1
|
Li Z, Xiong W, Liang Z, Wang J, Zeng Z, Kołat D, Li X, Zhou D, Xu X, Zhao L. Critical role of the gut microbiota in immune responses and cancer immunotherapy. J Hematol Oncol 2024; 17:33. [PMID: 38745196 PMCID: PMC11094969 DOI: 10.1186/s13045-024-01541-w] [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: 10/25/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
The gut microbiota plays a critical role in the progression of human diseases, especially cancer. In recent decades, there has been accumulating evidence of the connections between the gut microbiota and cancer immunotherapy. Therefore, understanding the functional role of the gut microbiota in regulating immune responses to cancer immunotherapy is crucial for developing precision medicine. In this review, we extract insights from state-of-the-art research to decipher the complicated crosstalk among the gut microbiota, the systemic immune system, and immunotherapy in the context of cancer. Additionally, as the gut microbiota can account for immune-related adverse events, we discuss potential interventions to minimize these adverse effects and discuss the clinical application of five microbiota-targeted strategies that precisely increase the efficacy of cancer immunotherapy. Finally, as the gut microbiota holds promising potential as a target for precision cancer immunotherapeutics, we summarize current challenges and provide a general outlook on future directions in this field.
Collapse
Affiliation(s)
- Zehua Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, England
| | - Weixi Xiong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Brain Science and Brain-Inspired Technology of West China Hospital, Sichuan University, Chengdu, China
| | - Zhu Liang
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, England
- Target Discovery Institute, Center for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, England
| | - Jinyu Wang
- Departments of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Ziyi Zeng
- Department of Neonatology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Xi Li
- Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Brain Science and Brain-Inspired Technology of West China Hospital, Sichuan University, Chengdu, China
| | - Xuewen Xu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Linyong Zhao
- Department of General Surgery and Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Zahorchak AF, DeRiggi ML, Muzzio JL, Sutherland V, Humar A, Lakkis FG, Hsu YMS, Thomson AW. Manufacturing and validation of Good Manufacturing Practice-compliant regulatory dendritic cells for infusion into organ transplant recipients. Cytotherapy 2023; 25:432-441. [PMID: 36639251 DOI: 10.1016/j.jcyt.2022.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/28/2022] [Accepted: 11/18/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AIMS Regulatory (or "tolerogenic") dendritic cells (DCregs) are a highly promising, innovative cell therapy for the induction or restoration of antigen-specific tolerance in immune-mediated inflammatory disorders. These conditions include organ allograft rejection, graft-versus-host disease following bone marrow transplantation and various autoimmune disorders. DCregs generated for adoptive transfer have potential to reduce patients' dependence on non-specific immunosuppressive drugs that can induce serious side effects and enhance the risk of infection and certain types of cancer. Here, our aim was to provide a detailed account of our experience manufacturing and validating comparatively large numbers of Good Manufacturing Practice-grade DCregs for systemic (intravenous) infusion into 28 organ (liver) transplant recipients and to discuss factors that influence the satisfaction of release criteria and attainment of target cell numbers. RESULTS DCregs were generated in granulocyte-macrophage colony stimulating factor and interleukin (IL)-4 from elutriated monocyte fractions isolated from non-mobilized leukapheresis products of consenting healthy adult prospective liver transplant donors. Vitamin D3 was added on day 0 and 4 and IL-10 on day 4 during the 7-day culture period. Release and post-release criteria included cell viability, purity, phenotype, sterility and functional assessment. The overall conversion rate of monocytes to DCregs was 28 ± 8.2%, with 94 ± 5.1% product viability. The mean cell surface T-cell co-inhibitory to co-stimulatory molecule (programmed death ligand-1:CD86) mean fluorescence intensity ratio was 3.9 ± 2.2, and the mean ratio of anti-inflammatory:pro-inflammatory cytokine product (IL-10:IL-12p70) secreted upon CD40 ligation was 60 ± 63 (median = 40). The mean total number of DCregs generated from a single leukapheresis product (n = 25 donors) and from two leukapheresis products (n = 3 donors) was 489 ± 223 × 106 (n = 28). The mean total number of DCregs infused was 5.9 ± 2.8 × 106 per kg body weight. DCreg numbers within a target cell range of 2.5-10 × 106/kg were achieved for 25 of 27 (92.6%) of products generated. CONCLUSIONS High-purity DCregs meeting a range of quality criteria were readily generated from circulating blood monocytes under Good Manufacturing Practice conditions to meet target cell numbers for infusion into prospective organ transplant recipients.
Collapse
Affiliation(s)
- Alan F Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Misty L DeRiggi
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jennifer L Muzzio
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Veronica Sutherland
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Abhinav Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fadi G Lakkis
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yen-Michael S Hsu
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA; Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
3
|
Li J, Zhao C, Xu Y, Song L, Chen Y, Xu Y, Ma Y, Wang S, Xu A, He F. Remodeling of the osteoimmune microenvironment after biomaterials implantation in murine tibia: Single-cell transcriptome analysis. Bioact Mater 2023; 22:404-422. [PMID: 36311047 PMCID: PMC9588995 DOI: 10.1016/j.bioactmat.2022.10.009] [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: 07/04/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 11/12/2022] Open
Abstract
Osseointegration seems to be a foreign body reaction equilibrium due to the complicated interactions between the immune and skeletal systems. The heterogeneity of the osteoimmune microenvironment in the osseointegration of implant materials remains elusive. Here, a single-cell study involving 40043 cells is conducted, and a total of 10 distinct cell clusters are identified from five different groups. A preliminary description of the osteoimmune microenvironment revealed the diverse cellular heterogeneity and dynamic changes modulated by implant properties. The increased immature neutrophils, Ly6C + CCR2hi monocytes, and S100a8hi macrophages induce an aggressive inflammatory response and eventually lead to the formation of fibrous capsule around the stainless steel implant. The enrichment of mature neutrophils, FcgR1hi and differentiated immunomodulatory macrophages around the titanium implant indicates favorable osseointegration under moderate immune response. Neutrophil-depletion mice are conducted to explore the role of neutrophils in osseointegration. Neutrophils may improve bone formation by enhancing the recruitment of BMSCs via the CXCL12/CXCR3 signal axis. These findings contribute to a better knowledge of osteoimmunology and are valuable for the design and modification of 'osteoimmune-smart' biomaterials in the bone regeneration field.
Collapse
Key Words
- BMP2, Bone Morphogenetic Proteins 2
- CXCL12, Chemokine (C-X-C mode) Ligand 12
- CXCR, CXC Chemokine Receptor
- FcgR, Fc Gamma Receptor
- IFN-γ, Interferon-gamma
- IL-1β, Interleukin-1 beta
- Implant
- MHC, Major Histocompatibility Complex
- MIP, Macrophage inflammatory cytokines
- MPO, Myeloperoxidase
- NE, Neutrophil Elastase
- NF-κB, Nuclear Factor Kappa-light-chain-enhancer of Activated B cells
- NOD, Nucleotide Binding Oligomerization Domain
- Neutrophil
- OPG, Osteoprotegerin
- Osseointegration
- Osteoimmunology
- RANKL, Nuclear Factor B receptor Activator Ligand
- RUNX2, Runt-related Transcription Factor 2
- S100a8, S100 Calcium Binding Protein A8
- SDF-1α, Stromal Cell-derived Factor-1 alpha
- STAT, Signal Transduction and Transcription Activator
- Single-cell transcriptomics
- TLR, Toll Like Receptor
- TNFα, Tumor Necrosis Factor-alpha
- TRAP, Tartrate Resistant Acid Phosphatase
Collapse
Affiliation(s)
- Jia Li
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Congrui Zhao
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yangbo Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Lu Song
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yanqi Chen
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yuzi Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yang Ma
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Antian Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Fuming He
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W, Sun Z. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology 2021; 164:476-493. [PMID: 34322877 PMCID: PMC8517597 DOI: 10.1111/imm.13397] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.
Collapse
Affiliation(s)
- Xiuxiu Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of MedicineZhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yaxin Guo
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Bo Shao
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Luyang Zhao
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinbo Liu
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guixian Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenqiang Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
5
|
Gold Nanoparticles: Multifaceted Roles in the Management of Autoimmune Disorders. Biomolecules 2021; 11:biom11091289. [PMID: 34572503 PMCID: PMC8470500 DOI: 10.3390/biom11091289] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Gold nanoparticles (GNPs) have been recently applied for various diagnostic and therapeutic purposes. The unique properties of these nanoparticles (NPs), such as relative ease of synthesis in various sizes, shapes and charges, stability, high drug-loading capacity and relative availability for modification accompanied by non-cytotoxicity and biocompatibility, make them an ideal field of research in bio-nanotechnology. Moreover, their potential to alleviate various inflammatory factors, nitrite species, and reactive oxygen production and the capacity to deliver therapeutic agents has attracted attention for further studies in inflammatory and autoimmune disorders. Furthermore, the characteristics of GNPs and surface modification can modulate their toxicity, biodistribution, biocompatibility, and effects. This review discusses in vitro and in vivo effects of GNPs and their functionalized forms in managing various autoimmune disorders (Ads) such as rheumatoid arthritis, type 1 diabetes, and multiple sclerosis.
Collapse
|
6
|
Muth S, Klaric A, Radsak M, Schild H, Probst HC. CD27 expression on Treg cells limits immune responses against tumors. J Mol Med (Berl) 2021; 100:439-449. [PMID: 34423375 PMCID: PMC8843905 DOI: 10.1007/s00109-021-02116-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Regulatory T cells (Tregs) suppress immune responses and thus contribute to immune homeostasis. On the downside, Tregs also limit immune responses against tumors promoting the progression of cancer. Among the many mechanisms implied in Treg-mediated suppression, the inhibition of dendritic cells (DCs) has been shown to be central in peripheral tolerance induction as well as in cancers. We have shown previously that the maintenance of peripheral T cell tolerance critically depends on cognate interactions between Tregs and DCs and that the CTL priming by unsuppressed steady state DCs is mediated via CD70. Here, we have investigated whether the CD70/CD27 axis is also involved in Treg-mediated suppression of anti-tumor immunity. Using a mixed bone marrow chimeric mouse model in which we can deplete regulatory T cells in a temporally controlled fashion, we show that Treg-expressed CD27 prevents the breakdown of peripheral tolerance and limits anti-tumor immunity. Furthermore, ablation of Treg expressed CD27 acts synergistically with PD-1 checkpoint inhibition to improve CTL mediated immunity against a solid tumor. Our data thus identify Treg-expressed CD27 as a potential target in cancer immunotherapy. KEY MESSAGES : Treg expressed CD27 maintains steady state DC tolerogenic Treg expressed CD27 limits anti-tumor immunity Ablation of Treg expressed CD27 synergizes with PD-1 blockade to improve CTL mediated tumor control.
Collapse
Affiliation(s)
- Sabine Muth
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany.
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany.
| | - Annekatrin Klaric
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Markus Radsak
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
- IIIrd Department of Medicine Hematology, Oncology, Pneumology, University Medical Center Mainz, Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Hans Christian Probst
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany.
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
7
|
The Interplay between Nutrition, Innate Immunity, and the Commensal Microbiota in Adaptive Intestinal Morphogenesis. Nutrients 2021; 13:nu13072198. [PMID: 34206809 PMCID: PMC8308283 DOI: 10.3390/nu13072198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract is a functionally and anatomically segmented organ that is colonized by microbial communities from birth. While the genetics of mouse gut development is increasingly understood, how nutritional factors and the commensal gut microbiota act in concert to shape tissue organization and morphology of this rapidly renewing organ remains enigmatic. Here, we provide an overview of embryonic mouse gut development, with a focus on the intestinal vasculature and the enteric nervous system. We review how nutrition and the gut microbiota affect the adaptation of cellular and morphologic properties of the intestine, and how these processes are interconnected with innate immunity. Furthermore, we discuss how nutritional and microbial factors impact the renewal and differentiation of the epithelial lineage, influence the adaptation of capillary networks organized in villus structures, and shape the enteric nervous system and the intestinal smooth muscle layers. Intriguingly, the anatomy of the gut shows remarkable flexibility to nutritional and microbial challenges in the adult organism.
Collapse
|
8
|
Joeris T, Gomez-Casado C, Holmkvist P, Tavernier SJ, Silva-Sanchez A, Klotz L, Randall TD, Mowat AM, Kotarsky K, Malissen B, Agace WW. Intestinal cDC1 drive cross-tolerance to epithelial-derived antigen via induction of FoxP3 +CD8 + T regs. Sci Immunol 2021; 6:6/60/eabd3774. [PMID: 34088744 DOI: 10.1126/sciimmunol.abd3774] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 01/25/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Although CD8+ T cell tolerance to tissue-specific antigen (TSA) is essential for host homeostasis, the mechanisms underlying peripheral cross-tolerance and whether they may differ between tissue sites remain to be fully elucidated. Here, we demonstrate that peripheral cross-tolerance to intestinal epithelial cell (IEC)-derived antigen involves the generation and suppressive function of FoxP3+CD8+ T cells. FoxP3+CD8+ Treg generation was dependent on intestinal cDC1, whose absence led to a break of tolerance and epithelial destruction. Mechanistically, intestinal cDC1-derived PD-L1, TGFβ, and retinoic acid contributed to the generation of gut-tropic CCR9+CD103+FoxP3+CD8+ Tregs Last, CD103-deficient CD8+ T cells lacked tolerogenic activity in vivo, indicating a role for CD103 in FoxP3+CD8+ Treg function. Our results describe a role for FoxP3+CD8+ Tregs in cross-tolerance in the intestine for which development requires intestinal cDC1.
Collapse
Affiliation(s)
- Thorsten Joeris
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kemitorvet, Kgs. Lyngby 2800, Denmark, Denmark.,Immunology Section, Lund University, Lund 221 84, Sweden
| | | | | | - Simon J Tavernier
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent 9000, Belgium.,VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, 9000 Ghent, Belgium
| | - Aaron Silva-Sanchez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Luisa Klotz
- University Hospital Münster, Department of Neurology with Institute of Translational Neurology, Münster 48149, Germany
| | - Troy D Randall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Allan M Mowat
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| | - Knut Kotarsky
- Immunology Section, Lund University, Lund 221 84, Sweden
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, INSERM, CNRS, Marseille, France
| | - William W Agace
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kemitorvet, Kgs. Lyngby 2800, Denmark, Denmark. .,Immunology Section, Lund University, Lund 221 84, Sweden
| |
Collapse
|
9
|
Modulatory effect of rapamycin and tacrolimus on monocyte-derived dendritic cells phenotype and function. Immunobiology 2020; 226:152031. [PMID: 33278711 DOI: 10.1016/j.imbio.2020.152031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 10/16/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immunosuppressive-drugs are needed after solid organ transplantation to prevent allograft rejection but induce severe side effects. Understanding the alloimmune response is critical to modulate it and to achieve graft operational tolerance. The role of regulatory T cells and tolerogenic dendritic cells (Tol-DCs) is undoubtedly essential in tolerance induction. Tacrolimus is considered as the cornerstone of immunosuppression in solid organ transplantation. mTOR inhibitor such as rapamycin are thought to induce tolerance and are used as anticancer drugs in several cancers. The aim of this study was to better understand the effect of these immunosuppressive drugs on the differentiation, maturation and function of human monocyte derived dendritic cells (DCs). MATERIAL AND METHODS DCs were differentiated from monocytes of healthy donors with either rapamycin (Rapa-DCs) or tacrolimus (Tac-DCs). The phenotype was evaluated by flow cytometry analysis. The production of pro- and anti-inflammatory cytokines was assessed by ELISA. The mRNA expression level of IDO and PD-L1 was assessed by RTqPCR. Mixed leukocytes reactions were performed to analyse suppressive activity of DCs. RESULTS Rapa-DC were characterised by a lower expression of the co-stimulatory molecules and CD83 than control-DCs (CTR-DC) (p < 0.05). In contrast, tacrolimus had no effect on the expression of surface markers compared to CTR-DCs. Rapamycin reduced both IL-12 and IL-10 secretions (p < 0.05). Rapa-DCs had a suppressive effect on CD4+ allogenic T cells compared to CTR-DCs (p < 0.05). However, neither Rapa-DCs nor Tac-DCs favoured the emergence of a CD4+CD25highFoxp3+ population compared to CTR-DCs. Surprisingly, Rapa-DCs had a reduced expression of IDO and PD-L1 compared to Tac-DCs and CTR-DCs. CONCLUSION Rapa-DCs exhibit an incomplete phenotypic tolerogenic profile. To our knowledge this is the first paper showing a reduction of expression of pro-tolerogenic enzyme IDO in DCs. Tacrolimus does not change the phenotypical or functional characteristics of moDCs.
Collapse
|
10
|
Lenders V, Koutsoumpou X, Sargsian A, Manshian BB. Biomedical nanomaterials for immunological applications: ongoing research and clinical trials. NANOSCALE ADVANCES 2020; 2:5046-5089. [PMID: 36132021 PMCID: PMC9418019 DOI: 10.1039/d0na00478b] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/22/2020] [Indexed: 05/04/2023]
Abstract
Research efforts on nanomaterial-based therapies for the treatment of autoimmune diseases and cancer have spiked and have made rapid progress over the past years. Nanomedicine has been shown to contribute significantly to overcome current therapeutic limitations, exhibiting advantages compared to conventional therapeutics, such as sustained drug release, delayed drug degradation and site-specific drug delivery. Multiple nanodrugs have reached the clinic, but translation is often hampered by either low targeting efficiency or undesired side effects. Nanomaterials, and especially inorganic nanoparticles, have gained criticism due to their potential toxic effects, including immunological alterations. However, many strategies have been attempted to improve the therapeutic efficacy of nanoparticles and exploit their unique properties for the treatment of inflammation and associated diseases. In this review, we elaborate on the immunomodulatory effects of nanomaterials, with a strong focus on the underlying mechanisms that lead to these specific immune responses. Nanomaterials to be discussed include inorganic nanoparticles such as gold, silica and silver, as well as organic nanomaterials such as polymer-, dendrimer-, liposomal- and protein-based nanoparticles. Furthermore, various approaches for tuning nanomaterials in order to enhance their efficacy and attenuate their immune stimulation or suppression, with respect to the therapeutic application, are described. Additionally, we illustrate how the acquired insights have been used to design immunotherapeutic strategies for a variety of diseases. The potential of nanomedicine-based therapeutic strategies in immunotherapy is further illustrated by an up to date overview of current clinical trials. Finally, recent efforts into enhancing immunogenic cell death through the use of nanoparticles are discussed.
Collapse
Affiliation(s)
- Vincent Lenders
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Xanthippi Koutsoumpou
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Ara Sargsian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Bella B Manshian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| |
Collapse
|
11
|
Dong M, Wang X, L T, Wang J, Yang Y, Liu Y, Jing Y, Zhao H, Chen J. Mir-27a-3p attenuates bronchiolitis obliterans in vivo via the regulation of dendritic cells' maturation and the suppression of myofibroblasts' differentiation. Clin Transl Med 2020; 10:e140. [PMID: 32898329 PMCID: PMC7423186 DOI: 10.1002/ctm2.140] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023] Open
Abstract
Bronchiolitis obliterans (BO), is a chronic rejection phenotype characterized by chronic small airway fibrous obliteration, hinders the patients who suffer from lung transplanting for surviving longer. Cell-based therapies using dendritic cells (DCs) and T regulatory cells (Tregs) have been developed to regulate allograft rejection, and to induce and maintain immune tolerance. In the present study, the effects of mir-27a-3p on regulating DCs as well as resulting effects on BO attenuation have been investigated. According to our reporter assays, the potential targets of mir-27a-3p were Smad2, sprouty2, and Smad4, respectively. Furthermore, sprouty2 inhibition by mir-27-3p indirectly activated extracellular regulated protein kinases (ERK) and increased IL-10 production in DCs. This led to a positive feedback loop that maintained the immature state of DCs via IL-10/JAK/STAT3 pathway, and caused an increase in Foxp3+ CD4+ T cells amount as well as TGF-β level. Furthermore, mir-27a-3p regulated TGF-β function, inhibited TGF-β/Smad pathway, and suppressed myofibroblast differentiation through influencing the function of Smad2 and Smad4. In short, the study indicated the effect of mir-27a-3p on suppressing DC maturation, which implicated the potential clinical application in treating postlung transplant BO.
Collapse
Affiliation(s)
- Ming Dong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Xin Wang
- Department of Pediatric Surgery, Tianjin Children's Hospital, Tianjin, P. R. China
| | - Tong L
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Jing Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Yunwei Yang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yi Liu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yaqing Jing
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Honglin Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| |
Collapse
|
12
|
Feng X, Liu J, Xu W, Li G, Ding J. Tackling autoimmunity with nanomedicines. Nanomedicine (Lond) 2020; 15:1585-1597. [DOI: 10.2217/nnm-2020-0102] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tolerogenic immunotherapy aims to blunt pathogenic inflammation without affecting systemic immunity. However, the anti-inflammatory drugs and immunosuppressive biologics that are used in the clinic usually result in nonspecific immune cell suppression and off-target toxicity. For this reason, strategies have been developed to induce antigen-specific immune tolerance through the delivery of disease-relevant antigens by nanocarriers as a benefit of their preferential internalization by antigen-presenting cells. Herein, we discuss the recent advances in the nanotechnology-based antigen-specific tolerance approaches. Some of these designs are based on nanoparticles delivering antigens and immunoregulatory agents to modulate antigen-presenting pathways, while others directly target T cells via nanoparticle-based artificial antigen-presenting cells. These antigen-specific therapies are hoped to replace systemic immune suppression and provide long-term disease remission.
Collapse
Affiliation(s)
- Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
- University of Science & Technology of China, 96 Jinzhai Road, Hefei, 230026, PR China
| | - Jiaxue Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
- University of Science & Technology of China, 96 Jinzhai Road, Hefei, 230026, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
13
|
Sushnitha M, Evangelopoulos M, Tasciotti E, Taraballi F. Cell Membrane-Based Biomimetic Nanoparticles and the Immune System: Immunomodulatory Interactions to Therapeutic Applications. Front Bioeng Biotechnol 2020; 8:627. [PMID: 32626700 PMCID: PMC7311577 DOI: 10.3389/fbioe.2020.00627] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Nanoparticle-based drug delivery systems have been synthesized from a wide array of materials. The therapeutic success of these platforms hinges upon their ability to favorably interact with the biological environment (both systemically and locally) and recognize the diseased target tissue. The immune system, composed of a highly coordinated organization of cells trained to recognize foreign bodies, represents a key mediator of these interactions. Although components of this system may act as a barrier to nanoparticle (NP) delivery, the immune system can also be exploited to target and trigger signaling cues that facilitate the therapeutic response stemming from systemic administration of NPs. The nano-bio interface represents the key facilitator of this communication exchange, where the surface properties of NPs govern their in vivo fate. Cell membrane-based biomimetic nanoparticles have emerged as one approach to achieve targeted drug delivery by actively engaging and communicating with the biological milieu. In this review, we will highlight the relationship between these biomimetic nanoparticles and the immune system, emphasizing the role of tuning the nano-bio interface in the immunomodulation of diseases. We will also discuss the therapeutic applications of this approach with biomimetic nanoparticles, focusing on specific diseases ranging from cancer to infectious diseases. Lastly, we will provide a critical evaluation on the current state of this field of cell membrane-based biomimetic nanoparticles and its future directions in immune-based therapy.
Collapse
Affiliation(s)
- Manuela Sushnitha
- Department of Bioengineering, Rice University, Houston, TX, United States
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
14
|
Cauwels A, Tavernier J. Tolerizing Strategies for the Treatment of Autoimmune Diseases: From ex vivo to in vivo Strategies. Front Immunol 2020; 11:674. [PMID: 32477325 PMCID: PMC7241419 DOI: 10.3389/fimmu.2020.00674] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Autoimmune diseases such as multiple sclerosis (MS), type I diabetes (T1D), inflammatory bowel diseases (IBD), and rheumatoid arthritis (RA) are chronic, incurable, incapacitating and at times even lethal conditions. Worldwide, millions of people are affected, predominantly women, and their number is steadily increasing. Currently, autoimmune patients require lifelong immunosuppressive therapy, often accompanied by severe adverse side effects and risks. Targeting the fundamental cause of autoimmunity, which is the loss of tolerance to self- or innocuous antigens, may be achieved via various mechanisms. Recently, tolerance-inducing cellular therapies, such as tolerogenic dendritic cells (tolDCs) and regulatory T cells (Tregs), have gained considerable interest. Their safety has already been evaluated in patients with MS, arthritis, T1D, and Crohn’s disease, and clinical trials are underway to confirm their safety and therapeutic potential. Cell-based therapies are inevitably expensive and time-consuming, requiring laborious ex vivo manufacturing. Therefore, direct in vivo targeting of tolerogenic cell types offers an attractive alternative, and several strategies are being explored. Type I IFN was the first disease-modifying therapy approved for MS patients, and approaches to endogenously induce IFN in autoimmune diseases are being pursued vigorously. We here review and discuss tolerogenic cellular therapies and targeted in vivo tolerance approaches and propose a novel strategy for cell-specific delivery of type I IFN signaling to a cell type of choice.
Collapse
Affiliation(s)
- Anje Cauwels
- VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium.,Orionis Biosciences, Ghent, Belgium
| |
Collapse
|
15
|
Schaupp L, Muth S, Rogell L, Kofoed-Branzk M, Melchior F, Lienenklaus S, Ganal-Vonarburg SC, Klein M, Guendel F, Hain T, Schütze K, Grundmann U, Schmitt V, Dorsch M, Spanier J, Larsen PK, Schwanz T, Jäckel S, Reinhardt C, Bopp T, Danckwardt S, Mahnke K, Heinz GA, Mashreghi MF, Durek P, Kalinke U, Kretz O, Huber TB, Weiss S, Wilhelm C, Macpherson AJ, Schild H, Diefenbach A, Probst HC. Microbiota-Induced Type I Interferons Instruct a Poised Basal State of Dendritic Cells. Cell 2020; 181:1080-1096.e19. [PMID: 32380006 DOI: 10.1016/j.cell.2020.04.022] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/31/2019] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Environmental signals shape host physiology and fitness. Microbiota-derived cues are required to program conventional dendritic cells (cDCs) during the steady state so that they can promptly respond and initiate adaptive immune responses when encountering pathogens. However, the molecular underpinnings of microbiota-guided instructive programs are not well understood. Here, we report that the indigenous microbiota controls constitutive production of type I interferons (IFN-I) by plasmacytoid DCs. Using genome-wide analysis of transcriptional and epigenetic regulomes of cDCs from germ-free and IFN-I receptor (IFNAR)-deficient mice, we found that tonic IFNAR signaling instructs a specific epigenomic and metabolic basal state that poises cDCs for future pathogen combat. However, such beneficial biological function comes with a trade-off. Instructed cDCs can prime T cell responses against harmless peripheral antigens when removing roadblocks of peripheral tolerance. Our data provide fresh insights into the evolutionary trade-offs that come with successful adaptation of vertebrates to their microbial environment.
Collapse
Affiliation(s)
- Laura Schaupp
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Institute for Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Sabine Muth
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Leif Rogell
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Michael Kofoed-Branzk
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Felix Melchior
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stefan Lienenklaus
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany; Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Stephanie C Ganal-Vonarburg
- Department for BioMedical Research (DBMR), University Clinic for Visceral Surgery and Medicine, Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Matthias Klein
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Fabian Guendel
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Tobias Hain
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kristian Schütze
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Ulrike Grundmann
- Institute for Medical Microbiology and Hygiene, University of Freiburg Medical Center, Hermann-Herder-Str. 11, 79104 Freiburg, Germany
| | - Vanessa Schmitt
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Martina Dorsch
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Thomas Schwanz
- Institute of Medical Microbiology and Hygiene, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany
| | - Sven Jäckel
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Tobias Bopp
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; University Cancer Center Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; German Cancer Consortium (DKTK)
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Posttranscriptional Gene Regulation, Cancer Research and Experimental Hemostasis, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Karsten Mahnke
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, D-69120 Heidelberg, Germany
| | - Gitta Anne Heinz
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Pawel Durek
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany; Department for Neuroanatomy, Anatomy and Cell Biology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Siegfried Weiss
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Andrew J Macpherson
- Department for BioMedical Research (DBMR), University Clinic for Visceral Surgery and Medicine, Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; Helmholtz Institute Translational Oncology, Obere Zahlbacher Straße 63, 55131 Mainz, Germany.
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany.
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW To evaluate role of the lymph node in immune regulation and tolerance in transplantation and recent advances in the delivery of antigen and immune modulatory signals to the lymph node. RECENT FINDINGS Lymph nodes are a primary site of immune cell priming, activation, and modulation, and changes within the lymph node microenvironment have the potential to induce specific regulation, suppression, and potentially tolerance. Antigen enters the lymph node either from tissues via lymphatics, from blood via high endothelial venules, or directly via injection. Here we review different techniques and materials to deliver antigen to the lymph node including microparticles or nanoparticles, ex-vivo antigen presenting cell manipulation, and use of receptor conjugation for specific intralymph node targeting locations. SUMMARY The promising results point to powerful techniques to harness the lymph node microenvironment and direct systemic immune regulation. The materials, techniques, and approaches suggest that translational and clinical trials in nonhuman primate and patients may soon be possible.
Collapse
|
17
|
Krzastek SC, Goliadze E, Zhou S, Petrossian A, Youniss F, Sundaresan G, Wang L, Zweit J, Guruli G. Dendritic cell trafficking in tumor-bearing mice. Cancer Immunol Immunother 2018; 67:1939-1947. [PMID: 29943070 PMCID: PMC11028156 DOI: 10.1007/s00262-018-2187-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/13/2018] [Indexed: 12/16/2022]
Abstract
Prostate cancer is one of the leading causes of cancer deaths, with no curative treatments once it spreads. Alternative therapies, including immunotherapy, have shown limited efficacy. Dendritic cells (DC) have been widely used in the treatment of various malignancies. DC capture antigens and move to the lymphoid organs where they prime naive T cells. Interaction between DC and T cells are most active in lymph nodes and suppression of DC trafficking to lymph nodes impairs the immune response. In this work, we aimed to study trafficking of DC in vivo via various routes of delivery, to optimize the effectiveness of DC-based therapy. A DC labeling system was developed using 1,1'-dioctadecyltetramethyl indotricarbocyanine Iodine for in vivo fluorescent imaging. DC harvested from C57B/6 mice were matured, labeled, and injected intravenously, subcutaneously, or intratumorally, with or without antigen loading with whole tumor lysate, into C57B/6 mice inoculated with RM-1 murine prostate tumor cells. Signal intensity was measured in vivo and ex vivo. Signal intensity at the tumor site increased over time, suggesting trafficking of DC to the tumor with all modes of injection. Subcutaneous injection showed preferential trafficking to lymph nodes and tumor. Intravenous injection showed trafficking to lungs, intestines, and spleen. Subcutaneous injection of DC pulsed with whole tumor lysate resulted in the highest increase in signal intensity at the tumor site and lymph nodes, suggesting subcutaneous injection of primed DC leads to highest preferential trafficking of DC to the immunocompetent organs.
Collapse
Affiliation(s)
- Sarah C Krzastek
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Ekaterine Goliadze
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Shaoqing Zhou
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Albert Petrossian
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Fatma Youniss
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Gobalakrishnan Sundaresan
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Li Wang
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jamal Zweit
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgi Guruli
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
18
|
Vitetta L, Vitetta G, Hall S. Immunological Tolerance and Function: Associations Between Intestinal Bacteria, Probiotics, Prebiotics, and Phages. Front Immunol 2018; 9:2240. [PMID: 30356736 PMCID: PMC6189397 DOI: 10.3389/fimmu.2018.02240] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Post-birth there is a bacterial assault on all mucosal surfaces. The intestinal microbiome is an important participant in health and disease. The pattern of composition and concentration of the intestinal microbiome varies greatly. Therefore, achieving immunological tolerance in the first 3-4 years of life is critical for maintaining health throughout a lifetime. Probiotic bacteria are organisms that afford beneficial health effects to the host and in certain instances may protect against the development of disease. The potential benefits of modifying the composition of the intestinal microbial cohort for therapeutic benefit is evident in the use in high risks groups such as premature infants, children receiving antibiotics, rotavirus infections in non-vaccinated children and traveler's diarrhea in adults. Probiotics and prebiotics are postulated to have immunomodulating capabilities by influencing the intestinal microbial cohort and dampening the activity of pathobiont intestinal microbes, such as Klebsiella pneumonia and Clostridia perfringens. Lactobacilli and Bifidobacteria are examples of probiotics found in the large intestine and so far, the benefits afforded to probiotics have varied in efficacy. Most likely the efficacy of probiotic bacteria has a multifactorial dependency, namely on a number of factors that include agents used, the dose, the pattern of dosing, and the characteristics of the host and the underlying luminal microbial environment and the activity of bacteriophages. Bacteriophages, are small viruses that infect and lyse intestinal bacteria. As such it can be posited that these viruses display an effective local protective control mechanism for the intestinal barrier against commensal pathobionts that indirectly may assist the host in controlling bacterial concentrations in the gut. A co-operative activity may be envisaged between the intestinal epithelia, mucosal immunity and the activity of bacteriophages to eliminate pathobiots, highlighting the potential role of bacteriophages in assisting with maintaining intestinal homeostasis. Hence bacteriophage local control of inflammation and immune responses may be an additional immunological defense mechanism that exploits bacteriophage-mucin glycoprotein interactions that controls bacterial diversity and abundance in the mucin layers of the gut. Moreover, and importantly the efficacy of probiotics may be dependent on the symbiotic incorporation of prebiotics, and the abundance and diversity of the intestinal microbiome encountered. The virome may be an important factor that determines the efficacy of some probiotic formulations.
Collapse
Affiliation(s)
- Luis Vitetta
- Discipline of Pharmacology, Faculty of Medicine and Health, School of Medicine, The University of Sydney, Camperdown, NSW, Australia
- Medlab Clinical Ltd., Sydney, NSW, Australia
| | | | - Sean Hall
- Medlab Clinical Ltd., Sydney, NSW, Australia
| |
Collapse
|
19
|
Kiouptsi K, Reinhardt C. Contribution of the commensal microbiota to atherosclerosis and arterial thrombosis. Br J Pharmacol 2018; 175:4439-4449. [PMID: 30129122 DOI: 10.1111/bph.14483] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/05/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
The commensal gut microbiota is an environmental factor that has been implicated in the development of cardiovascular disease. The development of atherosclerotic lesions is largely influenced not only by the microbial-associated molecular patterns of the gut microbiota but also by the meta-organismal trimethylamine N-oxide pathway. Recent studies have described a role for the gut microbiota in platelet activation and arterial thrombosis. This review summarizes the results from gnotobiotic mouse models and clinical data that linked microbiota-induced pattern recognition receptor signalling with atherogenesis. Based on recent insights, we here provide an overview of how the gut microbiota could affect endothelial cell function and platelet activation, to promote arterial thrombosis. LINKED ARTICLES: This article is part of a themed section on When Pharmacology Meets the Microbiome: New Targets for Therapeutics? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.24/issuetoc.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany.,German Center for Cardiovascular Research, Partner Site RheinMain, Mainz, Germany
| |
Collapse
|
20
|
Bigley V, Cytlak U, Collin M. Human dendritic cell immunodeficiencies. Semin Cell Dev Biol 2018; 86:50-61. [PMID: 29452225 DOI: 10.1016/j.semcdb.2018.02.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 11/28/2017] [Accepted: 02/10/2018] [Indexed: 12/21/2022]
Abstract
The critical functions of dendritic cells (DCs) in immunity and tolerance have been demonstrated in many animal models but their non-redundant roles in humans are more difficult to probe. Human primary immunodeficiency (PID), resulting from single gene mutations, may result in DC deficiency or dysfunction. This relatively recent recognition illuminates the in vivo role of human DCs and the pathophysiology of the associated clinical syndromes. In this review, the development and function of DCs as established in murine models and human in vitro systems, discussed. This forms the basis of predicting the effects of DC deficiency in vivo and understanding the consequences of specific mutations on DC development and function. DC deficiency syndromes are associated with heterozygous GATA2 mutation, bi-allelic and heterozygous IRF8 mutation and heterozygous IKZF1 mutation. The intricate involvement of DCs in the balance between immunity and tolerance is leading to increased recognition of their involvement in a number of other immunodeficiencies and autoimmune conditions. Owing to the precise control of transcription factor gene expression by super-enhancer elements, phenotypic anomalies are relatively commonly caused by heterozygous mutations.
Collapse
Affiliation(s)
- Venetia Bigley
- Human DC Lab, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Urszula Cytlak
- Human DC Lab, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew Collin
- Human DC Lab, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
21
|
Manches O, Muniz LR, Bhardwaj N. Dendritic Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
22
|
Audiger C, Rahman MJ, Yun TJ, Tarbell KV, Lesage S. The Importance of Dendritic Cells in Maintaining Immune Tolerance. THE JOURNAL OF IMMUNOLOGY 2017; 198:2223-2231. [PMID: 28264998 DOI: 10.4049/jimmunol.1601629] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/11/2016] [Indexed: 12/30/2022]
Abstract
Immune tolerance is necessary to prevent the immune system from reacting against self, and thus to avoid the development of autoimmune diseases. In this review, we discuss key findings that position dendritic cells (DCs) as critical modulators of both thymic and peripheral immune tolerance. Although DCs are important for inducing both immunity and tolerance, increased autoimmunity associated with decreased DCs suggests their nonredundant role in tolerance induction. DC-mediated T cell immune tolerance is an active process that is influenced by genetic variants, environmental signals, as well as the nature of the specific DC subset presenting Ag to T cells. Answering the many open questions with regard to the role of DCs in immune tolerance could lead to the development of novel therapies for the prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Cindy Audiger
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - M Jubayer Rahman
- Immune Tolerance Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Tae Jin Yun
- Laboratory of Cellular Physiology and Immunology, Clinical Research Institute of Montreal, Montreal, Quebec H2W 1R7, Canada; and.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Kristin V Tarbell
- Immune Tolerance Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sylvie Lesage
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
23
|
Kepert I, Fonseca J, Müller C, Milger K, Hochwind K, Kostric M, Fedoseeva M, Ohnmacht C, Dehmel S, Nathan P, Bartel S, Eickelberg O, Schloter M, Hartmann A, Schmitt-Kopplin P, Krauss-Etschmann S. D-tryptophan from probiotic bacteria influences the gut microbiome and allergic airway disease. J Allergy Clin Immunol 2016; 139:1525-1535. [PMID: 27670239 DOI: 10.1016/j.jaci.2016.09.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 05/13/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic immune diseases, such as asthma, are highly prevalent. Currently available pharmaceuticals improve symptoms but cannot cure the disease. This prompted demands for alternatives to pharmaceuticals, such as probiotics, for the prevention of allergic disease. However, clinical trials have produced inconsistent results. This is at least partly explained by the highly complex crosstalk among probiotic bacteria, the host's microbiota, and immune cells. The identification of a bioactive substance from probiotic bacteria could circumvent this difficulty. OBJECTIVE We sought to identify and characterize a bioactive probiotic metabolite for potential prevention of allergic airway disease. METHODS Probiotic supernatants were screened for their ability to concordantly decrease the constitutive CCL17 secretion of a human Hodgkin lymphoma cell line and prevent upregulation of costimulatory molecules of LPS-stimulated human dendritic cells. RESULTS Supernatants from 13 of 37 tested probiotic strains showed immunoactivity. Bioassay-guided chromatographic fractionation of 2 supernatants according to polarity, followed by total ion chromatography and mass spectrometry, yielded C11H12N2O2 as the molecular formula of a bioactive substance. Proton nuclear magnetic resonance and enantiomeric separation identified D-tryptophan. In contrast, L-tryptophan and 11 other D-amino acids were inactive. Feeding D-tryptophan to mice before experimental asthma induction increased numbers of lung and gut regulatory T cells, decreased lung TH2 responses, and ameliorated allergic airway inflammation and hyperresponsiveness. Allergic airway inflammation reduced gut microbial diversity, which was increased by D-tryptophan. CONCLUSIONS D-tryptophan is a newly identified product from probiotic bacteria. Our findings support the concept that defined bacterial products can be exploited in novel preventative strategies for chronic immune diseases.
Collapse
Affiliation(s)
- Inge Kepert
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany
| | - Juliano Fonseca
- Research Unit Analytical BioGeoChemistry (BGC), Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Constanze Müller
- Research Unit Analytical BioGeoChemistry (BGC), Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Katrin Milger
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany
| | - Kerstin Hochwind
- Research Unit Microbe-Plant Interactions, Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Matea Kostric
- Research Unit Environmental Genomics, Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Maria Fedoseeva
- Center of Allergy and Environment (ZAUM), Technische Universität and Helmholtz Zentrum München, Member of the German Center for Lung research (DZL), Oberschleissheim, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technische Universität and Helmholtz Zentrum München, Member of the German Center for Lung research (DZL), Oberschleissheim, Germany
| | - Stefan Dehmel
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany
| | - Petra Nathan
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany
| | - Sabine Bartel
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany; Division of Experimental Asthma Research, Research Center Borstel, Leibniz Center for Medicine and Biosciences, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany
| | - Michael Schloter
- Research Unit Environmental Genomics, Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Anton Hartmann
- Research Unit Microbe-Plant Interactions, Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry (BGC), Helmholtz Zentrum München, Oberschleissheim, Germany; Analytical Food Chemistry, Technische Universität Muenchen, Freising, Germany
| | - Susanne Krauss-Etschmann
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital, Member of the German Center for Lung Research (DZL), and Helmholtz Zentrum München, Munich, Germany; Division of Experimental Asthma Research, Research Center Borstel, Leibniz Center for Medicine and Biosciences, Member of the German Center for Lung Research (DZL), Borstel, Germany; Institute for Experimental Medicine, Christian-Albrechts-Universitaet zu Kiel, Kiel, Germany.
| |
Collapse
|
24
|
Biologics and the lung: TSLP and other epithelial cell-derived cytokines in asthma. Pharmacol Ther 2016; 169:104-112. [PMID: 27365223 DOI: 10.1016/j.pharmthera.2016.06.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic airway inflammatory disorder with characteristic symptoms of dyspnea, wheeze, chest tightness and cough, and physiological abnormalities of variable airway obstruction, airway hyperresponsiveness, and in some patients with chronic long standing disease reduced lung function. The physiological abnormalities are due to chronic airway inflammation and underlying structural changes to the airway wall. The interaction between the airway epithelium and the environment is crucial to the pathobiology of asthma. Several recent discoveries have highlighted a crucial role of airway epithelial derived cytokines such as interleukin (IL)-25, IL-33 and thymic stromal lymphopoietin (TSLP). These cytokines are collectively known as epithelial "alarmins", which act solely or in concert to activate and potentiate the innate and humoral arms of the immune system in the presence of actual or perceive damage. Understanding the role of alarmins and how they are activated and released may allow the development of novel new therapeutics to treat asthma. This review describes the interactions between inhaled air, the pulmonary microbiome, airway epithelial cell layer and the alarmins, IL-25, IL-33 and TSLP. There is already compelling evidence for a role of TSLP in the airway responses to environmental allergens in allergic asthmatics, as well as in maintaining airway eosinophilic inflammation in these subjects. Further work is required to develop human monoclonal antibodies (hMabs) directed against IL-25 and IL-33 or their receptors, to help understand their role in the initiation and/or persistence of asthma.
Collapse
|
25
|
Papazian D, Hansen S, Würtzen PA. Airway responses towards allergens - from the airway epithelium to T cells. Clin Exp Allergy 2016; 45:1268-87. [PMID: 25394747 DOI: 10.1111/cea.12451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The prevalence of allergic diseases such as allergic rhinitis is increasing, affecting up to 30% of the human population worldwide. Allergic sensitization arises from complex interactions between environmental exposures and genetic susceptibility, resulting in inflammatory T helper 2 (Th2) cell-derived immune responses towards environmental allergens. Emerging evidence now suggests that an epithelial dysfunction, coupled with inherent properties of environmental allergens, can be responsible for the inflammatory responses towards allergens. Several epithelial-derived cytokines, such as thymic stromal lymphopoietin (TSLP), IL-25 and IL-33, influence tissue-resident dendritic cells (DCs) as well as Th2 effector cells. Exposure to environmental allergens does not elicit Th2 inflammatory responses or any clinical symptoms in nonatopic individuals, and recent findings suggest that a nondamaged, healthy epithelium lowers the DCs' ability to induce inflammatory T-cell responses towards allergens. The purpose of this review was to summarize the current knowledge on which signals from the airway epithelium, from first contact with inhaled allergens all the way to the ensuing Th2-cell responses, influence the pathology of allergic diseases.
Collapse
Affiliation(s)
- D Papazian
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,ALK, Hørsholm, Denmark
| | - S Hansen
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
26
|
Edelblum KL. Dissecting the Requirement for Secondary Lymphoid Organs in Peripheral Regulatory T-Cell Development. Cell Mol Gastroenterol Hepatol 2016; 2:253-254. [PMID: 28174714 PMCID: PMC5042369 DOI: 10.1016/j.jcmgh.2016.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- Karen L. Edelblum
- Correspondence Address correspondence to: Karen L. Edelblum, PhD, Department of Pathology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB E673, Newark, New Jersey 07103.Department of Pathology and Laboratory MedicineCenter for Immunity and InflammationRutgers New Jersey Medical School185 South Orange AvenueMSB E673NewarkNew Jersey 07103
| |
Collapse
|
27
|
Targeting dendritic cells: a promising strategy to improve vaccine effectiveness. Clin Transl Immunology 2016; 5:e66. [PMID: 27217957 PMCID: PMC4815026 DOI: 10.1038/cti.2016.6] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) targeting is a novel strategy to enhance vaccination efficacy. This approach is based on the in situ delivery of antigen via antibodies that are specific for endocytic receptors expressed at the surface of DCs. Here we review the complexity of the DC subsets and the antigen presentation pathways that need to be considered in the settings of DC targeting. We also summarize current knowledge about antigen delivery to DCs via DEC-205, Clec9A and Clec12A, receptor targets that strongly enhance cellular and humoral immune responses. Finally, we discuss the intracellular trafficking criteria of the targeted receptors that may impact their effectiveness as DC targets.
Collapse
|
28
|
Secretory IgA in complex with Lactobacillus rhamnosus potentiates mucosal dendritic cell-mediated Treg cell differentiation via TLR regulatory proteins, RALDH2 and secretion of IL-10 and TGF-β. Cell Mol Immunol 2016; 14:546-556. [PMID: 26972771 DOI: 10.1038/cmi.2015.110] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/02/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022] Open
Abstract
The importance of secretory IgA in controlling the microbiota is well known, yet how the antibody affects the perception of the commensals by the local immune system is still poorly defined. We have previously shown that the transport of secretory IgA in complex with bacteria across intestinal microfold cells results in an association with dendritic cells in Peyer's patches. However, the consequences of such an interaction on dendritic cell conditioning have not been elucidated. In this study, we analyzed the impact of the commensal Lactobacillus rhamnosus, alone or associated with secretory IgA, on the responsiveness of dendritic cells freshly recovered from mouse Peyer's patches, mesenteric lymph nodes, and spleen. Lactobacillus rhamnosus-conditioned mucosal dendritic cells are characterized by increased expression of Toll-like receptor regulatory proteins [including single immunoglobulin interleukin-1 receptor-related molecule, suppressor of cytokine signaling 1, and Toll-interacting molecule] and retinaldehyde dehydrogenase 2, low surface expression of co-stimulatory markers, high anti- versus pro-inflammatory cytokine production ratios, and induction of T regulatory cells with suppressive function. Association with secretory IgA enhanced the anti-inflammatory/regulatory Lactobacillus rhamnosus-induced conditioning of mucosal dendritic cells, particularly in Peyer's patches. At the systemic level, activation of splenic dendritic cells exposed to Lactobacillus rhamnosus was partially dampened upon association with secretory IgA. These data suggest that secretory IgA, through coating of commensal bacteria, contributes to the conditioning of mucosal dendritic cells toward tolerogenic profiles essential for the maintenance of intestinal homeostasis.
Collapse
|
29
|
Käfer R, Usanova S, Montermann E, Loquai C, Reske-Kunz AB, Bros M. Inhibitors of β-catenin affect the immuno-phenotype and functions of dendritic cells in an inhibitor-specific manner. Int Immunopharmacol 2016; 32:118-124. [DOI: 10.1016/j.intimp.2016.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/08/2016] [Accepted: 01/14/2016] [Indexed: 12/27/2022]
|
30
|
Thomson AW, Zahorchak AF, Ezzelarab MB, Butterfield LH, Lakkis FG, Metes DM. Prospective Clinical Testing of Regulatory Dendritic Cells in Organ Transplantation. Front Immunol 2016; 7:15. [PMID: 26858719 PMCID: PMC4729892 DOI: 10.3389/fimmu.2016.00015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/12/2016] [Indexed: 01/03/2023] Open
Abstract
Dendritic cells (DC) are rare, professional antigen-presenting cells with ability to induce or regulate alloimmune responses. Regulatory DC (DCreg) with potential to down-modulate acute and chronic inflammatory conditions that occur in organ transplantation can be generated in vitro under a variety of conditions. Here, we provide a rationale for evaluation of DCreg therapy in clinical organ transplantation with the goal of promoting sustained, donor-specific hyporesponsiveness, while lowering the incidence and severity of rejection and reducing patients’ dependence on anti-rejection drugs. Generation of donor- or recipient-derived DCreg that suppress T cell responses and prolong transplant survival in rodents or non-human primates has been well-described. Recently, good manufacturing practice (GMP)-grade DCreg have been produced at our Institution for prospective use in human organ transplantation. We briefly review experience of regulatory immune therapy in organ transplantation and describe our experience generating and characterizing human monocyte-derived DCreg. We propose a phase I/II safety study in which the influence of donor-derived DCreg combined with conventional immunosuppression on subclinical and clinical rejection and host alloimmune responses will be examined in detail.
Collapse
Affiliation(s)
- Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alan F Zahorchak
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Mohamed B Ezzelarab
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Lisa H Butterfield
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fadi G Lakkis
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diana M Metes
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Sun Y, Jin X, Liu X, Zhang M, Liu W, Li Z, Han N, Tan M, Chi D, Zhu J, Zhang B, Yu B, Wu J. MicroRNA let-7i regulates dendritic cells maturation targeting interleukin-10 via the Janus kinase 1-signal transducer and activator of transcription 3 signal pathway subsequently induces prolonged cardiac allograft survival in rats. J Heart Lung Transplant 2015; 35:378-388. [PMID: 26755202 DOI: 10.1016/j.healun.2015.10.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/25/2015] [Accepted: 10/31/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In this study, we investig1ated whether microRNA let-7i regulates dendric cell maturation targeting interleukin-10 (IL-10) via the Janus kinase 1-signal transducer and activator of transcription 3 (JAK1-STAT3) signal pathway subsequently prolongs rat cardiac allograft survival. METHODS Quantitative real-time reverse transcriptase polymerase chain reaction, enzyme linked immunosorbent assay, and dual-luciferase assay were performed to verify whether IL-10 was the target of let-7i, and regulatory T cells were assessed by flow cytometry and immunohistochemical study. Western blot was performed to detect JAK1, STAT3, and phosphorylated STAT3 expression. Lewis recipients of Dark Agouti hearts were transfused with phosphate-buffered saline, lipopolysaccharide (LPS)-mature dendritic cells (mDCs), or let-7i-inhibitor-mDCs. Allograft survival times were recorded, and histologic studies were performed. RESULTS Expression of IL-10 messenger RNA level and production of IL-10 were increased in let-7i-inhibitor-mDCs compared with LPS-mDCs. Luciferase activity showed that the translational level of the IL-10 luciferase reporter was decreased by let-7i mimic but increased by let-7i-inhibitor. MicroRNA let-7i inhibitor suppressed DC maturation; however, pretreatment of IL-10 small interfering RNA attenuated the suppression. Expression of JAK1, STAT3, and phosphorylated STAT3 in mDCs were suppressed by let-7i mimic, and pre-treatment of IL-10 small interfering RNA, however, were upregulated by let-7i inhibitor. Lewis recipients transfused with let-7i-inhibitor-mDCs significantly prolonged Dark Agouti cardiac allograft survival. The allografts transfused with let-7i-inhibitor-mDCs showed slight cell infiltration and significantly preserved graft structure. Inhibition of let-7i increased CD4(+)CD25(+)forkhead box P3(+) regulatory T cells and modulated cytokine profiles in vivo and in vitro. CONCLUSIONS MicroRNA let-7i regulated DC maturation and function targeting IL-10 through the JAK1-STAT3 pathway. Moreover, transfusion of LPS-induced mDCs transfected with let-7i inhibitor induced prolonged cardiac allograft survival and generated regulatory T cells.
Collapse
Affiliation(s)
- Yong Sun
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyuan Jin
- Department of Thoracic Surgery, the Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianglan Liu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Maomao Zhang
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Liu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenchao Li
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Na Han
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miaoxin Tan
- Department of Cardiology, the First Hospital of Fangshan District, Beijing, China
| | - Di Chi
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingyi Zhu
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Zhang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Yu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Wu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
32
|
MyD88 dependence of beryllium-induced dendritic cell trafficking and CD4⁺ T-cell priming. Mucosal Immunol 2015; 8:1237-47. [PMID: 25760420 PMCID: PMC4567547 DOI: 10.1038/mi.2015.14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/12/2015] [Indexed: 02/04/2023]
Abstract
Beryllium exposure results in beryllium hypersensitivity in a subset of exposed individuals, leading to granulomatous inflammation and fibrosis in the lung. In addition to its antigenic properties, beryllium has potent adjuvant activity that contributes to sensitization via unknown pathways. Here we show that beryllium induces cellular death and release of interleukin (IL)-1α and DNA into the lung. Release of IL-1α was inflammasome independent and required for beryllium-induced neutrophil recruitment into the lung. Beryllium enhanced classical dendritic cell (cDC) migration from the lung to draining lymph nodes (LNs) in an IL-1R-independent manner, and the accumulation of activated cDCs in the LN was associated with increased priming of CD4(+) T cells. DC migration was reduced in Toll-like receptor 9 knockout (TLR9KO) mice; however, cDCs in the LNs of TLR9-deficient mice were highly activated, suggesting a role for more than one innate receptor in the effects on DCs. The adjuvant effects of beryllium on CD4(+) T-cell priming were similar in wild-type, IL-1R-, caspase-1-, TLR2-, TLR4-, TLR7-, and TLR9-deficient mice. In contrast, DC migration, activation, and the adjuvant effects of beryllium were significantly reduced in myeloid differentiation primary response gene 88 knockout (MyD88KO) mice. Collectively, these data suggest that beryllium exposure results in the release of damage-associated molecular patterns that engage MyD88-dependent receptors to enhance pulmonary DC function.
Collapse
|
33
|
Requirement of full TCR repertoire for regulatory T cells to maintain intestinal homeostasis. Proc Natl Acad Sci U S A 2015; 112:12770-5. [PMID: 26420876 DOI: 10.1073/pnas.1516617112] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The regulation of intestinal homeostasis by the immune system involves the dynamic interplay between gut commensal microbiota and resident immune cells. It is well known that a large and diverse lymphocyte antigen receptor repertoire enables the immune system to recognize and respond to a wide range of invading pathogens. There is also an emerging appreciation for a critical role the T-cell receptor (TCR) repertoire serves in the maintenance of peripheral tolerance by regulatory T cells (Tregs). Nevertheless, how the diversity of the TCR repertoire in Tregs affects intestinal homeostasis remains unknown. To address this question, we studied mice whose T cells express a restricted TCR repertoire. We observed the development of spontaneous colitis, accompanied by the induction of T-helper type 17 cells in the colon that is driven by gut commensal microbiota. We provide further evidence that a restricted TCR repertoire causes a loss of tolerogenicity to microbiota, accompanied by a paucity of peripherally derived, Helios(-) Tregs and hyperactivation of migratory dendritic cells. These results thus reveal a new facet of the TCR repertoire in which Tregs require a diverse TCR repitoire for intestinal homeostasis, suggesting an additional driving force in the evolutional significance of the TCR repertoire.
Collapse
|
34
|
The TGF-β superfamily in dendritic cell biology. Cytokine Growth Factor Rev 2015; 26:647-57. [PMID: 26115564 DOI: 10.1016/j.cytogfr.2015.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022]
Abstract
The TGF-β superfamily consists of a large group of pleiotropic cytokines that are involved in the regulation of many developmental, physiological and pathological processes. Dendritic cells are antigen-presenting cells that play a key role in innate and adaptive immune responses. Dendritic cells have a complex relationship with the TGF-β cytokine superfamily being both source and targets for many of these cytokines. Some TGF-β family members are expressed by dendritic cells and modulate immune responses, for instance through the induction of T cell polarization. Others play a crucial role in the development and function of the different dendritic cell subsets. This review summarizes the current knowledge on the role of TGF-β family cytokines in dendritic cell biology, focusing on TGF-β as well as on other, less characterized, members of these important immune mediators.
Collapse
|
35
|
Serra P, Santamaria P. Nanoparticle-based autoimmune disease therapy. Clin Immunol 2015; 160:3-13. [PMID: 25704658 DOI: 10.1016/j.clim.2015.02.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 01/10/2023]
Abstract
The goal of immunotherapy against autoimmunity is to block pathogenic inflammation without impairing immunity against infections and tumours. Regulatory T-cells (Tregs) play a central role in maintaining immune homeostasis, and autoimmune inflammation is frequently associated with decreased numbers and/or function of these T-cells. Therapies harnessing Tregs to treat autoimmune inflammation remain under-developed with caveats ranging from the lack of antigenic and disease specificity to the potential phenotypic and functional instability of in vitro-expanded Treg cells in vivo. Here, we review nanotechnology-based approaches designed to promote immune tolerance through various mechanisms, ranging from systemic or local suppression of antigen-presenting cells and deletion of antigen-specific T-cells, to the systemic expansion of antigen- and disease-specific Treg cells in vivo.
Collapse
Affiliation(s)
- Pau Serra
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain.
| | - Pere Santamaria
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain; Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cummings School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
36
|
Zandman-Goddard G, Pierangeli SS, Gertel S, Blank M. Tolerogenic dendritic cells specific for β2-glycoprotein-I Domain-I, attenuate experimental antiphospholipid syndrome. J Autoimmun 2014; 54:72-80. [PMID: 24972993 DOI: 10.1016/j.jaut.2014.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/04/2014] [Indexed: 11/19/2022]
Abstract
Tolerogenic dendritic cells (tDCs) have the potential to control the outcome of autoimmunity by modulating the immune response. The aim of this study was to uncover the tolerance efficacy attributed to beta-2-glycoprotein-I (β2GPI) tDCs or β2GPI domain-I (D-I) and domain-V (D-V)-tDCs in mice with antiphospholipid syndrome (APS). tDCs were pulsed with β2GPI or D-I or D-V derivatives. Our results revealed that β2GPI related tDCs phenotype includes CD80(high), CD86(high) CD40(high) MHC class II(high). The miRNA profiling encompass miRNA 23b(high), miRNA 142-3p(low) and miRNA 221(low). In addition the β2GPI related tDCs showed reduced secretion of IL-1β, IL-12 and IL-23. D-I tDCs treatment was more efficient than β2GPI tDCs in inducing of tolerance in APS mice, manifested by lowered titers of anti- β2GPI antibodies (Abs) and reduced percentage of fetal loss. Tolerance induction was accompanied by poor T cell response to β2GPI, high numbers of CD4 + CD25 + FOXP3 + T-regulatory cells (Treg), reduced levels of IFNγ, IL-17 and increased expression of IL-10 and TGFβ. Tolerance was successfully transferred by Treg cells from the tolerized mice to β2GPI immunized mice. We conclude that predominantly D-I-tDCs and β2GPI tDCs have the potential to attenuate experimental APS by induction of Treg cells, reduction of anti- β2GPI Abs titers and increased expression of anti-inflammatory cytokines. We suggest that β2-GPI-D-I-tDCs may offer a novel approach for developing therapy for APS patients.
Collapse
Affiliation(s)
- Gisele Zandman-Goddard
- The Zabludowicz Center For Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Sackler Faculty of Medicine Tel-Aviv University, 52621, Israel; Department of Medicine C, Wolfson Medical Center, Sackler Faculty of Medicine Tel-Aviv University, Holon, Israel
| | - Silvia S Pierangeli
- Antiphospholipid Standardization Laboratory, Division of Rheumatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Smadar Gertel
- The Zabludowicz Center For Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Sackler Faculty of Medicine Tel-Aviv University, 52621, Israel
| | - Miri Blank
- The Zabludowicz Center For Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Sackler Faculty of Medicine Tel-Aviv University, 52621, Israel.
| |
Collapse
|