molecular and metabolic changes occurring in tolDC induced by tolerogenic agents, clinical applications of tolDC-based therapy, and finally new perspectives in the generation of effective tolDC.
Collapse
Affiliation(s)
- Konstantina Morali
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gloria Giacomello
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- PhD Course in Medicina Traslazionale e Molecolare (DIMET), University of Milano Bicocca, Italy
| | - Michela Vuono
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- PhD Course in Molecular Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
2
|
Chen KY, Sun WL, Wu SM, Feng PH, Lin CF, Chen TT, Lu YH, Ho SC, Chen YH, Lee KY. Reduced Tolerogenic Program Death-Ligand 1-Expressing Conventional Type 1 Dendritic Cells Are Associated with Rapid Decline in Chronic Obstructive Pulmonary Disease. Cells 2024; 13:878. [PMID: 38786101 PMCID: PMC11119227 DOI: 10.3390/cells13100878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized, at least in part, by autoimmunity through amplified T helper 1 and 17 (Th1 and Th17) immune responses. The loss of immune tolerance controlled by programmed death-ligand 1 (PD-L1) may contribute to this. OBJECTIVES We studied the tolerogenic role of PD-L1+ dendritic cells (DCs) and their subtypes in relation to specific T cell immunity and the clinical phenotypes of COPD. METHODS We used flow cytometry to analyze PD-L1 expression by the DCs and their subtypes in the peripheral blood mononuclear cells (PBMCs) from normal participants and those with COPD. T cell proliferation and the signature cytokines of T cell subtypes stimulated with elastin as autoantigens were measured using flow cytometry and enzyme-linked immunosorbent assays (ELISA), respectively. MEASUREMENT AND MAIN RESULTS A total of 83 participants were enrolled (normal, n = 29; COPD, n = 54). A reduced PD-L1+ conventional dendritic cell 1 (cDC1) ratio in the PBMCs of the patients with COPD was shown (13.7 ± 13.7%, p = 0.03). The decrease in the PD-L1+ cDC1 ratio was associated with a rapid decline in COPD (p = 0.02) and correlated with the CD4+ T cells (r = -0.33, p = 0.02). This is supported by the NCBI GEO database accession number GSE56766, the researchers of which found that the gene expressions of PD-L1 and CD4, but not CD8 were negatively correlated from PBMC in COPD patients (r = -0.43, p = 0.002). Functionally, the PD-L1 blockade enhanced CD4+ T cell proliferation stimulated by CD3/elastin (31.2 ± 22.3%, p = 0.04) and interleukin (IL)-17A production stimulated by both CD3 (156.3 ± 54.7, p = 0.03) and CD3/elastin (148 ± 64.9, p = 0.03) from the normal PBMCs. The PD-L1 blockade failed to increase IL-17A production in the cDC1-depleted PBMCs. By contrast, there was no significant change in interferon (IFN)-γ, IL-4, or IL-10 after the PD-L1 blockade. Again, these findings were supported by the NCBI GEO database accession number GSE56766, the researchers of which found that only the expression of RORC, a master transcription factor driving the Th17 cells, was significantly negatively correlated to PD-L1 (r = -0.33, p = 0.02). CONCLUSIONS Circulating PD-L1+ cDC1 was reduced in the patients with COPD, and the tolerogenic role was suppressed with susceptibility to self-antigens and linked to rapid decline caused by Th17-skewed chronic inflammation.
Collapse
Affiliation(s)
- Kuan-Yuan Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-Y.C.); (T.-T.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Tzu-Tao Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-Y.C.); (T.-T.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yueh-Hsun Lu
- Department of Radiology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan;
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yueh-Hsi Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
| | - Kang-Yun Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-Y.C.); (T.-T.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (W.-L.S.); (S.-M.W.); (P.-H.F.); (S.-C.H.); (Y.-H.C.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
3
|
Meisser SS, Mitamura Y, Altunbulakli C, Bandier J, Opstrup MS, Gadsbøll ASØ, Li M, Tan G, Akdis M, Akdis CA, Geisler C, Johansen JD, Bonefeld CM. Regulation of immune response genes in the skin of allergic and clinically tolerant individuals exposed to p-phenylenediamine. Allergy 2024; 79:1291-1305. [PMID: 38263750 DOI: 10.1111/all.16031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/10/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND p-Phenylenediamine (PPD) is a potent contact allergen found in many hair colour products. However, not all individuals develop allergic contact dermatitis (ACD) although they are regularly exposed to PPD. It is unclear whether these asymptomatic individuals are true non-responders to PPD or whether they mount a response to PPD without showing any symptoms. METHODS Skin biopsies were collected from 11 asymptomatic hairdressers regularly exposed to PPD and from 10 individuals with known ACD on day 4 after patch testing with 1% PPD in petrolatum and petrolatum exclusively as control. RNA sequencing and confocal microscopy were performed. RESULTS T cell activation, inflammation and apoptosis pathways were up-regulated by PPD in both asymptomatic and allergic individuals. Compared to asymptomatic individuals with a negative patch test, individuals with a strong reaction to PPD strongly up-regulated both pro- and anti-inflammatory cytokines genes. Interestingly, PPD treatment induced significant up-regulation of several genes for chemokines, classical type 2 dendritic cell markers and regulatory T cell markers in both asymptomatic and allergic individuals. In addition, apoptosis signalling pathway was activated in both non-responders and allergic individuals. CONCLUSION This study demonstrates that there are no true non-responders to PPD but that the immune response elicited by PPD differs between individuals and can lead to either tolerance, subclinical inflammation or allergy.
Collapse
Affiliation(s)
- Sanne S Meisser
- National Allergy Research Centre, Department of Dermato-Allergology, Copenhagen University Hospital Gentofte, Hellerup, Denmark
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Can Altunbulakli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Josefine Bandier
- National Allergy Research Centre, Department of Dermato-Allergology, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Morten S Opstrup
- National Allergy Research Centre, Department of Dermato-Allergology, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Anne-Sofie Ø Gadsbøll
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
- Christine Kühne- Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Carsten Geisler
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeanne D Johansen
- National Allergy Research Centre, Department of Dermato-Allergology, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Charlotte M Bonefeld
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Xu J, Xiao N, Zhou D, Xie L. Disease tolerance: a protective mechanism of lung infections. Front Cell Infect Microbiol 2023; 13:1037850. [PMID: 37207185 PMCID: PMC10189053 DOI: 10.3389/fcimb.2023.1037850] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/30/2023] [Indexed: 05/21/2023] Open
Abstract
Resistance and tolerance are two important strategies employed by the host immune response to defend against pathogens. Multidrug-resistant bacteria affect the resistance mechanisms involved in pathogen clearance. Disease tolerance, defined as the ability to reduce the negative impact of infection on the host, might be a new research direction for the treatment of infections. The lungs are highly susceptible to infections and thus are important for understanding host tolerance and its precise mechanisms. This review focuses on the factors that induce lung disease tolerance, cell and molecular mechanisms involved in tissue damage control, and the relationship between disease tolerance and sepsis immunoparalysis. Understanding the exact mechanism of lung disease tolerance could allow better assessment of the immune status of patients and provide new ideas for the treatment of infections.
Collapse
Affiliation(s)
- Jianqiao Xu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Nan Xiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- *Correspondence: Dongsheng Zhou, ; Lixin Xie,
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
- *Correspondence: Dongsheng Zhou, ; Lixin Xie,
| |
Collapse
|
5
|
Staphylococcus aureus-induced immunosuppression mediated by IL-10 and IL-27 facilitates nasal colonisation. PLoS Pathog 2022; 18:e1010647. [PMID: 35776778 PMCID: PMC9282462 DOI: 10.1371/journal.ppat.1010647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/14/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus persistently colonises the anterior nares of a significant proportion of the healthy population, however the local immune response elicited during S. aureus nasal colonisation remains ill-defined. Local activation of IL-17/IL-22 producing T cells are critical for controlling bacterial clearance from the nasal cavity. However, recurrent and long-term colonisation is commonplace indicating efficient clearance does not invariably occur. Here we identify a central role for the regulatory cytokine IL-10 in facilitating bacterial persistence during S. aureus nasal colonisation in a murine model. IL-10 is produced rapidly within the nasal cavity following S. aureus colonisation, primarily by myeloid cells. Colonised IL-10-/- mice demonstrate enhanced IL-17+ and IL-22+ T cell responses and more rapidly clear bacteria from the nasal tissues as compared with wild-type mice. S. aureus also induces the regulatory cytokine IL-27 within the nasal tissue, which acts upstream of IL-10 promoting its production. IL-27 blockade reduces IL-10 production within the nasal cavity and improves bacterial clearance. TLR2 signalling was confirmed to be central to controlling the IL-10 response. Our findings conclude that during nasal colonisation S. aureus creates an immunosuppressive microenvironment through the local induction of IL-27 and IL-10, to dampen protective T cell responses and facilitate its persistence. Nasal colonisation by the bacterium Staphylococcus aureus is a very common occurrence in the human population. However there is a lack of knowledge on the immune response that controls nasal colonisation. It is known that a local pro-inflammatory immune response is important for bacterial clearance, however sustained colonisation is commonplace suggesting efficient clearance may not be occurring. Here we demonstrate for the first time that S. aureus is manipulating the host immune response by promoting immunosuppression in the nasal cavity which enables bacterial survival. We found that the regulatory proteins IL-10 and IL-27 are central to this suppressive response and result in reduced protective T cell responses. We also demonstrate that S. aureus is inducing IL-27 production to enhance IL-10 production in order to prolong bacterial colonisation. Our findings show that the host-pathogen interaction during nasal colonisation is more complex than previously described and that S. aureus is capable of manipulating the regulatory immune response of the host for its’ own benefit.
Collapse
|
6
|
Iberg CA, Hawiger D. Natural and Induced Tolerogenic Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:733-744. [PMID: 32015076 DOI: 10.4049/jimmunol.1901121] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are highly susceptible to extrinsic signals that modify the functions of these crucial APCs. Maturation of DCs induced by diverse proinflammatory conditions promotes immune responses, but certain signals also induce tolerogenic functions in DCs. These "induced tolerogenic DCs" help to moderate immune responses such as those to commensals present at specific anatomical locations. However, also under steady-state conditions, some DCs are characterized by inherent tolerogenic properties. The immunomodulatory mechanisms constitutively present in such "natural tolerogenic DCs" help to promote tolerance to peripheral Ags. By extending tolerance initially established in the thymus, these functions of DCs help to regulate autoimmune and other immune responses. In this review we will discuss the mechanisms and functions of natural and induced tolerogenic DCs and offer further insight into how their possible manipulations may ultimately lead to more precise treatments for various immune-mediated conditions and diseases.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
7
|
Peters M, Peters K, Bufe A. Regulation of lung immunity by dendritic cells: Implications for asthma, chronic obstructive pulmonary disease and infectious disease. Innate Immun 2020; 25:326-336. [PMID: 31291810 PMCID: PMC7103613 DOI: 10.1177/1753425918821732] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Since the first description of dendritic cells by Steinman and Cohn in 1973, this
important cell type has gained increasing attention. Over 4000 papers have been
published on this topic annually during the last few years. At the beginning,
dendritic cells were recognized for their immune stimulatory properties and
their importance in initiating an adaptive immune response. Later, it was found
that dendritic cells do not only initiate but also regulate immune responses.
This attribute makes the so-called regulatory dendritic cells highly important
for the prevention of exaggerated immune responses. Immune cells make contact
with different Ags every day and must be tightly controlled to prevent excessive
inflammation and subsequent organ destruction, particularly in organs such as
the gut and lungs. Here, we give a brief overview of our current knowledge on
how immune responses are controlled by dendritic cells, highlighting how they
are involved in the induction of peripheral tolerance. We focus on what is known
about these processes in the lung, with a closer look at their role in the
induction and control of diseases such as bronchial asthma, chronic obstructive
pulmonary disease and lung infections. Finally, we summarize some current
approaches to modulate the behavior of dendritic cells that may hopefully lead
to future therapeutics to control exaggerated immune responses.
Collapse
Affiliation(s)
- Marcus Peters
- Department of Experimental Pneumology, Ruhr-University Bochum, Germany
| | - Karin Peters
- Department of Experimental Pneumology, Ruhr-University Bochum, Germany
| | - Albrecht Bufe
- Department of Experimental Pneumology, Ruhr-University Bochum, Germany
| |
Collapse
|
8
|
Carvalheiro T, Zimmermann M, Radstake TRDJ, Marut W. Novel insights into dendritic cells in the pathogenesis of systemic sclerosis. Clin Exp Immunol 2020; 201:25-33. [PMID: 31970748 PMCID: PMC7290079 DOI: 10.1111/cei.13417] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (SSc) is a severe autoimmune fibrotic disease characterized by fibrosis, vasculopathy, and immune dysregulation. Dendritic cells (DCs) are the most potent antigen-presenting cells, specialized in pathogen sensing, with high capacity to shape the immune responses. The most recent technological advances have allowed the discovery of new DC subsets with potential implications in inflammatory conditions. Alterations of DC distribution in circulation and affected tissue as well as impaired DC function have been described in SSc patients, pointing towards a crucial role of these cells in SSc pathogenesis. In particular, recent studies have shown the importance of plasmacytoid DCs either by their high capacity to produce type I interferon or other inflammatory mediators implicated in SSc pathology, such as chemokine C-X-C motif ligand 4 (CXCL4). In-vivo models of SSc have been vital to clarify the implications of DCs in this disease, especially DCs depletion and specific gene knock-down studies. This review provides these new insights into the contribution of the different DCs subsets in the pathogenesis of SSc, as well as to the novel developments on DCs in in-vivo models of SSc and the potential use of DCs and their mediators as therapeutic targets.
Collapse
Affiliation(s)
- T Carvalheiro
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M Zimmermann
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - T R D J Radstake
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - W Marut
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
9
|
Júlio de Souza AL, Beatriz Mahler Pereira A, Robison de Oliveira J, Santos Ramalho L, Ismarsi de Souza H, Lacerda Nascimento A, Uddin M, Sérgio Pereira P, Nascimento Silva Teixeira D, Roberto da Silva P, Alves de Albuquerque D, Helena Faccioli L, de Paula Rogério A. Dermatophagoides pteronyssinus-induced pro-inflammatory responses mediated via STAT3 and NF-kappaB signaling pathways in human bronchial epithelial cells - Inhibitory effects of Lafoensia pacari and ellagic acid. J Pharmacol Sci 2020; 142:157-164. [PMID: 32008948 DOI: 10.1016/j.jphs.2020.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022] Open
Abstract
The house dust mite allergen Dermatophagoides pteronyssinus (Der p) is a major driver of allergic asthma. Studies from our group demonstrated anti-eosinophilic effects of ethanolic extract of Lafoensia pacari stem bark (and ellagic acid, isolated from L. pacari extract), used as traditional medicine in Brazil to naturally treat inflammatory conditions. Here, we extended these results through performing phytochemical analysis of the constituents of L. pacari using liquid chromatography-mass spectrometry and evaluating the anti-inflammatory effects of both L. pacari and ellagic acid in the human BEAS-2B bronchial epithelial cell line stimulated with Der p. Ellagic acid (major constituent), gallic acid, ferulic acid, chlorogenic acid and rosmarinic acid, but not flavonoids (rutin, kaempferol, luteolin and quercetin), were found in the L. pacari. Pro-inflammatory mediators, IL-6, IL-8 and CCL-2 production were increased in BEAS-2B stimulated with Der p (10 μg/mL, 24 h) compared to control. L. pacari (250 μg/mL) and ellagic acid (100 μM) significantly reduced the concentration of these mediators. L. pacari increased the production of the anti-inflammatory cytokine, IL-10. These results were associated with the downregulation of NF-κB and STAT3 pathways. These findings indicate a novel anti-inflammatory action for L. pacari and ellagic acid in the airways allergic inflammatory response.
Collapse
Affiliation(s)
- Ana Letícia Júlio de Souza
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Aline Beatriz Mahler Pereira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Jhony Robison de Oliveira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Luciana Santos Ramalho
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Henrique Ismarsi de Souza
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | | | - Mohib Uddin
- Respiratory Global Medicines Development, AstraZeneca, Gothenburg, Sweden.
| | - Paulo Sérgio Pereira
- Programa de Pós-Graduação em Agroquímica, Instituto Federal Goiano, Rio Verde, GO, Brazil.
| | - David Nascimento Silva Teixeira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Paulo Roberto da Silva
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| | - Deijanira Alves de Albuquerque
- Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil.
| | - Lucia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Alexandre de Paula Rogério
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triângulo Mineiro, Uberaba, MG 38025-350, Brazil.
| |
Collapse
|
10
|
Lu Y, Xu W, Gu Y, Chang X, Wei G, Rong Z, Qin L, Chen X, Zhou F. Non-small Cell Lung Cancer Cells Modulate the Development of Human CD1c + Conventional Dendritic Cell Subsets Mediated by CD103 and CD205. Front Immunol 2019; 10:2829. [PMID: 31921114 PMCID: PMC6914740 DOI: 10.3389/fimmu.2019.02829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced non-small cell lung cancer (NSCLC) leads to a high death rate in patients and is a major threat to human health. NSCLC induces an immune suppressive microenvironment and escapes from immune surveillance in vivo. At present, the molecular mechanisms of NSCLC immunopathogenesis and the immune suppressive microenvironment induced by NSCLC have not been fully elucidated. Here, we focus on the effect of NSCLC cells on the development and differentiation of human CD1c+ conventional dendritic cell (DC) subsets mediated by CD205 and CD103. The peripheral blood mononuclear cells (PBMCs) were isolated from NSCLC patients and healthy donors. DCs were induced and cocultured with primary NSCLC cells or tumor cell line H1299. DCs without incubation with tumor cells are control. The protein expression of costimulatory molecules such as CD80 and CD86, HLA-DR, pro-/anti-inflammatory cytokines such as IL-10 and IL-12, and CD205 and CD103 on CD1c+ DCs was detected by flow cytometry. Our data revealed two new subpopulations of human CD1c+ DCs (CD1c+CD205+CD103+ and CD1c+CD205+CD103− DC) in healthy donors and NSCLC patients. NSCLC cells modulate the development of the CD1c+CD205+CD103+ DC and CD1c+CD205+CD103− DC subpopulations in vitro and ex vivo. NSCLC cells also suppress the expression of signal molecules such as CD40, CD80, CD86, and HLA-DR on CD1c+ DCs. In addition, the production of pro-inflammatory cytokines, including IL-12 and IL-23, is downregulated by NSCLC cells; however, the secretion of anti-inflammatory cytokines, such as IL-10 and IL-27, by CD1c+ DCs is upregulated by NSCLC cells. Our results suggest that NSCLC cells may induce immune tolerogenic DCs, which block DC-mediated anti-tumor immunity in NSCLC patients. Our data may be helpful in revealing new cellular mechanisms related to the induction of tolerogenic CD1c+ DCs by NSCLCs and the development of an immune suppressive microenvironment that causes tumor cells to escape immune surveillance. Our results indicate a potential role for CD1c+ DC subsets mediated by CD205 and CD103 in DC-mediated immunotherapy to target NSCLC in the future.
Collapse
Affiliation(s)
- Yong Lu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Wenlong Xu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Yanli Gu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Xu Chang
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Guojian Wei
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Zhien Rong
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Li Qin
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Xiaoping Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China.,Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fang Zhou
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| |
Collapse
|
11
|
Ritprajak P, Kaewraemruaen C, Hirankarn N. Current Paradigms of Tolerogenic Dendritic Cells and Clinical Implications for Systemic Lupus Erythematosus. Cells 2019; 8:cells8101291. [PMID: 31640263 PMCID: PMC6830089 DOI: 10.3390/cells8101291] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/05/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
Tolerogenic dendritic cells (tolDCs) are central players in the initiation and maintenance of immune tolerance and subsequent prevention of autoimmunity. Recent advances in treatment of autoimmune diseases including systemic lupus erythematosus (SLE) have focused on inducing specific tolerance to avoid long-term use of immunosuppressive drugs. Therefore, DC-targeted therapies to either suppress DC immunogenicity or to promote DC tolerogenicity are of high interest. This review describes details of the typical characteristics of in vivo and ex vivo tolDC, which will help to select a protocol that can generate tolDC with high functional quality for clinical treatment of autoimmune disease in individual patients. In addition, we discuss the recent studies uncovering metabolic pathways and their interrelation intertwined with DC tolerogenicity. This review also highlights the clinical implications of tolDC-based therapy for SLE treatment, examines the current clinical therapeutics in patients with SLE, which can generate tolDC in vivo, and further discusses on possibility and limitation on each strategy. This synthesis provides new perspectives on development of novel therapeutic approaches for SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand.
- Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Chamraj Kaewraemruaen
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand.
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
12
|
Ahad A, Stevanin M, Smita S, Mishra GP, Gupta D, Waszak S, Sarkar UA, Basak S, Gupta B, Acha-Orbea H, Raghav SK. NCoR1: Putting the Brakes on the Dendritic Cell Immune Tolerance. iScience 2019; 19:996-1011. [PMID: 31522122 PMCID: PMC6744395 DOI: 10.1016/j.isci.2019.08.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/19/2019] [Accepted: 08/13/2019] [Indexed: 01/24/2023] Open
Abstract
Understanding the mechanisms fine-tuning immunogenic versus tolerogenic balance in dendritic cells (DCs) is of high importance for therapeutic approaches. We found that NCoR1-mediated direct repression of the tolerogenic program in conventional DCs is essential for induction of an optimal immunogenic response. NCoR1 depletion upregulated a wide variety of tolerogenic genes in activated DCs, which consequently resulted in increased frequency of FoxP3+ regulatory T cells. Mechanistically, NCoR1 masks the PU.1-bound super-enhancers on major tolerogenic genes after DC activation that are subsequently bound by nuclear factor-κB. NCoR1 knockdown (KD) reduced RelA nuclear translocation and activity, whereas RelB was unaffected, providing activated DCs a tolerogenic advantage. Moreover, NCoR1DC−/- mice depicted enhanced Tregs in draining lymph nodes with increased disease burden upon bacterial and parasitic infections. Besides, adoptive transfer of activated NCoR1 KD DCs in infected animals showed a similar phenotype. Collectively, our results demonstrated NCoR1 as a promising target to control DC-mediated immune tolerance. NCoR1 directly represses tolerogenic program in mouse cDCs Depletion of NCoR1 in cDCs enhanced Treg development ex vivo and in vivo NCoR1 masks PU.1-bound super-enhancers on tolerogenic genes in cDCs NCoR1DC−/− animals depicted enhanced Treg frequency and infection load
Collapse
Affiliation(s)
- Abdul Ahad
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Mathias Stevanin
- Department of Biochemistry CIIL, University of Lausanne (UNIL), Epalinges CH-1066, Switzerland
| | - Shuchi Smita
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Gyan Prakash Mishra
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India; Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024, India
| | - Dheerendra Gupta
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India
| | - Sebastian Waszak
- European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Uday Aditya Sarkar
- Systems Immunology Laboratory, National Institute of Immunology (NII), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology (NII), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Bhawna Gupta
- Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024, India
| | - Hans Acha-Orbea
- Department of Biochemistry CIIL, University of Lausanne (UNIL), Epalinges CH-1066, Switzerland.
| | - Sunil Kumar Raghav
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India; Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
13
|
Mesenchymal stem cell therapy induces FLT3L and CD1c + dendritic cells in systemic lupus erythematosus patients. Nat Commun 2019; 10:2498. [PMID: 31175312 PMCID: PMC6555800 DOI: 10.1038/s41467-019-10491-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 05/14/2019] [Indexed: 02/06/2023] Open
Abstract
Allogeneic mesenchymal stem cells (MSCs) exhibit immunoregulatory function in human autoimmune diseases such as systemic lupus erythematosus (SLE), but the underlying mechanisms remain incompletely understood. Here we show that the number of peripheral tolerogenic CD1c+ dendritic cells (DCs) and the levels of serum FLT3L are significantly decreased in SLE patients especially with lupus nephritis, compared to healthy controls. Transplantation of allogeneic umbilical cord-derived MSCs (UC-MSCs) significantly up-regulates peripheral blood CD1c+DCs and serum FLT3L. Mechanistically, UC-MSCs express FLT3L that binds to FLT3 on CD1c+DCs to promote the proliferation and inhibit the apoptosis of tolerogenic CD1c+DCs. Conversely, reduction of FLT3L with small interfering RNA in MSCs abolishes the up-regulation of tolerogenic CD1c+DCs in lupus patients treated with MSCs. Interferon-γ induces FLT3L expression in UC-MSCs through JAK/STAT signaling pathway. Thus, allogeneic MSCs might suppress inflammation in lupus through up-regulating tolerogenic DCs. Promising pilot clinical trials of mesenchymal stem cells (MSCs) therapy of lupus await validation in larger, controlled trials. Here the authors show that MSCs expand CD1c+ dendritic cells in cell culture by producing FLT3L, and that in lupus patients, circulating CD1c+ dendritic cells and FLT3L are increased following MSCs therapy.
Collapse
|
14
|
van der Burg NMD, Depelsenaire ACI, Crichton ML, Kuo P, Phipps S, Kendall MAF. A low inflammatory, Langerhans cell-targeted microprojection patch to deliver ovalbumin to the epidermis of mouse skin. J Control Release 2019; 302:190-200. [PMID: 30940498 DOI: 10.1016/j.jconrel.2019.03.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/20/2019] [Accepted: 03/29/2019] [Indexed: 12/21/2022]
Abstract
In a low inflammatory skin environment, Langerhans cells (LCs) - but not dermal dendritic cells (dDCs) - contribute to the pivotal process of tolerance induction. Thus LCs are a target for specific-tolerance therapies. LCs reside just below the stratum corneum, within the skin's viable epidermis. One way to precisely deliver immunotherapies to LCs while remaining minimally invasive is with a skin delivery device such as a microprojection arrays (MPA). Today's MPAs currently achieve rapid delivery (e.g. within minutes of application), but are focussed primarily at delivery of therapeutics to the dermis, deeper within the skin. Indeed, no MPA currently delivers specifically to the epidermal LCs of mouse skin. Without any convenient, pre-clinical device available, advancement of LC-targeted therapies has been limited. In this study, we designed and tested a novel MPA that delivers ovalbumin to the mouse epidermis (eMPA) while maintaining a low, local inflammatory response (as defined by low erythema after 24 h). In comparison to available dermal-targeted MPAs (dMPA), only eMPAs with larger projection tip surface areas achieved shallow epidermal penetration at a low application energy. The eMPA characterised here induced significantly less erythema after 24 h (p = 0.0004), less epidermal swelling after 72 h (p < 0.0001) and 52% less epidermal cell death than the dMPA. Despite these differences in skin inflammation, the eMPA and dMPA promoted similar levels of LC migration out of the skin. However, only the eMPA promoted LCs to migrate with a low MHC II expression and in the absence of dDC migration. Implementing this more mouse-appropriate and low-inflammatory eMPA device to deliver potential immunotherapeutics could improve the practicality and cell-specific targeting of such therapeutics in the pre-clinical stage. Leading to more opportunities for LC-targeted therapeutics such as for allergy immunotherapy and asthma.
Collapse
Affiliation(s)
- Nicole M D van der Burg
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Alexandra C I Depelsenaire
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Michael L Crichton
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Paula Kuo
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QL 4102, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, QL 4006, Australia
| | - Mark A F Kendall
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia; The Australian National University, Canberra, Australian Capital Territory 2600, Australia.
| |
Collapse
|
15
|
Zhou F, Zhang GX, Rostami A. Distinct Role of IL-27 in Immature and LPS-Induced Mature Dendritic Cell-Mediated Development of CD4 + CD127 +3G11 + Regulatory T Cell Subset. Front Immunol 2018; 9:2562. [PMID: 30483251 PMCID: PMC6244609 DOI: 10.3389/fimmu.2018.02562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
Interleukin-27 (IL-27) plays an important role in regulation of anti-inflammatory responses and autoimmunity; however, the molecular mechanisms of IL-27 in modulation of immune tolerance and autoimmunity have not been fully elucidated. Dendritic cells (DCs) play a central role in regulating immune responses mediated by innate and adaptive immune systems, but regulatory mechanisms of DCs in CD4+ T cell-mediated immune responses have not yet been elucidated. Here we show that IL-27 treated mature DCs induced by LPS inhibit immune tolerance mediated by LPS-stimulated DCs. IL-27 treatment facilitates development of the CD4+ CD127+3G11+ regulatory T cell subset in vitro and in vivo. By contrast, IL-27 treated immature DCs fail to modulate development of the CD4+CD127+3G11+ regulatory T cell sub-population in vitro and in vivo. Our results suggest that IL-27 may break immune tolerance induced by LPS-stimulated mature DCs through modulating development of a specific CD4+ regulatory T cell subset mediated by 3G11 and CD127. Our data reveal a new cellular regulatory mechanism of IL-27 that targets DC-mediated immune responses in autoimmune diseases such as multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE).
Collapse
Affiliation(s)
- Fang Zhou
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
16
|
Sun W, Wei JW, Li H, Wei FQ, Li J, Wen WP. Adoptive cell therapy of tolerogenic dendritic cells as inducer of regulatory T cells in allergic rhinitis. Int Forum Allergy Rhinol 2018; 8:1291-1299. [PMID: 30281915 DOI: 10.1002/alr.22217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/22/2018] [Accepted: 09/09/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Wei Sun
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Jia-Wei Wei
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Hang Li
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Fan-Qin Wei
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Jian Li
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Wei-Ping Wen
- Department of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Guangzhou Key Laboratory of Otorhinolaryngology-Head and Neck Surgery; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| |
Collapse
|
17
|
Kerdidani D, Magkouta S, Chouvardas P, Karavana V, Glynos K, Roumelioti F, Zakynthinos S, Wauters E, Janssens W, Lambrechts D, Kollias G, Tsoumakidou M. Cigarette Smoke-Induced Emphysema Exhausts Early Cytotoxic CD8 + T Cell Responses against Nascent Lung Cancer Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1558-1569. [PMID: 30037849 DOI: 10.4049/jimmunol.1700700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/25/2018] [Indexed: 01/08/2023]
Abstract
Chronic obstructive pulmonary disease is a chronic inflammatory disorder with an increased incidence of lung cancer. The emphysema component of chronic obstructive pulmonary disease confers the greatest proportion to lung cancer risk. Although tumors create inflammatory conditions to escape immunity, the immunological responses that control growth of nascent cancer cells in pre-established inflammatory microenvironments are unknown. In this study, we addressed this issue by implanting OVA-expressing cancer cells in the lungs of mice with cigarette smoke-induced emphysema. Emphysema augmented the growth of cancer cells, an effect that was dependent on T cytotoxic cells. OVA-specific OTI T cells showed early signs of exhaustion upon transfer in emphysema tumor hosts that was largely irreversible because sorting, expansion, and adoptive transfer failed to restore their antitumor activity. Increased numbers of PD-L1- and IDO-positive CD11c+ myeloid dendritic cells (DCs) infiltrated emphysema tumors, whereas sorted emphysema tumor DCs poorly stimulated OTI T cells. Upon adoptive transfer in immunocompetent hosts, T cells primed by emphysema tumor DCs were unable to halt tumor growth. DCs exposed to the emphysema tumor microenvironment downregulated MHC class II and costimulatory molecules, whereas they upregulated PD-L1/IDO via oxidative stress-dependent mechanisms. T cell activation increased upon PD-L1 blockade in emphysema DC-T cell cocultures and in emphysema tumor hosts in vivo. Analysis of the transcriptome of primary human lung tumors showed a strong association between computed tomography-based emphysema scoring and downregulation of immunogenic processes. Thus, suppression of adaptive immunity against lung cancer cells links a chronic inflammatory disorder, emphysema, to cancer, with clinical implications for emphysema patients to be considered optimal candidates for cancer immunotherapies.
Collapse
Affiliation(s)
- Dimitra Kerdidani
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Sophia Magkouta
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Panagiotis Chouvardas
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Vassiliki Karavana
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Konstantinos Glynos
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Fani Roumelioti
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece
| | - Spyros Zakynthinos
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Els Wauters
- Respiratory Oncology Unit, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Leuven Lung Cancer Group, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Wim Janssens
- Respiratory Oncology Unit, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Leuven Lung Cancer Group, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium; and.,Laboratory for Translational Genetics, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - George Kollias
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Maria Tsoumakidou
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece;
| |
Collapse
|
18
|
Ni L, Dong C. Roles of Myeloid and Lymphoid Cells in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Front Immunol 2018; 9:1431. [PMID: 29977245 PMCID: PMC6021485 DOI: 10.3389/fimmu.2018.01431] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the third largest cause of human mortality in the world after stroke and heart disease. COPD is characterized by sustained inflammation of the airways, leading to destruction of lung tissue and declining pulmonary function. The main risk factor is known to be cigarette smoke currently. However, the strategies for prevention and treatment have not altered significantly for many years. A growing body of evidences indicates that the immune system plays a pivotal role in the pathogenesis of COPD. The repeated and progressive activation of immune cells is at least in part the source of this chronic inflammation. In this review paper, we have conducted an extensive literature search of the studies of immune cells in COPD patients. The objective is to assess the contributions of different immune cell types, the imbalance of pro/anti-inflammatory immune cells, such as M1/M2 macrophages, Tc1/Tc10, and Th17/Treg, and their mediators in the peripheral blood as well as in the lung to the pathogenesis of COPD. Therefore, understanding their roles in COPD development will help us find the potential target to modify this disease. This review focuses predominantly on data derived from human studies but will refer to animal studies where they help understand the disease in humans.
Collapse
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
19
|
Dysregulated Functions of Lung Macrophage Populations in COPD. J Immunol Res 2018; 2018:2349045. [PMID: 29670919 PMCID: PMC5835245 DOI: 10.1155/2018/2349045] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/29/2017] [Indexed: 01/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a diverse respiratory disease characterised by bronchiolitis, small airway obstruction, and emphysema. Innate immune cells play a pivotal role in the disease's progression, and in particular, lung macrophages exploit their prevalence and strategic localisation to orchestrate immune responses. To date, alveolar and interstitial resident macrophages as well as blood monocytes have been described in the lungs of patients with COPD contributing to disease pathology by changes in their functional repertoire. In this review, we summarise recent evidence from human studies and work with animal models of COPD with regard to altered functions of each of these myeloid cell populations. We primarily focus on the dysregulated capacity of alveolar macrophages to secrete proinflammatory mediators and proteases, induce oxidative stress, engulf microbes and apoptotic cells, and express surface and intracellular markers in patients with COPD. In addition, we discuss the differences in the responses between alveolar macrophages and interstitial macrophages/monocytes in the disease and propose how the field should advance to better understand the implications of lung macrophage functions in COPD.
Collapse
|
20
|
Alivernini S, Tolusso B, Ferraccioli G, Gremese E, Kurowska-Stolarska M, McInnes IB. Driving chronicity in rheumatoid arthritis: perpetuating role of myeloid cells. Clin Exp Immunol 2018; 193:13-23. [PMID: 29315512 PMCID: PMC6038003 DOI: 10.1111/cei.13098] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022] Open
Abstract
Acute inflammation is a complex and tightly regulated homeostatic process that includes leucocyte migration from the vasculature into tissues to eliminate the pathogen/injury, followed by a pro‐resolving response promoting tissue repair. However, if inflammation is uncontrolled as in chronic diseases such as rheumatoid arthritis (RA), it leads to tissue damage and disability. Synovial tissue inflammation in RA patients is maintained by sustained activation of multiple inflammatory positive‐feedback regulatory pathways in a variety of cells, including myeloid cells. In this review, we will highlight recent evidence uncovering biological mechanisms contributing to the aberrant activation of myeloid cells that contributes to perpetuation of inflammation in RA, and discuss emerging data on anti‐inflammatory mediators contributing to sustained remission that may inform a novel category of therapeutic targets.
Collapse
Affiliation(s)
- S Alivernini
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - B Tolusso
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - G Ferraccioli
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - E Gremese
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - M Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| | - I B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| |
Collapse
|
21
|
Obregon C, Kumar R, Pascual MA, Vassalli G, Golshayan D. Update on Dendritic Cell-Induced Immunological and Clinical Tolerance. Front Immunol 2017; 8:1514. [PMID: 29250057 PMCID: PMC5715373 DOI: 10.3389/fimmu.2017.01514] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) as highly efficient antigen-presenting cells are at the interface of innate and adaptive immunity. As such, they are key mediators of immunity and antigen-specific immune tolerance. Due to their functional specialization, research efforts have focused on the characterization of DCs subsets involved in the initiation of immunogenic responses and in the maintenance of tissue homeostasis. Tolerogenic DCs (tolDCs)-based therapies have been designed as promising strategies to prevent and control autoimmune diseases as well as allograft rejection after solid organ transplantation (SOT). Despite successful experimental studies and ongoing phase I/II clinical trials using autologous tolDCs in patients with type 1 diabetes, rheumatoid arthritis, multiple sclerosis, and in SOT recipients, additional basic research will be required to determine the optimal DC subset(s) and conditioning regimens for tolDCs-based treatments in vivo. In this review, we discuss the characteristics of human DCs and recent advances in their classification, as well as the role of DCs in immune regulation and their susceptibility to in vitro or in vivo manipulation for the development of tolerogenic therapies, with a focus on the potential of tolDCs for the treatment of autoimmune diseases and the prevention of allograft rejection after SOT.
Collapse
Affiliation(s)
- Carolina Obregon
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Rajesh Kumar
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Manuel Antonio Pascual
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.,Department of Surgery, Transplantation Centre, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Vassalli
- Département coeur-vaisseaux, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.,Fondazione Cardiocentro Ticino, Swiss Institute of Regenerative Medicine (SIRM), Lugano, Switzerland
| | - Déla Golshayan
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.,Department of Surgery, Transplantation Centre, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
22
|
Sendo S, Saegusa J, Okano T, Takahashi S, Akashi K, Morinobu A. CD11b+Gr-1 dim Tolerogenic Dendritic Cell-Like Cells Are Expanded in Interstitial Lung Disease in SKG Mice. Arthritis Rheumatol 2017; 69:2314-2327. [PMID: 28805019 DOI: 10.1002/art.40231] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 08/09/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE SKG mice develop interstitial lung disease (ILD) resembling rheumatoid arthritis-associated ILD in humans. The aim of this study was to clarify the mechanism underlying the lung pathology by analyzing lung-infiltrating cells in SKG mice with ILD. METHODS We assessed the severity of zymosan A (ZyA)-induced ILD in SKG mice histologically, and we examined lung-infiltrating cells by flow cytometry. Total lung cells and isolated monocytic myeloid-derived suppressor cells (MDSCs) were cultured in vitro with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4. The proliferation of 5,6-carboxyfluorescein diacetate N-succinimidyl ester-labeled naive T cells cocultured with isolated CD11b+Gr-1dim cells and MDSCs was evaluated by flow cytometry. CD11b+Gr-1dim cells were adoptively transferred to ZyA-treated SKG mice. RESULTS MDSCs, Th17 cells, and group 1 and 3 innate lymphoid cells (ILC1s and ILC3s) were increased in the lungs; the proportion of these cells varied with ILD severity. In this process, we found that a unique cell population, CD11b+Gr-1dim cells, was expanded in the severely inflamed lungs. Approximately half of the CD11b+Gr-1dim cells expressed CD11c. CD11b+Gr-1dim cells were induced from monocytic MDSCs with GM-CSF in vitro and were considered tolerogenic because they suppressed T cell proliferation. These CD11b+Gr-1dim cells have never been described previously, and we termed them CD11b+Gr-1dim tolerogenic dendritic cell (DC)-like cells. Th17 cells, ILC1s, and ILC3s in the inflamed lung produced GM-CSF, which may have expanded CD11b+Gr-1dim tolerogenic DC-like cells in vivo. Furthermore, adoptive transfer of CD11b+Gr-1dim tolerogenic DC-like cells significantly suppressed progression of ILD in SKG mice. CONCLUSION We identified unique suppressive myeloid cells that were differentiated from monocytic MDSCs in SKG mice with ILD, and we termed them CD11b+Gr-1dim tolerogenic DC-like cells.
Collapse
Affiliation(s)
- Sho Sendo
- Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Saegusa
- Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takaichi Okano
- Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Kengo Akashi
- Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akio Morinobu
- Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
23
|
Baharom F, Rankin G, Blomberg A, Smed-Sörensen A. Human Lung Mononuclear Phagocytes in Health and Disease. Front Immunol 2017; 8:499. [PMID: 28507549 PMCID: PMC5410584 DOI: 10.3389/fimmu.2017.00499] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022] Open
Abstract
The lungs are vulnerable to attack by respiratory insults such as toxins, allergens, and pathogens, given their continuous exposure to the air we breathe. Our immune system has evolved to provide protection against an array of potential threats without causing collateral damage to the lung tissue. In order to swiftly detect invading pathogens, monocytes, macrophages, and dendritic cells (DCs)-together termed mononuclear phagocytes (MNPs)-line the respiratory tract with the key task of surveying the lung microenvironment in order to discriminate between harmless and harmful antigens and initiate immune responses when necessary. Each cell type excels at specific tasks: monocytes produce large amounts of cytokines, macrophages are highly phagocytic, whereas DCs excel at activating naïve T cells. Extensive studies in murine models have established a division of labor between the different populations of MNPs at steady state and during infection or inflammation. However, a translation of important findings in mice is only beginning to be explored in humans, given the challenge of working with rare cells in inaccessible human tissues. Important progress has been made in recent years on the phenotype and function of human lung MNPs. In addition to a substantial population of alveolar macrophages, three subsets of DCs have been identified in the human airways at steady state. More recently, monocyte-derived cells have also been described in healthy human lungs. Depending on the source of samples, such as lung tissue resections or bronchoalveolar lavage, the specific subsets of MNPs recovered may differ. This review provides an update on existing studies investigating human respiratory MNP populations during health and disease. Often, inflammatory MNPs are found to accumulate in the lungs of patients with pulmonary conditions. In respiratory infections or inflammatory diseases, this may contribute to disease severity, but in cancer patients this may improve clinical outcomes. By expanding on this knowledge, specific lung MNPs may be targeted or modulated in order to attain favorable responses that can improve preventive or treatment strategies against respiratory infections, lung cancer, or lung inflammatory diseases.
Collapse
Affiliation(s)
- Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Gregory Rankin
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
24
|
Freeman CM, Curtis JL. Lung Dendritic Cells: Shaping Immune Responses throughout Chronic Obstructive Pulmonary Disease Progression. Am J Respir Cell Mol Biol 2017; 56:152-159. [PMID: 27767327 PMCID: PMC6222925 DOI: 10.1165/rcmb.2016-0272tr] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/12/2016] [Indexed: 12/24/2022] Open
Abstract
Hallmarks of chronic obstructive pulmonary disease (COPD) include innate inflammation and remodeling of small airways, which begin in early disease, and the development of lung lymphoid follicles (LLF), indicative of adaptive immunity, in more spirometrically severe stages. Common to these processes in all stages is orchestration by dendritic cells (DCs). Recently improved understanding of the analogous lung DC subsets in humans and mice has allowed for better integration and interpretation of the experimental and clinical pathological literature. In this review, we summarize the evidence from human and animal studies to place lung DCs into the context of COPD pathogenesis. We highlight recent studies that demonstrate a potential role for DCs in airway remodeling and that call into question the long-standing belief that intraepithelial DCs actively sample airway lumens. We discuss how DCs drive LLF formation directly and indirectly and also examine the ability of DCs within LLF to instruct downstream effector functions of natural killer cells, CD4+ T cells, and regulatory T cells. Greater awareness of the multifaceted functions of DCs will be essential in the quest to identify new therapeutic modalities to treat COPD.
Collapse
Affiliation(s)
- Christine M. Freeman
- Research Service, and
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, and
| | - Jeffrey L. Curtis
- Pulmonary and Critical Care Medicine Section, Medicine Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, and
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
25
|
Jirmo AC, Daluege K, Happle C, Albrecht M, Dittrich AM, Busse M, Habener A, Skuljec J, Hansen G. IL-27 Is Essential for Suppression of Experimental Allergic Asthma by the TLR7/8 Agonist R848 (Resiquimod). THE JOURNAL OF IMMUNOLOGY 2016; 197:4219-4227. [PMID: 27799314 DOI: 10.4049/jimmunol.1601094] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/27/2016] [Indexed: 12/24/2022]
Abstract
Different models of experimental allergic asthma have shown that the TLR7/8 agonist resiquimod (R848) is a potential inhibitor of type 2 helper cell-driven inflammatory responses. However, the mechanisms mediating its therapeutic effects are not fully understood. Using a model of experimental allergic asthma, we show that induction of IL-27 by R848 is critical for the observed ameliorative effects. R848 significantly inhibited all hallmarks of experimental allergic asthma, including airway hyperreactivity, eosinophilic airway inflammation, mucus hypersecretion, and Ag-specific Ig production. Whereas R848 significantly reduced IL-5, IL-13, and IL-17, it induced IFN-γ and IL-27. Neutralization of IL-27 completely reversed the therapeutic effect of R848 in the experimental asthma model, demonstrating dependence of R848-mediated suppression on IL-27. In vitro, R848 induced production of IL-27 by murine alveolar macrophages and dendritic cells and enhanced expression of programmed death-ligand 1, whose expression on monocytes and dendritic cells has been shown to regulate peripheral tolerance in both murine and human studies. Moreover, in vitro IL-27 enhanced secretion of IFN-γ whereas it inhibited IL-5 and IL-13, demonstrating its direct effect on attenuating Th2 responses. Taken together, our study proves that R848-mediated suppression of experimental asthma is dependent on IL-27. These data provide evidence of a central role of IL-27 for the control of Th2-mediated allergic diseases.
Collapse
Affiliation(s)
- Adan Chari Jirmo
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research, 30625 Hannover, Germany
| | - Kathleen Daluege
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research, 30625 Hannover, Germany
| | - Melanie Albrecht
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Mandy Busse
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Anika Habener
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research, 30625 Hannover, Germany
| | - Jelena Skuljec
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research, 30625 Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; and .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research, 30625 Hannover, Germany
| |
Collapse
|
26
|
Upham JW, Xi Y. Dendritic Cells in Human Lung Disease: Recent Advances. Chest 2016; 151:668-673. [PMID: 27729261 DOI: 10.1016/j.chest.2016.09.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/18/2016] [Accepted: 09/29/2016] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells. Because of their particular ability to initiate and regulate cell mediated and humoral immune responses, there is considerable interest in the role that DCs play in the pathogenesis of various lung diseases, especially those in which there is an excessive immune response to specific antigens (as in asthma) or a deficient immune response (as in lung cancer). A number of DC subpopulations have been defined in the lungs, including myeloid or conventional DCs that initiate T-cell immunity and antibody production and plasmacytoid DCs that have an important role in antiviral immunity and immune tolerance. Although an extensive body of literature has documented the role that DCs play in experimental models of lung disease, this review will highlight recent advances in our understanding of DC function in human disease, including asthma, COPD, antimicrobial immunity, and lung cancer. The future is likely to see new approaches whereby antigens and small molecules are targeted to receptors on particular DC subpopulations in order to modify pulmonary immune responses.
Collapse
Affiliation(s)
- John W Upham
- School of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Australia; Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia.
| | - Yang Xi
- School of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Australia
| |
Collapse
|
27
|
Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol 2016; 138:984-1010. [DOI: 10.1016/j.jaci.2016.06.033] [Citation(s) in RCA: 603] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022]
|
28
|
Abstract
Dendritic cells (DCs) lie at the heart of the innate immune system, specialised at recognising danger signals in many forms including foreign material, infection or tissue damage and initiating powerful adaptive immune and inflammatory responses. In barrier sites such as the lung, the instrumental role that DCs play at the interface between the environment and the host places them in a pivotal position in determining the severity of inflammatory disease. The past few years has seen a significant increase in our fundamental understanding of the subsets of DCs involved in pulmonary immunity, as well as the mechanisms by which they are activated and which they may use to coordinate downstream inflammation and pathology. In this review, we will summarise current understanding of the multi-faceted role that DCs play in the induction, maintenance and regulation of lung immunopathology, with an emphasis on allergic pulmonary disease.
Collapse
|
29
|
Baharom F, Thomas S, Rankin G, Lepzien R, Pourazar J, Behndig AF, Ahlm C, Blomberg A, Smed-Sörensen A. Dendritic Cells and Monocytes with Distinct Inflammatory Responses Reside in Lung Mucosa of Healthy Humans. THE JOURNAL OF IMMUNOLOGY 2016; 196:4498-509. [PMID: 27183618 DOI: 10.4049/jimmunol.1600071] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/04/2016] [Indexed: 12/24/2022]
Abstract
Every breath we take contains potentially harmful pathogens or allergens. Dendritic cells (DCs), monocytes, and macrophages are essential in maintaining a delicate balance of initiating immunity without causing collateral damage to the lungs because of an exaggerated inflammatory response. To document the diversity of lung mononuclear phagocytes at steady-state, we performed bronchoscopies on 20 healthy subjects, sampling the proximal and distal airways (bronchial wash and bronchoalveolar lavage, respectively), as well as mucosal tissue (endobronchial biopsies). In addition to a substantial population of alveolar macrophages, we identified subpopulations of monocytes, myeloid DCs (MDCs), and plasmacytoid DCs in the lung mucosa. Intermediate monocytes and MDCs were highly frequent in the airways compared with peripheral blood. Strikingly, the density of mononuclear phagocytes increased upon descending the airways. Monocytes from blood and airways produced 10-fold more proinflammatory cytokines than MDCs upon ex vivo stimulation. However, airway monocytes were less inflammatory than blood monocytes, suggesting a more tolerant nature. The findings of this study establish how to identify human lung mononuclear phagocytes and how they function in normal conditions, so that dysregulations in patients with respiratory diseases can be detected to elucidate their contribution to immunity or pathogenesis.
Collapse
Affiliation(s)
- Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Saskia Thomas
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Gregory Rankin
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Rico Lepzien
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Jamshid Pourazar
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Annelie F Behndig
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Clas Ahlm
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, 901 85 Umeå, Sweden
| | - Anders Blomberg
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden;
| |
Collapse
|
30
|
Wawrzyniak P, Akdis CA, Finkelman FD, Rothenberg ME. Advances and highlights in mechanisms of allergic disease in 2015. J Allergy Clin Immunol 2016; 137:1681-1696. [PMID: 27090934 DOI: 10.1016/j.jaci.2016.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/03/2016] [Accepted: 02/17/2016] [Indexed: 01/08/2023]
Abstract
This review highlights some of the advances in mechanisms of allergic disease, particularly anaphylaxis, including food allergy, drug hypersensitivity, atopic dermatitis (AD), allergic conjunctivitis, and airway diseases. During the last year, a mechanistic advance in food allergy was achieved by focusing on mechanisms of allergen sensitization. Novel biomarkers and treatment for mastocytosis were presented in several studies. Novel therapeutic approaches in the treatment of atopic dermatitis and psoriasis showed that promising supplementation of the infant's diet in the first year of life with immunoactive prebiotics might have a preventive role against early development of AD and that therapeutic approaches to treat AD in children might be best directed to the correction of a TH2/TH1 imbalance. Several studies were published emphasizing the role of the epithelial barrier in patients with allergic diseases. An impaired skin barrier as a cause for sensitization to food allergens in children and its relationship to filaggrin mutations has been an important development. Numerous studies presented new approaches for improvement of epithelial barrier function and novel biologicals used in the treatment of inflammatory skin and eosinophilic diseases. In addition, novel transcription factors and signaling molecules that can develop as new possible therapeutic targets have been reported.
Collapse
Affiliation(s)
- Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| | - Fred D Finkelman
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, and the Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
31
|
O'Keeffe M, Mok WH, Radford KJ. Human dendritic cell subsets and function in health and disease. Cell Mol Life Sci 2015; 72:4309-25. [PMID: 26243730 PMCID: PMC11113503 DOI: 10.1007/s00018-015-2005-0] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/15/2015] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
Abstract
The method of choice for the development of new vaccines is to target distinct dendritic cell subsets with antigen in vivo and to harness their function in situ to enhance cell-mediated immunity or induce tolerance to specific antigens. The innate functions of dendritic cells themselves may also be targeted by inhibitors or activators that would target a specific function such as interferon production, potentially important in autoimmune disease and chronic viral infections. Importantly targeting dendritic cells requires detailed knowledge of both the surface phenotype and function of each dendritic cell subset, including how they may respond to different types of vaccine adjuvants, their ability to produce soluble mediators and to process and present antigens and induce priming of naïve T cells. This review summarizes our knowledge of the functional attributes of the human dendritic cell subsets in the steady state and upon activation and their roles in human disease.
Collapse
Affiliation(s)
- Meredith O'Keeffe
- Centre for Biomedical Research, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Clayton, VIC, 3800, Australia
| | - Wai Hong Mok
- Mater Research Institute, University of Queensland, 37 Kent St, Woolloongabba, QLD, 4012, Australia
| | - Kristen J Radford
- Mater Research Institute, University of Queensland, 37 Kent St, Woolloongabba, QLD, 4012, Australia.
| |
Collapse
|
32
|
The role of IL-10 in microbiome-associated immune modulation and disease tolerance. Cytokine 2015; 75:291-301. [DOI: 10.1016/j.cyto.2014.11.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 02/06/2023]
|