1
|
Dong L, Wu N, Cai Z, Yan S, Pan J, Yang L, Zhu J, Wang Y, Li J, Tao J. Tolerogenic Monocyte-Derived Langerhans Cells Promote Melanoma Progression and Immunotherapy Resistance through Aryl Hydrocarbon Receptor-Cyclooxygenase-2 Activation. J Invest Dermatol 2025:S0022-202X(25)00404-X. [PMID: 40216154 DOI: 10.1016/j.jid.2025.02.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 04/27/2025]
Abstract
Langerhans cells (LCs), as epidermal-resident antigen-presenting cells, are among the first to encounter early carcinogenic changes in the skin, such as those seen in melanoma. During inflammation or cancer, circulating monocytes are recruited into the epidermis, replacing resident LCs that have migrated to draining lymph nodes. However, the specific roles of LC subsets in the progression of melanoma remain unclear. In this study, we observed a decrease in resident LCs and an increase in monocyte-derived LCs (moLCs) within melanoma tumor tissues as the disease progressed. Notably, the frequency of moLCs was higher in patients with anti-PD-1 therapy-resistant melanoma than in those responsive to the therapy. Using muLangerin-DTR transgenic and moLC-specific knockout mouse models, we demonstrated that the depletion of resident LCs accelerated melanoma growth, whereas the depletion of moLCs suppressed tumor progression. Mechanistically, moLCs exhibited elevated expression of cyclooxygenase-2-related genes compared with resident LCs. TGF-β was found to activate aryl hydrocarbon receptor-cyclooxygenase-2 signaling in moLCs, leading to increased production of prostaglandin E2 and expression of PD-L1, which collectively contributed to the immunosuppressive effects of moLCs on CD8+ cytotoxic T lymphocytes. In summary, our findings highlight the functional heterogeneity of LC subsets in melanoma progression, offering deeper insights into LC biology and potential therapeutic strategies.
Collapse
Affiliation(s)
- Liyun Dong
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Naming Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Zhen Cai
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Shiyi Yan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Juan Pan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Yujue Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Jun Li
- Department of Dermatology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China.
| |
Collapse
|
2
|
Nakashima K, Imai T, Shiraishi A, Unose R, Goto H, Nagatomo Y, Kojima-Ishii K, Mushimoto Y, Nishiyama K, Yamamura K, Nagata H, Ishimura M, Kusuhara K, Koga Y, Sakai Y, Ohga S. The immunoreactive signature of monocyte-derived dendritic cells from patients with Down syndrome. Clin Exp Immunol 2024; 217:291-299. [PMID: 38916251 PMCID: PMC11310712 DOI: 10.1093/cei/uxae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/17/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024] Open
Abstract
The clinical spectrum of Down syndrome (DS) ranges from congenital malformations to premature aging and early-onset senescence. Excessive immunoreactivity and oxidative stress are thought to accelerate the pace of aging in DS patients; however, the immunological profile remains elusive. We investigated whether peripheral blood monocyte-derived dendritic cells (MoDCs) in DS patients respond to lipopolysaccharide (LPS) distinctly from non-DS control MoDCs. Eighteen DS patients (age 2-47 years, 12 males) and 22 controls (age 4-40 years, 15 males) were enrolled. CD14-positive monocytes were immunopurified and cultured for 7 days in the presence of granulocyte-macrophage colony-stimulating factor and IL-4, yielding MoDCs in vitro. After the LPS-stimulation for 48 hours from days 7 to 9, culture supernatant cytokines were measured by multiplex cytokine bead assays, and bulk-prepared RNA from the cells was used for transcriptomic analyses. MoDCs from DS patients produced cytokines/chemokines (IL-6, IL-8, TNF-α, MCP-1, and IP-10) at significantly higher levels than those from controls in response to LPS. RNA sequencing revealed that DS-derived MoDCs differentially expressed 137 genes (74 upregulated and 63 downregulated) compared with controls. A gene enrichment analysis identified 5 genes associated with Toll-like receptor signaling (KEGG: hsa04620, P = 0.00731) and oxidative phosphorylation (hsa00190, P = 0.0173) pathways. MoDCs obtained from DS patients showed higher cytokine or chemokine responses to LPS than did control MoDCs. Gene expression profiles suggest that hyperactive Toll-like receptor and mitochondrial oxidative phosphorylation pathways configure the immunoreactive signature of MoDCs in DS patients.
Collapse
Affiliation(s)
- Kentaro Nakashima
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Imai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Shiraishi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoko Unose
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironori Goto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusaku Nagatomo
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kanako Kojima-Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Mushimoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei Nishiyama
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichiro Yamamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hazumu Nagata
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuhki Koga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
3
|
Skariah N, James OJ, Swamy M. Signalling mechanisms driving homeostatic and inflammatory effects of interleukin-15 on tissue lymphocytes. DISCOVERY IMMUNOLOGY 2024; 3:kyae002. [PMID: 38405398 PMCID: PMC10883678 DOI: 10.1093/discim/kyae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
There is an intriguing dichotomy in the function of cytokine interleukin-15-at low levels, it is required for the homeostasis of the immune system, yet when it is upregulated in response to pathogenic infections or in autoimmunity, IL-15 drives inflammation. IL-15 associates with the IL-15Rα within both myeloid and non-haematopoietic cells, where IL-15Rα trans-presents IL-15 in a membrane-bound form to neighboring cells. Alongside homeostatic maintenance of select lymphocyte populations such as NK cells and tissue-resident T cells, when upregulated, IL-15 also promotes inflammatory outcomes by driving effector function and cytotoxicity in NK cells and T cells. As chronic over-expression of IL-15 can lead to autoimmunity, IL-15 expression is tightly regulated. Thus, blocking dysregulated IL-15 and its downstream signalling pathways are avenues for immunotherapy. In this review we discuss the molecular pathways involved in IL-15 signalling and how these pathways contribute to both homeostatic and inflammatory functions in IL-15-dependent mature lymphoid populations, focusing on innate, and innate-like lymphocytes in tissues.
Collapse
Affiliation(s)
- Neema Skariah
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Olivia J James
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Mahima Swamy
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
4
|
Alimohammadi S, Pénzes Z, Horváth D, Gyetvai Á, Bácsi A, Kis NG, Németh Á, Arany J, Oláh A, Lisztes E, Tóth BI, Bíró T, Szöllősi AG. TRPV4 Activation Increases the Expression of CD207 (Langerin) of Monocyte-Derived Langerhans Cells without Affecting their Maturation. J Invest Dermatol 2022; 143:801-811.e10. [PMID: 36502939 DOI: 10.1016/j.jid.2022.10.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 12/14/2022]
Abstract
Langerhans cells (LCs) are the sole professional antigen-presenting cell normally found in the human epidermal compartment. Research into their physiological role is hindered by the fact that they are invariably activated during isolation from the skin. To overcome this challenge, we turned to a monocyte-derived LC (moLC) model, which we characterized with RNA sequencing, and compared the transcriptome of moLCs with that of donor-matched immature dendritic cells. We found that moLCs express markers characteristic of LC2 cells as well as TRPV4. TRPV4 is especially important in the skin because it has been linked to the conservation of the skin barrier, immunological responses, as well as acute and chronic itch, but we know little about its function on LCs. Our results show that TRPV4 activation increased the expression of Langerin and led to increased intracellular calcium concentration in moLCs. Regarding the functionality of moLCs, we found that TRPV4 agonism had a mitigating effect on their inflammatory responses because it decreased their cytokine production and T-cell activating capability. Because TRPV4 has emerged as a potential therapeutic target in dermatological conditions, it is important to highlight LCs as, to our knowledge, a previously unreported target of these therapies.
Collapse
Affiliation(s)
- Shahrzad Alimohammadi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pénzes
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Dorottya Horváth
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Gyetvai
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ákos Németh
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Health Sciences, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - József Arany
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erika Lisztes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs István Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, Münster, Germany
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
5
|
Fu C, Ma T, Zhou L, Mi QS, Jiang A. Dendritic Cell-Based Vaccines Against Cancer: Challenges, Advances and Future Opportunities. Immunol Invest 2022; 51:2133-2158. [PMID: 35946383 DOI: 10.1080/08820139.2022.2109486] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As the most potent professional antigen presenting cells, dendritic cells (DCs) have the ability to activate both naive CD4 and CD8 T cells. Recognized for their exceptional ability to cross-present exogenous antigens to prime naive antigen-specific CD8 T cells, DCs play a critical role in generating CD8 T cell immunity, as well as mediating CD8 T cell tolerance to tumor antigens. Despite the ability to potentiate host CD8 T cell-mediated anti-tumor immunity, current DC-based cancer vaccines have not yet achieved the promised success clinically with the exception of FDA-approved Provenge. Interestingly, recent studies have shown that type 1 conventional DCs (cDC1s) play a critical role in cross-priming tumor-specific CD8 T cells and determining the anti-tumor efficacy of cancer immunotherapies including immune checkpoint blockade (ICB). Together with promising clinical results in neoantigen-based cancer vaccines, there is a great need for DC-based vaccines to be further developed and refined either as monotherapies or in combination with other immunotherapies. In this review, we will present a brief review of DC development and function, discuss recent progress, and provide a perspective on future directions to realize the promising potential of DC-based cancer vaccines.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Tianle Ma
- Department of Computer Science and Engineering, School of Engineering and Computer Science, Oakland University, Rochester, Michigan, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
6
|
Xuan S, Li Y, Wu Y, Adcock IM, Zeng X, Yao X. Langerin-expressing dendritic cells in pulmonary immune-related diseases. Front Med (Lausanne) 2022; 9:909057. [PMID: 36160158 PMCID: PMC9490018 DOI: 10.3389/fmed.2022.909057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) are “frontline” immune cells dedicated to antigen presentation. They serve as an important bridge connecting innate and adaptive immunity, and express various receptors for antigen capture. DCs are divided into various subclasses according to their differential expression of cell surface receptors and different subclasses of DCs exhibit specific immunological characteristics. Exploring the common features of each sub-category has became the focus of many studies. There are certain amounts of DCs expressing langerin in airways and peripheral lungs while the precise mechanism by which langerin+ DCs drive pulmonary disease is unclear. Langerin-expressing DCs can be further subdivided into numerous subtypes based on the co-expressed receptors, but here, we identify commonalities across these subtypes that point to the major role of langerin. Better understanding is required to clarify key disease pathways and determine potential new therapeutic approaches.
Collapse
Affiliation(s)
- Shurui Xuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuebei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunhui Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ian M. Adcock
- Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xiaoning Zeng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Xin Yao
| |
Collapse
|
7
|
The Roles of Skin Langerhans Cells in Immune Tolerance and Cancer Immunity. Vaccines (Basel) 2022; 10:vaccines10091380. [PMID: 36146458 PMCID: PMC9503294 DOI: 10.3390/vaccines10091380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 12/19/2022] Open
Abstract
Langerhans cells (LC) are a unique population of tissue-resident macrophages with dendritic cell (DC) functionality that form a network of cells across the epidermis of the skin. Their location at the skin barrier suggests an important role for LC as immune sentinels at the skin surface. The classification of LC as DC over the past few decades has driven the scientific community to extensively study how LC function as DC-like cells that prime T cell immunity. However, LC are a unique type of tissue-resident macrophages, and recent evidence also supports an immunoregulatory role of LC at steady state and during specific inflammatory conditions, highlighting the impact of cutaneous environment in shaping LC functionality. In this mini review, we discuss the recent literature on the immune tolerance function of LC in homeostasis and disease conditions, including malignant transformation and progression; as well as LC functional plasticity for adaption to microenvironmental cues and the potential connection between LC population heterogeneity and functional diversity. Future investigation into the molecular mechanisms that LC use to integrate different microenvironment cues and adapt immunological responses for controlling LC functional plasticity is needed for future breakthroughs in tumor immunology, vaccine development, and treatments for inflammatory skin diseases.
Collapse
|
8
|
Development of a serum-free induction medium for the induction of human CD40+CD209+ dendritic cells from CD14+ monocytes. J Taiwan Inst Chem Eng 2022. [DOI: 10.1016/j.jtice.2022.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
9
|
Combes TW, Orsenigo F, Stewart A, Mendis ASJR, Dunn-Walters D, Gordon S, Martinez FO. CSF1R defines the mononuclear phagocyte system lineage in human blood in health and COVID-19. IMMUNOTHERAPY ADVANCES 2021; 1:ltab003. [PMID: 35915730 PMCID: PMC7928847 DOI: 10.1093/immadv/ltab003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/31/2022] Open
Abstract
Summary
Mononuclear phagocytes defend tissues, present antigens, and mediate recovery and healing. To date, we lack a marker to unify mononuclear phagocytes in humans or that informs us about their origin. Here, we reassess mononuclear phagocyte ontogeny in human blood through the lineage receptor CSF1R, in the steady state and in COVID-19. We define CSF1R as the first sensitive and reproducible pan-phagocyte lineage marker, to identify and enumerate all conventional monocytes, and the myeloid dendritic cells. In the steady state, CSF1R is sufficient for sorting and immuno-magnetic isolation. In pathology, changes in CSF1R are more sensitive than CD14 and CD16. In COVID-19, a significant drop in membrane CSF1R is useful for stratifying patients, beyond the power of cell categories published thus far, which fail to capture COVID-19 specific events. Importantly, CSF1R defines cells which are neither conventional monocytes nor DCs, which are missed in published analysis. CSF1R decrease can be linked ex vivo to high CSF1 levels. Blood assessment of CSF1R+ cells opens a developmental window to the Mononuclear Phagocyte System in transit from bone marrow to tissues, supports isolation and phenotypic characterisation, identifies novel cell types, and singles out CSF1R inhibition as therapeutic target in COVID-19 and other diseases.
Collapse
Affiliation(s)
- Theo W Combes
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Federica Orsenigo
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Alexander Stewart
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | | | | | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | |
Collapse
|
10
|
Bellmann L, Zelle-Rieser C, Milne P, Resteu A, Tripp CH, Hermann-Kleiter N, Zaderer V, Wilflingseder D, Hörtnagl P, Theochari M, Schulze J, Rentzsch M, Del Frari B, Collin M, Rademacher C, Romani N, Stoitzner P. Notch-Mediated Generation of Monocyte-Derived Langerhans Cells: Phenotype and Function. J Invest Dermatol 2021; 141:84-94.e6. [PMID: 32522485 PMCID: PMC7758629 DOI: 10.1016/j.jid.2020.05.098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 01/16/2023]
Abstract
Langerhans cells (LCs) in the skin are a first line of defense against pathogens but also play an essential role in skin homeostasis. Their exclusive expression of the C-type lectin receptor Langerin makes them prominent candidates for immunotherapy. For vaccine testing, an easily accessible cell platform would be desirable as an alternative to the time-consuming purification of LCs from human skin. Here, we present such a model and demonstrate that monocytes in the presence of GM-CSF, TGF-β1, and the Notch ligand DLL4 differentiate within 3 days into CD1a+Langerin+cells containing Birbeck granules. RNA sequencing of these monocyte-derived LCs (moLCs) confirmed gene expression of LC-related molecules, pattern recognition receptors, and enhanced expression of genes involved in the antigen-presenting machinery. On the protein level, moLCs showed low expression of costimulatory molecules but prominent expression of C-type lectin receptors. MoLCs can be matured, secrete IL-12p70 and TNF-α, and stimulate proliferation and cytokine production in allogeneic CD4+ and CD8+ T cells. In regard to vaccine testing, a recently characterized glycomimetic Langerin ligand conjugated to liposomes demonstrated specific and fast internalization into moLCs. Hence, these short-term in vitro‒generated moLCs represent an interesting tool to screen LC-based vaccines in the future.
Collapse
Key Words
- a647, alexafluor-647
- dc, dendritic cell
- lc, langerhans cell
- mhc, major histocompatibility complex
- mlr, mixed leukocyte reaction
- molc, monocyte-derived lc
- polyi:c, polyinosinic:polycytidylic acid
- rna-seq, rna sequencing
- th, t helper
- tlr, toll-like receptor
Collapse
Affiliation(s)
- Lydia Bellmann
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Zelle-Rieser
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul Milne
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anastasia Resteu
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christoph H Tripp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Viktoria Zaderer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul Hörtnagl
- Central Institute for Blood Transfusion and Immunological Department, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Theochari
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jessica Schulze
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Mareike Rentzsch
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthew Collin
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Nikolaus Romani
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Human genetic dissection of papillomavirus-driven diseases: new insight into their pathogenesis. Hum Genet 2020; 139:919-939. [PMID: 32435828 DOI: 10.1007/s00439-020-02183-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Human papillomaviruses (HPVs) infect mucosal or cutaneous stratified epithelia. There are 5 genera and more than 200 types of HPV, each with a specific tropism and virulence. HPV infections are typically asymptomatic or result in benign tumors, which may be disseminated or persistent in rare cases, but a few oncogenic HPVs can cause cancers. This review deals with the human genetic and immunological basis of interindividual clinical variability in the course of HPV infections of the skin and mucosae. Typical epidermodysplasia verruciformis (EV) is characterized by β-HPV-driven flat wart-like and pityriasis-like cutaneous lesions and non-melanoma skin cancers in patients with inborn errors of EVER1-EVER2-CIB1-dependent skin-intrinsic immunity. Atypical EV is associated with other infectious diseases in patients with inborn errors of T cells. Severe cutaneous or anogenital warts, including anogenital cancers, are also driven by certain α-, γ-, μ or ν-HPVs in patients with inborn errors of T lymphocytes and antigen-presenting cells. The genetic basis of HPV diseases at other mucosal sites, such as oral multifocal epithelial hyperplasia or juvenile recurrent respiratory papillomatosis (JRRP), remains poorly understood. The human genetic dissection of HPV-driven lesions will clarify the molecular and cellular basis of protective immunity to HPVs, and should lead to novel diagnostic, preventive, and curative approaches in patients.
Collapse
|
12
|
Zhang LX, Chen RL, Liao XY, You X, Gao FG. Ex vivo IL-15 replenishment augments bone marrow precursor cell-mediated adaptive immunity via PI3K-Akt pathway. J Leukoc Biol 2020; 108:177-188. [PMID: 32293057 DOI: 10.1002/jlb.1ma0220-337rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/11/2020] [Accepted: 03/18/2020] [Indexed: 01/10/2023] Open
Abstract
This study tested the hypothesis that PI3K-Akt activity contributes to the superior immune function of IL-15-administrated bone marrow precursor cells (BMPC). Our previous studies revealed that PI3K-Akt play vital role in dendritic cells (DCs) cross-presentation and DC-based CTL priming. Despite the fact that IL-15 serves multiple functions in its therapeutic potential for the induction and maintenance of T cell response, the exact role of PI3K-Akt in IL-15 increased adaptive immunity is still poorly understood. In this study, we demonstrated that ex vivo IL-15 administration increased BMPC capability of antigen uptake and the expression of costimulatory molecules (such as CD80 and 4-1BB(CD137) ligand [4-1BBL]) and MHC class I molecule via PI3K-Akt pathway. Importantly, PI3K-Akt activity was not only necessary for IL-15 augmented BMPC cross-presentation and CTL priming, but also facilitated IL-15 increased therapeutic potential of the cytolytic capacity and maintenance of BMPC-activated T cells. Thus, these data suggested that PI3K-Akt activity contribute to the superior immune function of IL-15-administrated BMPC and thereby might be therapeutic potential for adaptive immunity.
Collapse
Affiliation(s)
- Li Xiao Zhang
- Department of Basic Medicine Science, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Rui Ling Chen
- Department of Basic Medicine Science, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Xiao Yan Liao
- Department of Basic Medicine Science, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Xiang You
- Department of Basic Medicine Science, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Feng Guang Gao
- Department of Basic Medicine Science, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shang Hai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Abstract
During the past few years, there has been a substantial increase in the understanding of innate immunity. Dendritic cells are emerging as key players in the orchestration of this early phase of immune responses, with a role that will translate into the subsequent type of adaptive immune response against infection. Here we provide an overview of dendritic cell differentiation and function, with particular emphasis on those features unique to the immune defense of the peritoneal cavity and in the context of peritoneal dialysis-associated immune responses. The reader is referred to the primary references included in the accompanying list for specific details in this fascinating field.
Collapse
Affiliation(s)
- Michelle L. McCully
- The FOCIS Centre for Clinical Immunology and Immunotherapeutics, Robarts Research Institute, and the Departments of Microbiology and Immunology, and Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Joaquín Madrenas
- The FOCIS Centre for Clinical Immunology and Immunotherapeutics, Robarts Research Institute, and the Departments of Microbiology and Immunology, and Medicine, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
14
|
Pereiro P, Figueras A, Novoa B. Insights into teleost interferon-gamma biology: An update. FISH & SHELLFISH IMMUNOLOGY 2019; 90:150-164. [PMID: 31028897 DOI: 10.1016/j.fsi.2019.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/20/2019] [Accepted: 04/02/2019] [Indexed: 06/09/2023]
Abstract
Interferon-gamma (IFN-ϒ) is probably one of the most relevant cytokines orchestrating the immune response in vertebrates. Although the activities mediated by this molecule are well known in mammals, several aspects of the IFN-ϒ system in teleosts remain a riddle to scientists. Numerous studies support a potentially similar role of the fish IFN-ϒ signalling pathway in some well-described immunological processes induced by this cytokine in mammals. Nevertheless, the existence in some teleost species of duplicated ifng genes and an additional gene derived from ifng known as interferon-γ-related (ifngrel), among other things, raises new interesting questions about the mode of action of these various molecules in fish. Moreover, certain IFN-ϒ-mediated activities recently observed in mammals are still fully unknown in fish. Another attractive but mainly unexplored curious property of IFN-ϒ in vertebrates is its potential dual role depending on the type of pathogen. In addition, some aspects mediated by this molecule could favour the resolution of a bacterial infection but be harmful in the context of a viral disease, and vice versa. This review collects old and new aspects of IFN-ϒ research in teleosts and discusses new questions and pathways of investigation based on recent discoveries in mammals.
Collapse
Affiliation(s)
- Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM), CSIC, Vigo, Spain; Laboratory of Biotechnology and Aquatic Genomics, Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile
| | | | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM), CSIC, Vigo, Spain.
| |
Collapse
|
15
|
Abstract
With the spotlight on cancer immunotherapy and the expanding use of immune checkpoint inhibitors, strategies to improve the response rate and duration of current cancer immunotherapeutics are highly sought. In that sense, investigators around the globe have been putting spurs on the development of effective cancer vaccines in humans after decades of efforts that led to limited clinical success. In more than three decades of research in pursuit of targeted and personalized immunotherapy, several platforms have been incorporated into the list of cancer vaccines from live viral or bacterial agents harboring antigens to synthetic peptides with the hope of stronger and durable immune responses that will tackle cancers better. Unlike adoptive cell therapy, cancer vaccines can take advantage of using a patient's entire immune system that can include more than engineered receptors or ligands in developing antigen-specific responses. Advances in molecular technology also secured the use of genetically modified genes or proteins of interest to enhance the chance of stronger immune responses. The formulation of vaccines to increase chances of immune recognition such as nanoparticles for peptide delivery is another area of great interest. Studies indicate that cancer vaccines alone may elicit tumor-specific cellular or humoral responses in immunologic assays and even regression or shrinkage of the cancer in select trials, but novel strategies, especially in combination with other cancer therapies, are under study and are likely to be critical to achieve and optimize reliable objective responses and survival benefit. In this review, cancer vaccine platforms with different approaches to deliver tumor antigens and boost immunity are discussed with the intention of summarizing what we know and what we need to improve in the clinical trial setting.
Collapse
Affiliation(s)
- Hoyoung M. Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
16
|
IL-15 and a Two-Step Maturation Process Improve Bone Marrow-Derived Dendritic Cell Cancer Vaccine. Cancers (Basel) 2019; 11:cancers11010040. [PMID: 30621204 PMCID: PMC6356194 DOI: 10.3390/cancers11010040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
In the last 20 years, dendritic cells (DCs) have been largely used as a platform for therapeutic vaccination in cancer patients. However, despite its proven safety and ability to induce cancer specific immune responses, the clinical benefits of DC-based immunotherapy are currently very limited. Thus, novel approaches are still needed to boost its efficacy. Our group recently showed that squaric acid treatment of antigens is an important adjuvant that can increase vaccine-induced downstream immune responses and therapeutic outcomes. Here we further improved this dendritic cell vaccine formulation by developing a new method for differentiating and maturing DCs from their bone marrow precursors. Our data demonstrate that bone marrow-derived DCs differentiated with GM-CSF and IL-15 and matured with a maturation cocktail in two steps present a more mature and immunogenic phenotype, compared to standard DC preparations. Further suppression of the prostaglandin E₂ pathway achieved even more immunogenic DC phenotypes. This vaccine was more potent at delaying tumor growth, improved animal survival and induced a more immunogenic and Th1-skewed T cell response in an ovarian cancer mouse model. These promising results support future efforts for the clinical translation of this approach.
Collapse
|
17
|
CECHIM GIOVANA, CHIES JOSÉA. In vitro generation of human monocyte-derived dendritic cells methodological aspects in a comprehensive review. ACTA ACUST UNITED AC 2019. [DOI: 10.1590/0001-3765201920190310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Kwon KW, Kim SJ, Kim H, Kim WS, Kang SM, Choi E, Ha SJ, Yoon JH, Shin SJ. IL-15 Generates IFN-γ-producing Cells Reciprocally Expressing Lymphoid-Myeloid Markers during Dendritic Cell Differentiation. Int J Biol Sci 2019; 15:464-480. [PMID: 30745835 PMCID: PMC6367559 DOI: 10.7150/ijbs.25743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 12/02/2018] [Indexed: 11/23/2022] Open
Abstract
Recently, interest in IL-15-differentiated cells has increased; however, the phenotypic definition of IL-15-differentiated bone marrow-derived cells (IL-15-DBMCs) is still under debate, particularly the generation of IFN-γ-producing innate cells such as premature NK (pre-mNK) cells, natural killer dendritic cells (NKDCs), interferon-producing killer dendritic cells (IKDCs), and type 1 innate lymphoid cells (ILC1s), all of which are IL-15-dependent. Here, we revisited the immunophenotypic characteristics of IFN-γ-producing IL-15-DBMCs and their functional role in the control of intracellular Mycobacterium tuberculosis (Mtb) infection. When comparing the cytokine levels between bone marrow-derived dendritic cells (BMDCs) and IL-15-DBMCs upon stimulation with various TLR agonists, only the CD11cint population of IL-15-DBMCs produced significant levels of IFN-γ, decreased levels of MHC-II, and increased levels of B220. Neither BMDCs nor IL-15-DBMCs were found to express DX5 or NK1.1, which are representative markers for the NK cell lineage and IKDCs. When the CD11cintB220+ population of IL-15-DBMCs was enriched, the Thy1.2+Sca-1+ population showed a marked increase in IFN-γ production. In addition, while depletion of the B220+ and Thy1.2+ populations of IL-15-DBMCs, but not the CD19+ population, inhibited IFN-γ production, enrichment of these cell populations increased IFN-γ. Ultimately, co-culture of sorted IFN-γ-producing B220+Thy1.2+ IL-15-DBMCs with Mtb-infected macrophages resulted in control of the intracellular growth of Mtb via the IFN-γ-nitric oxide axis in a donor cell number-dependent manner. Taken together, the results indicate that IFN-γ-producing IL-15-DBMCs could be redefined as CD11cintB220+Thy1.2+Sca-1+ cells, which phenotypically resemble both IKDCs and ILC1s, and may have therapeutic potential for controlling infectious intracellular bacteria such as Mtb.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - So Jeong Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soon Myung Kang
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, South Korea
| | - Joo-Heon Yoon
- The Airway Mucus Institute, and Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
19
|
Song J, Lang F, Zhao N, Guo Y, Zhang H. Vaginal Lactobacilli Induce Differentiation of Monocytic Precursors Toward Langerhans-like Cells: in Vitro Evidence. Front Immunol 2018; 9:2437. [PMID: 30410487 PMCID: PMC6211368 DOI: 10.3389/fimmu.2018.02437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/02/2018] [Indexed: 01/17/2023] Open
Abstract
Lactobacilli have immunomodulatory mechanisms that affect the host cell immune system, leading to inhibition of HIV-1 transmission. Thus, lactobacilli as mucosal delivery vehicles for developing HIV-1 vaccines have attracted interest in recent years. Herein, we investigated the immunomodulatory effects of six strains of Lactobacillus naturally isolated from vaginal samples, including Lactobacillus crispatus (L. crispatus), L. fermentum, L. jensenii, L. gasseri, L. delbrueckii and L. johnsonii, on differentiation of monocytic precursors. L. crispatus, L. fermentum and L. delbrueckii could drive human monocytic cell line THP-1 cells to differentiate into dendritic-like cells according to the morphology. Moreover, L. crispatus increased costimulatory molecules including CD40, CD80 and CD86, and Langerhans cell specific C-type lectin receptors CD207, while L. fermentum decreased these molecules in THP-1 cells. Furthermore, L. crispatus promoted the differentiation of THP-1 cells with specific markers, phagocytic features, cytokine production ability and reduced the expression of receptors for HIV-1 entry of Langerhans cells. However, in the presence of L. fermentum, THP-1 cells did not show the above alterations. Moreover, similar effects of L. crispatus and L. fermentum were observed in CD14+ monocytes. These data suggested that L. crispatus facilitates the differentiation of monocytic precursors toward Langerhans-like cells in vitro. We further identified the cell wall components of Lactobacillus and found that peptidoglycans (PGNs), rather than bacteriocins, S-layer protein and lipoteichoic acid, were key contributors to the induction of CD207 expression. However, PGNs originating from Bacillus subtilis, E. coli JM109 and E. coli DH5α did not elevate CD207 expression, indicating that only PGN derived from Lactobacillus could enhance CD207 expression. Finally, the recognized receptors of L. crispatus (such as TLR2 and TLR6) and the upstream transcription factors (PU.1, TAL1, TIF1γ, and POLR2A) of CD207 were examined, and the expression of these molecules was enhanced in THP-1 cells following L. crispatus treatment. Thus, this study offers powerful evidence that vaginal lactobacilli modulate monocytic precursor differentiation into Langerhans-like cells probably via activating the TLR2/6-TFs-CD207 axis. These data provide clues for further investigation of the original occurrence, development and differentiation of Langerhans cells from monocytes.
Collapse
Affiliation(s)
- Jie Song
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China.,Chongqing Center for Biomedical Research and Equipment Development, Chongqing Academy of Science and Technology, Chongqing, China.,Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Fengchao Lang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Na Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Yan Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Huatang Zhang
- Chongqing Center for Biomedical Research and Equipment Development, Chongqing Academy of Science and Technology, Chongqing, China.,Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
20
|
Lin JX, Leonard WJ. The Common Cytokine Receptor γ Chain Family of Cytokines. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028449. [PMID: 29038115 DOI: 10.1101/cshperspect.a028449] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15, and IL-21 form a family of cytokines based on their sharing the common cytokine receptor γ chain (γc), which was originally discovered as the third receptor component of the IL-2 receptor, IL-2Rγ. The IL2RG gene is located on the X chromosome and is mutated in humans with X-linked severe combined immunodeficiency (XSCID). The breadth of the defects in XSCID could not be explained solely by defects in IL-2 signaling, and it is now clear that γc is a shared receptor component of the six cytokines noted above, making XSCID a disease of defective cytokine signaling. Janus kinase (JAK)3 associates with γc, and JAK3-deficient SCID phenocopies XSCID, findings that served to stimulate the development of JAK3 inhibitors as immunosuppressants. γc family cytokines collectively control broad aspects of lymphocyte development, growth, differentiation, and survival, and these cytokines are clinically important, related to allergic and autoimmune diseases and cancer as well as immunodeficiency. In this review, we discuss the actions of these cytokines, their critical biological roles and signaling pathways, focusing mainly on JAK/STAT (signal transducers and activators of transcription) signaling, and how this information is now being used in clinical therapeutic efforts.
Collapse
Affiliation(s)
- Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1674
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1674
| |
Collapse
|
21
|
Innate Immune Response in Kidney Ischemia/Reperfusion Injury: Potential Target for Therapy. J Immunol Res 2017; 2017:6305439. [PMID: 28676864 PMCID: PMC5476886 DOI: 10.1155/2017/6305439] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/17/2017] [Indexed: 01/06/2023] Open
Abstract
Acute kidney injury caused by ischemia and subsequent reperfusion is associated with a high rate of mortality and morbidity. Ischemia/reperfusion injury in kidney transplantation causes delayed graft function and is associated with more frequent episodes of acute rejection and progression to chronic allograft nephropathy. Alloantigen-independent inflammation is an important process, participating in pathogenesis of injurious response, caused by ischemia and reperfusion. This innate immune response is characterized by the activity of classical cells belonging to the immune system, such as neutrophils, macrophages, dendritic cells, lymphocytes, and also tubular epithelial cells and endothelial cells. These immune cells not only participate in inflammation after ischemia exerting detrimental influence but also play a protective role in the healing response from ischemia/reperfusion injury. Delineating of complex mechanisms of their actions could be fruitful in future prevention and treatment of ischemia/reperfusion injury. Among numerous so far conducted experiments, observed immunomodulatory role of adenosine and adenosine receptor agonists in complex interactions of dendritic cells, natural killer T cells, and T regulatory cells is emphasized as promising in the treatment of kidney ischemia/reperfusion injury. Potential pharmacological approaches which decrease NF-κB activity and antagonize mechanisms downstream of activated Toll-like receptors are discussed.
Collapse
|
22
|
Heterogeneity, functional specialization and differentiation of monocyte‐derived dendritic cells. Immunol Cell Biol 2016; 95:244-251. [DOI: 10.1038/icb.2016.104] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023]
|
23
|
Gruenbacher G, Gander H, Rahm A, Idzko M, Nussbaumer O, Thurnher M. Ecto-ATPase CD39 Inactivates Isoprenoid-Derived Vγ9Vδ2 T Cell Phosphoantigens. Cell Rep 2016; 16:444-456. [PMID: 27346340 DOI: 10.1016/j.celrep.2016.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/29/2016] [Accepted: 05/26/2016] [Indexed: 01/11/2023] Open
Abstract
In humans, Vγ9Vδ2 T cells respond to self and pathogen-associated, diphosphate-containing isoprenoids, also known as phosphoantigens (pAgs). However, activation and homeostasis of Vγ9Vδ2 T cells remain incompletely understood. Here, we show that pAgs induced expression of the ecto-ATPase CD39, which, however, not only hydrolyzed ATP but also abrogated the γδ T cell receptor (TCR) agonistic activity of self and microbial pAgs (C5 to C15). Only mevalonate-derived geranylgeranyl diphosphate (GGPP, C20) resisted CD39-mediated hydrolysis and acted as a regulator of CD39 expression and activity. GGPP enhanced macrophage differentiation in response to the tissue stress cytokine interleukin-15. In addition, GGPP-imprinted macrophage-like cells displayed increased capacity to produce IL-1β as well as the chemokine CCL2 and preferentially activated CD161-expressing CD4(+) T cells in an innate-like manner. Our studies reveal a previously unrecognized immunoregulatory function of CD39 and highlight a particular role of GGPP among pAgs.
Collapse
Affiliation(s)
- Georg Gruenbacher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Hubert Gander
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Andrea Rahm
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria
| | - Marco Idzko
- Department of Pulmonary Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Oliver Nussbaumer
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, UK
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck and K1 Center Oncotyrol-Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria.
| |
Collapse
|
24
|
Picarda G, Chéneau C, Humbert JM, Bériou G, Pilet P, Martin J, Duteille F, Perrot P, Bellier-Waast F, Heslan M, Haspot F, Guillon F, Josien R, Halary FA. Functional Langerinhigh-Expressing Langerhans-like Cells Can Arise from CD14highCD16−Human Blood Monocytes in Serum-Free Condition. THE JOURNAL OF IMMUNOLOGY 2016; 196:3716-28. [DOI: 10.4049/jimmunol.1501304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 02/23/2016] [Indexed: 12/31/2022]
|
25
|
Smits ELJM, Stein B, Nijs G, Lion E, Van Tendeloo VF, Willemen Y, Anguille S, Berneman ZN. Generation and Cryopreservation of Clinical Grade Wilms' Tumor 1 mRNA-Loaded Dendritic Cell Vaccines for Cancer Immunotherapy. Methods Mol Biol 2016; 1393:27-35. [PMID: 27033213 DOI: 10.1007/978-1-4939-3338-9_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
First described in the 1970s, dendritic cells (DC) are currently subjects of intense investigation to exploit their unique antigen-presenting and immunoregulatory capacities. In cancer, DC show promise to elicit or amplify immune responses directed against cancer cells by activating natural killer (NK) cells and tumor antigen-specific T cells. Wilms' tumor 1 (WT1) protein is a tumor-associated antigen that is expressed in a majority of cancer types and has been designated as an antigen of major interest to be targeted in clinical cancer immunotherapy trials. In this chapter, we describe the generation, cryopreservation, and thawing of clinical grade autologous monocyte-derived DC vaccines that are loaded with WT1 by messenger RNA (mRNA) electroporation. This in-house-developed transfection method gives rise to presentation of multiple antigen epitopes and can be used for all patients without restriction of human leukocyte antigen (HLA) type.
Collapse
Affiliation(s)
- Evelien L J M Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium.
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.
| | - Barbara Stein
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Griet Nijs
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.T.431, Wilrijk, Antwerp, 2610, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
26
|
Papadopoulos A, Gagnaire A, Degos C, de Chastellier C, Gorvel JP. Brucella discriminates between mouse dendritic cell subsets upon in vitro infection. Virulence 2015; 7:33-44. [PMID: 26606688 PMCID: PMC4871654 DOI: 10.1080/21505594.2015.1108516] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Brucella is a Gram-negative bacterium responsible for brucellosis, a worldwide re-emerging zoonosis. Brucella has been shown to infect and replicate within Granulocyte macrophage colony-stimulating factor (GMCSF) in vitro grown bone marrow-derived dendritic cells (BMDC). In this cell model, Brucella can efficiently control BMDC maturation. However, it has been shown that Brucella infection in vivo induces spleen dendritic cells (DC) migration and maturation. As DCs form a complex network composed by several subpopulations, differences observed may be due to different interactions between Brucella and DC subsets. Here, we compare Brucella interaction with several in vitro BMDC models. The present study shows that Brucella is capable of replicating in all the BMDC models tested with a high infection rate at early time points in GMCSF-IL15 DCs and Flt3l DCs. GMCSF-IL15 DCs and Flt3l DCs are more activated than the other studied DC models and consequently intracellular bacteria are not efficiently targeted to the ER replicative niche. Interestingly, GMCSF-DC and GMCSF-Flt3l DC response to infection is comparable. However, the key difference between these 2 models concerns IL10 secretion by GMCSF DCs observed at 48 h post-infection. IL10 secretion can explain the weak secretion of IL12p70 and TNFα in the GMCSF-DC model and the low level of maturation observed when compared to GMCSF-IL15 DCs and Flt3l DCs. These models provide good tools to understand how Brucella induce DC maturation in vivo and may lead to new therapeutic design using DCs as cellular vaccines capable of enhancing immune response against pathogens.
Collapse
Affiliation(s)
- Alexia Papadopoulos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Aurélie Gagnaire
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Clara Degos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Chantal de Chastellier
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Jean-Pierre Gorvel
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| |
Collapse
|
27
|
Yu H, Sui Y, Wang Y, Sato N, Frey B, Xia Z, Waldmann TA, Berzofsky J. Interleukin-15 Constrains Mucosal T Helper 17 Cell Generation: Influence of Mononuclear Phagocytes. PLoS One 2015; 10:e0143001. [PMID: 26600079 PMCID: PMC4658142 DOI: 10.1371/journal.pone.0143001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 10/29/2015] [Indexed: 01/09/2023] Open
Abstract
Interleukin (IL)-15 has multiple roles in innate and adaptive immunity, especially regarding CD8+ T cells and natural killer cells. However, the role of IL-15 in regulating differentiation of T helper cell subsets and mononuclear phagocytes (MPs) in different tissues in vivo is unknown. Here we report that IL-15 indirectly regulates Th17 but not other Th subsets in the intestinal lamina propria (LP), apparently through effects on MPs. Th17 cells in the LP were more prevalent in IL-15 KO mice than their wild-type counterparts, and less prevalent in IL-15 transgenic mice than their wild-type littermates, even co-caged. MPs from the LP of these mice were sufficient to mimic the in vivo finding in vitro by skewing of cocultured wild type OVA-specific CD4+ T cells. However, production of IL-15 or lack thereof by these MPs was not sufficient to explain the skewing, as addition or blockade of IL-15 in the cultures had no effect. Rather, a skewing of the relative proportion of CD11b+, CD103+ and double positive LP MP subsets in transgenic and KO could explain the differences in Th17 cells. Thus, IL-15 may influence MP subsets in the gut in a novel way that alters the frequency of LP Th17 cells.
Collapse
Affiliation(s)
- Huifeng Yu
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
- * E-mail: (YS); (JB)
| | - Yichuan Wang
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
| | - Noriko Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Blake Frey
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
| | - Thomas A. Waldmann
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Jay Berzofsky
- Vaccine Branch, Center for Cancer Research, National Institute of Health, Bethesda, Maryland, United States of America
- * E-mail: (YS); (JB)
| |
Collapse
|
28
|
Anguille S, Smits EL, Bryant C, Van Acker HH, Goossens H, Lion E, Fromm PD, Hart DN, Van Tendeloo VF, Berneman ZN. Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy. Pharmacol Rev 2015; 67:731-53. [PMID: 26240218 DOI: 10.1124/pr.114.009456] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Although the earliest—rudimentary—attempts at exploiting the immune system for cancer therapy can be traced back to the late 18th Century, it was not until the past decade that cancer immunotherapeutics have truly entered mainstream clinical practice. Given their potential to stimulate both adaptive and innate antitumor immune responses, dendritic cells (DCs) have come under intense scrutiny in recent years as pharmacological tools for cancer immunotherapy. Conceptually, the clinical effectiveness of this form of active immunotherapy relies on the completion of three critical steps: 1) the DCs used as immunotherapeutic vehicles must properly activate the antitumor immune effector cells of the host, 2) these immune effector cells must be receptive to stimulation by the DCs and be competent to mediate their antitumor effects, which 3) requires overcoming the various immune-inhibitory mechanisms used by the tumor cells. In this review, following a brief overview of the pivotal milestones in the history of cancer immunotherapy, we will introduce the reader to the basic immunobiological and pharmacological principles of active cancer immunotherapy using DCs. We will then discuss how current research is trying to define the optimal parameters for each of the above steps to realize the full clinical potential of DC therapeutics. Given its high suitability for immune interventions, acute myeloid leukemia was chosen here to showcase the latest research trends driving the field of DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Sébastien Anguille
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Evelien L Smits
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Christian Bryant
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Heleen H Van Acker
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Herman Goossens
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Eva Lion
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Phillip D Fromm
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | | | - Viggo F Van Tendeloo
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Zwi N Berneman
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| |
Collapse
|
29
|
Massa C, Thomas C, Wang E, Marincola F, Seliger B. Different maturation cocktails provide dendritic cells with different chemoattractive properties. J Transl Med 2015; 13:175. [PMID: 26695182 PMCID: PMC4467838 DOI: 10.1186/s12967-015-0528-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/11/2015] [Indexed: 12/16/2022] Open
Abstract
Background Dendritic cells (DC) are currently implemented as immunotherapeutic strategy for the treatment of tumor patients based on their central role in the immune system. Despite good results were obtained in vitro and in animal models, their clinical use has provided limited success suggesting the requirement to optimise the protocol for their production. Methods A cDNA array was performed on FastDC obtained from the differentiation of human peripheral blood monocytes stimulated with the clinical gold standard or with two alternative maturation cocktails combining interferon (IFN)γ and ligands for different toll like receptors (TLR). Results A stronger modulation of the DC transcriptome with respect to immature DC was found in alternatively stimulated DC when compared to DC stimulated with the clinical gold standard. A major class of molecules differentially expressed using distinct DC stimulation protocols were chemokines. Validation of their differential expression pattern at the mRNA and protein level confirmed the secretion of inflammatory chemokines by the alternative DC. Functional analyses of the chemotactic properties of DC “wash out” supernatants highlighted the ability of alternative, but not of gold standard DC to efficiently recruit immune cells with a prevalence of monocytes. Effector cells belonging to the innate as well as adaptive immunity were also attracted and the interaction with alternative DC resulted in enhanced secretion of IFNγ and induction of cytotoxic activity. Using leukocytes from cancer patients, it was demonstrated that the monocyte-attracting activity targeted cells with an inflammatory phenotype characterised by high levels of HLA-DR expression. Conclusions Despite other classes of immune modulatory genes differently expressed in the alternative DC require to be investigated and characterised regarding their functional consequences, the reduced maturation state and chemoattractive properties of the gold standard versus alternative DC clearly promote the necessity to change the clinically used maturation cocktail of DC in order to improve the outcome of patients treated with DC-based vaccines.
Collapse
Affiliation(s)
- Chiara Massa
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| | - Carolin Thomas
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| | - Ena Wang
- Department of Transfusion Medicine, National Institute of Health Clinical Center, Bethesda, USA. .,Sidra Medical and Research Center, Doha, Qatar.
| | - Francesco Marincola
- Department of Transfusion Medicine, National Institute of Health Clinical Center, Bethesda, USA. .,Sidra Medical and Research Center, Doha, Qatar.
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
30
|
Anguille S, Van Acker HH, Van den Bergh J, Willemen Y, Goossens H, Van Tendeloo VF, Smits EL, Berneman ZN, Lion E. Interleukin-15 Dendritic Cells Harness NK Cell Cytotoxic Effector Function in a Contact- and IL-15-Dependent Manner. PLoS One 2015; 10:e0123340. [PMID: 25951230 PMCID: PMC4423923 DOI: 10.1371/journal.pone.0123340] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 03/02/2015] [Indexed: 01/02/2023] Open
Abstract
The contribution of natural killer (NK) cells to the treatment efficacy of dendritic cell (DC)-based cancer vaccines is being increasingly recognized. Much current efforts to optimize this form of immunotherapy are therefore geared towards harnessing the NK cell-stimulatory ability of DCs. In this study, we investigated whether generation of human monocyte-derived DCs with interleukin (IL)-15 followed by activation with a Toll-like receptor stimulus endows these DCs, commonly referred to as "IL-15 DCs", with the capacity to stimulate NK cells. In a head-to-head comparison with "IL-4 DCs" used routinely for clinical studies, IL-15 DCs were found to induce a more activated, cytotoxic effector phenotype in NK cells, in particular in the CD56bright NK cell subset. With the exception of GM-CSF, no significant enhancement of cytokine/chemokine secretion was observed following co-culture of NK cells with IL-15 DCs. IL-15 DCs, but not IL-4 DCs, promoted NK cell tumoricidal activity towards both NK-sensitive and NK-resistant targets. This effect was found to require cell-to-cell contact and to be mediated by DC surface-bound IL-15. This study shows that DCs can express a membrane-bound form of IL-15 through which they enhance NK cell cytotoxic function. The observed lack of membrane-bound IL-15 on "gold-standard" IL-4 DCs and their consequent inability to effectively promote NK cell cytotoxicity may have important implications for the future design of DC-based cancer vaccine studies.
Collapse
Affiliation(s)
- Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Heleen H. Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Johan Van den Bergh
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Viggo F. Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L. Smits
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
31
|
Vorobjova T, Uibo O, Heilman K, Uibo R. Increased density of tolerogenic dendritic cells in the small bowel mucosa of celiac patients. World J Gastroenterol 2015; 21:439-452. [PMID: 25593459 PMCID: PMC4292275 DOI: 10.3748/wjg.v21.i2.439] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/15/2014] [Accepted: 07/22/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the densities of dendritic cells (DCs) and FOXP3(+) regulatory T cells (Tregs) and their interrelations in the small bowel mucosa in untreated celiac disease (CD) patients with and without type 1 diabetes (T1D). METHODS Seventy-four patients (45 female, 29 male, mean age 11.1 ± 6.8 years) who underwent small bowel biopsy were studied. CD without T1D was diagnosed in 18 patients, and CD with T1D was diagnosed in 15 patients. Normal small bowel mucosa was found in two T1D patients. Thirty-nine patients (mean age 12.8 ± 4.9 years) with other diagnoses (functional dyspepsia, duodenal ulcer, erosive gastritis, etc.) formed the control group. All CD patients had partial or subtotal villous atrophy according to the Marsh classification: Marsh grade IIIa in 9, grade IIIb in 21 and grade IIIc in 3 cases. Thirty-nine patients without CD and 2 with T1D had normal small bowel mucosa (Marsh grade 0). The densities of CD11c(+), IDO(+), CD103(+), Langerin (CD207(+)) DCs and FOXP3(+) Tregs were investigated by immunohistochemistry (on paraffin-embedded specimens) and immunofluorescence (on cryostat sections) methods using a combination of mono- and double-staining. Sixty-six serum samples were tested for IgA-tissue transglutaminase (tTG) using a fully automated EliA™ Celikey(®) IgA assay (Pharmacia Diagnostics, Freiburg, Germany). RESULTS The density of CD11c(+) DCs was significantly increased in CD patients compared with patients with normal mucosa (21.67 ± 2.49 vs 13.58 ± 1.51, P = 0.007). The numbers of FOXP3(+) cells were significantly higher in CD patients (10.66 ± 1.50 vs 1.92 ± 0.37, P = 0.0002) and in patients with CD and coexisting T1D (8.11 ± 1.64 vs 1.92 ± 0.37, P = 0.002) compared with patients with normal mucosa. The density of FOXP3(+) cells significantly correlated with the histological grade of atrophic changes in the small bowel mucosa according to the March classification (r = 0.62; P < 0.0001) and with levels of IgA antibody (r = 0.55; P < 0.0001). The densities of IDO(+) DCs were significantly higher in CD patients (21.6 ± 2.67 vs 6.26 ± 0.84, P = 0.00003) and in patients with CD and coexisting T1D (19.08 ± 3.61 vs 6.26 ± 0.84, P = 0.004) compared with patients with normal mucosa. A significant correlation was identified between the densities of IDO(+) DCs and FOXP3(+) T cells (r = 0.76; P = 0.0001). The mean values of CD103(+) DCs were significantly higher in CD patients (10.66 ± 1.53 vs 6.34 ± 0.61, P = 0.01) and in patients with CD and associated T1D (11.13 ± 0.72 vs 6.34 ± 0.61, P = 0.00002) compared with subjects with normal small bowel mucosa. The mean value of Langerin(+) DCs was higher in CD patients compared with persons with normal mucosa (7.4 ± 0.92 vs 5.64 ± 0.46, P = 0.04). CONCLUSION The participation of diverse DC subsets in the pathological processes of CD and the possible involvement of tolerogenic DCs in Tregs development to maintain intestinal immunological tolerance in CD patients are revealed.
Collapse
|
32
|
Okada S, Han S, Patel ES, Yang LJ, Chang LJ. STAT3 signaling contributes to the high effector activities of interleukin-15-derived dendritic cells. Immunol Cell Biol 2015; 93:461-71. [PMID: 25582338 PMCID: PMC4450366 DOI: 10.1038/icb.2014.103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 11/04/2014] [Accepted: 11/13/2014] [Indexed: 02/05/2023]
Abstract
Dendritic cells (DCs) are important innate and adaptive immune effectors, and have a key role in antigen presentation and T-cell activation. Different lineages of DCs can be developed from hematopoietic progenitors following cytokine signaling, and the various lineages of DCs display distinct morphology, phenotype and functions. There has been limited information on differential cytokine-mediated molecular signaling in DCs. Analyses of surface molecules by flow cytometry and quantitative RNA profiling revealed differences between DCs derived from interleukin-4 (IL-4) versus IL-15 signaling, yet both lineages of DCs exhibited similar levels of surface molecules key to immune activation. Functional assays confirmed that IL-15-derived DCs elicited greater antigen-specific, primary and secondary CD8 and CD4 T-cell responses than did IL-4-derived DCs. Importantly, IL-15 DCs secreted substantial amounts of proinflammatory cytokines, including IL-6, interferon-γ (IFN-γ) and tumor necrosis factor-α (TNFα), which helped polarize a strong T-cell response. Assessment of signaling pathways revealed that IL-15 DCs exhibited a lower levels of activated signal transducer and activator of transcription 5 (STAT5), STAT6 and extracellular signal-regulated kinase 1/2 than IL-4 DCs, but after lipopolysaccharide (LPS)/TNFα treatment, the STAT3 and p38 mitogen-activated protein kinase (MAPK) activities were significantly enhanced in the IL-15 DCs. Surprisingly, contrary to the canonical IL-15-mediated STAT5 signaling pathway in lymphoid cells, IL-15 did not mediate a strong STAT5 or STAT3 activation in DCs. Further analysis using specific inhibitors to STAT3 and p38 MAPK pathways revealed that the STAT3 signaling, but not p38 MAPK signaling, contributed to IFN-γ production in DCs. Therefore, while IL-15 does not promote the STAT signaling in DCs, the increased STAT3 activity after LPS/TNFα treatment of the IL-15 DCs has a key role in their high IFN-γ effector activities.
Collapse
Affiliation(s)
- Starlyn Okada
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Shuhong Han
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ekta S Patel
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Li-Jun Yang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lung-Ji Chang
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Benteyn D, Heirman C, Bonehill A, Thielemans K, Breckpot K. mRNA-based dendritic cell vaccines. Expert Rev Vaccines 2014; 14:161-76. [PMID: 25196947 DOI: 10.1586/14760584.2014.957684] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer immunotherapy has been proposed as a powerful treatment modality. Active immunotherapy aspires to stimulate the patient's immune system, particularly T cells. These cells can recognize and kill cancer cells and can form an immunological memory. Dendritic cells (DCs) are the professional antigen-presenting cells of our immune system. They take up and process antigens to present them to T cells. Consequently, DCs have been investigated as a means to stimulate cancer-specific T-cell responses. An efficient strategy to program DCs is the use of mRNA, a well-defined and safe molecule that can be easily generated at high purity. Importantly, vaccines consisting of mRNA-modified DCs showed promising results in clinical trials. Therefore, we will introduce cancer immunotherapy and DCs and give a detailed overview on the application of mRNA to generate cancer-fighting DC vaccines.
Collapse
Affiliation(s)
- Daphné Benteyn
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Jette, Belgium
| | | | | | | | | |
Collapse
|
34
|
Van den Bergh JMJ, Guerti K, Willemen Y, Lion E, Cools N, Goossens H, Vorsters A, Van Tendeloo VFI, Anguille S, Van Damme P, Smits ELJM. HPV vaccine stimulates cytotoxic activity of killer dendritic cells and natural killer cells against HPV-positive tumour cells. J Cell Mol Med 2014; 18:1372-80. [PMID: 24979331 PMCID: PMC4124021 DOI: 10.1111/jcmm.12284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/17/2014] [Indexed: 01/09/2023] Open
Abstract
Cervarix™ is approved as a preventive vaccine against infection with the human papillomavirus (HPV) strains 16 and 18, which are causally related to the development of cervical cancer. We are the first to investigate in vitro the effects of this HPV vaccine on interleukin (IL)-15 dendritic cells (DC) as proxy of a naturally occurring subset of blood DC, and natural killer (NK) cells, two innate immune cell types that play an important role in antitumour immunity. Our results show that exposure of IL-15 DC to the HPV vaccine results in increased expression of phenotypic maturation markers, pro-inflammatory cytokine production and cytotoxic activity against HPV-positive tumour cells. These effects are mediated by the vaccine adjuvant, partly through Toll-like receptor 4 activation. Next, we demonstrate that vaccine-exposed IL-15 DC in turn induce phenotypic activation of NK cells, resulting in a synergistic cytotoxic action against HPV-infected tumour cells. Our study thus identifies a novel mode of action of the HPV vaccine in boosting innate immunity, including killing of HPV-infected cells by DC and NK cells.
Collapse
Affiliation(s)
- Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Terhune J, Berk E, Czerniecki BJ. Dendritic Cell-Induced Th1 and Th17 Cell Differentiation for Cancer Therapy. Vaccines (Basel) 2013; 1:527-49. [PMID: 26344346 PMCID: PMC4494209 DOI: 10.3390/vaccines1040527] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/18/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023] Open
Abstract
The success of cellular immunotherapies against cancer requires the generation of activated CD4+ and CD8+ T-cells. The type of T-cell response generated (e.g., Th1 or Th2) will determine the efficacy of the therapy, and it is generally assumed that a type-1 response is needed for optimal cancer treatment. IL-17 producing T-cells (Th17/Tc17) play an important role in autoimmune diseases, but their function in cancer is more controversial. While some studies have shown a pro-cancerous role for IL-17, other studies have shown an anti-tumor function. The induction of polarized T-cell responses can be regulated by dendritic cells (DCs). DCs are key regulators of the immune system with the ability to affect both innate and adaptive immune responses. These properties have led many researchers to study the use of ex vivo manipulated DCs for the treatment of various diseases, such as cancer and autoimmune diseases. While Th1/Tc1 cells are traditionally used for their potent anti-tumor responses, mounting evidence suggests Th17/Tc17 cells should be utilized by themselves or for the induction of optimal Th1 responses. It is therefore important to understand the factors involved in the induction of both type-1 and type-17 T-cell responses by DCs.
Collapse
Affiliation(s)
- Julia Terhune
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Erik Berk
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Brian J Czerniecki
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Rena Rowan Breast Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Gehring AJ, Haniffa M, Kennedy PT, Ho ZZ, Boni C, Shin A, Banu N, Chia A, Lim SG, Ferrari C, Ginhoux F, Bertoletti A. Mobilizing monocytes to cross-present circulating viral antigen in chronic infection. J Clin Invest 2013; 123:3766-76. [PMID: 23908113 DOI: 10.1172/jci66043] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 06/06/2013] [Indexed: 12/16/2022] Open
Abstract
Selection of antigens for therapeutic vaccination against chronic viral infections is complicated by pathogen genetic variations. We tested whether antigens present during persistent viral infections could provide a personalized antigenic reservoir for therapeutic T cell expansion in humans. We focused our study on the HBV surface antigen (HBsAg), which is present in microgram quantities in the serum of chronic HBV patients. We demonstrated by quantitative fluorescent microscopy that, out of 6 professional APC populations in the circulation, only CD14 monocytes (MNs) retained an HBsAg depot. Using TCR-redirected CD8+ T cells specific for MHC-I-restricted HBV epitopes, we showed that, despite being constantly exposed to antigen, ex vivo-isolated APCs did not constitutively activate HBV-specific CD8+ T cells. However, differentiation of HBsAg+ CD14 MNs from chronic patients to MN-derived DCs (moDCs) induced cross-presentation of the intracellular reservoir of viral antigen. We exploited this mechanism to cross-present circulating viral antigen and showed that moDCs from chronically infected patients stimulated expansion of autologous HBV-specific T cells. Thus, these data demonstrate that circulating viral antigen produced during chronic infection can serve as a personalized antigenic reservoir to activate virus-specific T cells.
Collapse
Affiliation(s)
- Adam J Gehring
- Infection and Immunity Programme, Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Anguille S, Lion E, Van den Bergh J, Van Acker HH, Willemen Y, Smits EL, Van Tendeloo VF, Berneman ZN. Interleukin-15 dendritic cells as vaccine candidates for cancer immunotherapy. Hum Vaccin Immunother 2013; 9:1956-61. [PMID: 23778748 DOI: 10.4161/hv.25373] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Owing to their professional antigen-presenting capacity and unique potential to induce tumor antigen-specific T cell immunity, dendritic cells (DCs) have attracted much interest over the past decades for therapeutic vaccination against cancer. Clinical trials have shown that the use of tumor antigen-loaded DCs in cancer patients is safe and that it has the potential to induce anti-tumor immunity which, in some cases, culminates in striking clinical responses. Unfortunately, in a considerable number of patients, DC vaccination is unable to mount effective anti-tumor immune responses and, if it does so, the resultant immunity is often insufficient to translate into tangible clinical benefit. This underscores the necessity to re-design and optimize the current procedures for DC vaccine manufacturing. A new generation of DC vaccines with improved potency has now become available for clinical use as a result of extensive pre-clinical research. One of the promising next-generation DC vaccine candidates are interleukin (IL)-15-differentiated DCs. In this commentary, we will compile the research data that have been obtained by our group and other groups with these so-called IL-15 DCs and summarize the evidence supporting the implementation of IL-15 DCs in DC-based cancer vaccination regimens.
Collapse
Affiliation(s)
- Sébastien Anguille
- Vaccine & Infectious Disease Institute; Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); University of Antwerp; Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine; Antwerp University Hospital; Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Hubo M, Trinschek B, Kryczanowsky F, Tuettenberg A, Steinbrink K, Jonuleit H. Costimulatory molecules on immunogenic versus tolerogenic human dendritic cells. Front Immunol 2013; 4:82. [PMID: 23565116 PMCID: PMC3615188 DOI: 10.3389/fimmu.2013.00082] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/20/2013] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) are sentinels of immunity, essential for homeostasis of T cell-dependent immune responses. Both functions of DC, initiation of antigen-specific T cell immunity and maintenance of tissue-specific tolerance originate from distinct stages of differentiation, immunogenic versus tolerogenic. Dependent on local micro milieu and inflammatory stimuli, tissue resident immature DC with functional plasticity differentiate into tolerogenic or immunogenic DC with stable phenotypes. They efficiently link innate and adaptive immunity and are ideally positioned to modify T cell-mediated immune responses. Since the T cell stimulatory properties of DC are significantly influenced by their expression of signal II ligands, it is critical to understand the impact of distinct costimulatory pathways on DC function. This review gives an overview of functional different human DC subsets with unique profiles of costimulatory molecules and outlines how different costimulatory pathways together with the immunosuppressive cytokine IL-10 bias immunogenic versus tolerogenic DC functions. Furthermore, we exemplarily describe protocols for the generation of two well-defined monocyte-derived DC subsets for their clinical use, immunogenic versus tolerogenic.
Collapse
Affiliation(s)
- Mario Hubo
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz Mainz, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Understanding dendritic cells and their role in cutaneous carcinoma and cancer immunotherapy. Clin Dev Immunol 2013; 2013:624123. [PMID: 23606870 PMCID: PMC3625554 DOI: 10.1155/2013/624123] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/07/2013] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DC) represent a diverse group of professional antigen-presenting cells that serve to link the innate and adaptive immune systems. Their capacity to initiate a robust and antigen-specific immune response has made them the ideal candidates for cancer immunotherapies. To date, the clinical impact of DC immunotherapy has been limited, which may, in part, be explained by the complex nature of DC biology. Multiple distinct subsets of DCs have been identified in the skin, where they can be broadly subcategorized into epidermal Langerhans cells (LC), myeloid-derived dermal dendritic cells (mDC) and plasmacytoid dendritic cells (pDC). Each subset is functionally unique and may activate alternate branches of the immune system. This may be relevant for the treatment of squamous cell carcinoma, where we have shown that the tumor microenvironment may preferentially suppress the activity of mDCs, while LCs remain potent stimulators of immunity. Here, we provide an in depth analysis of DC biology, with a particular focus on skin DCs and their role in cutaneous carcinoma. We further explore the current approaches to DC immunotherapy and provide evidence for the targeting of LCs as a promising new strategy in the treatment of skin cancer.
Collapse
|
40
|
Anguille S, Lion E, Tel J, de Vries IJM, Couderé K, Fromm PD, Van Tendeloo VF, Smits EL, Berneman ZN. Interleukin-15-induced CD56(+) myeloid dendritic cells combine potent tumor antigen presentation with direct tumoricidal potential. PLoS One 2012; 7:e51851. [PMID: 23284789 PMCID: PMC3532168 DOI: 10.1371/journal.pone.0051851] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/07/2012] [Indexed: 11/18/2022] Open
Abstract
Dendritic cells (DCs) are the quintessential antigen-presenting cells of the human immune system and play a prime role in coordinating innate and adaptive immune responses, explaining the strong and still growing interest in their application for cancer immunotherapy. Much current research in the field of DC-based immunotherapy focuses on optimizing the culture conditions for in vitro DC generation in order to assure that DCs with the best possible immunogenic qualities are being used for immunotherapy. In this context, monocyte-derived DCs that are alternatively induced by interleukin-15 (IL-15 DCs) have attracted recent attention due to their superior immunostimulatory characteristics. In this study, we show that IL-15 DCs, in addition to potent tumor antigen-presenting function, possess tumoricidal potential and thus qualify for the designation of killer DCs. Notwithstanding marked expression of the natural killer (NK) cell marker CD56 on a subset of IL-15 DCs, we found no evidence of a further phenotypic overlap between IL-15 DCs and NK cells. Allostimulation and antigen presentation assays confirmed that IL-15 DCs should be regarded as bona fide myeloid DCs not only from the phenotypic but also from the functional point of view. Concerning their cytotoxic activity, we demonstrate that IL-15 DCs are able to induce apoptotic cell death of the human K562 tumor cell line, while sparing tumor antigen-specific T cells. The cytotoxicity of IL-15 DCs is predominantly mediated by granzyme B and, to a small extent, by tumor necrosis factor-α (TNF-α)-related apoptosis-inducing ligand (TRAIL) but is independent of perforin, Fas ligand and TNF-α. In conclusion, our data provide evidence of a previously unappreciated role for IL-15 in the differentiation of human monocytes towards killer DCs. The observation that IL-15 DCs have killer DC capacity lends further support to their implementation in DC-based immunotherapy protocols.
Collapse
Affiliation(s)
- Sébastien Anguille
- University of Antwerp, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), Laboratory of Experimental Hematology, Antwerp, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hopkins RA, Connolly JE. The specialized roles of immature and mature dendritic cells in antigen cross-presentation. Immunol Res 2012; 53:91-107. [PMID: 22450675 DOI: 10.1007/s12026-012-8300-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Exogenous antigen cross-presentation is integral to the stimulation of cytotoxic T-lymphocytes against viruses and tumors. Central to this process are dendritic cells (DCs), which specialize in cross-presentation. DCs may be considered to exist in two radically different states of activation, generally referred to as immature and mature. In each of these states, the cell has a series of separate and specialized abilities for the induction of T-cell immunity. In the immature state, the DC is adept in surveying the periphery, acquiring and storing antigen, but has a limited capacity for direct T-cell activation. During a brief and defined window of time following DC stimulation, nearly every aspect of antigen handling changes, as it transitions from an entity focused on protein preservation to one capable of efficient cross-presentation. It is this time period and the underlying molecular mechanisms active here, which form the core of our studies on cross-presentation.
Collapse
Affiliation(s)
- Richard A Hopkins
- Program in Translational Immunology, Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03 Immunos, Biopolis, Singapore
| | | |
Collapse
|
42
|
Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm 2012; 2012:690643. [PMID: 22851815 PMCID: PMC3407661 DOI: 10.1155/2012/690643] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/17/2012] [Indexed: 02/08/2023] Open
Abstract
Earlier investigations have revealed a surprising complexity and variety in the range of interaction between cells of the innate and adaptive immune system. Our understanding of the specialized roles of dendritic cell (DC) subsets in innate and adaptive immune responses has been significantly advanced over the years. Because of their immunoregulatory capacities and because very small numbers of activated DC are highly efficient at generating immune responses against antigens, DCs have been vigorously used in clinical trials in order to elicit or amplify immune responses against cancer and chronic infectious diseases. A better insight in DC immunobiology and function has stimulated many new ideas regarding the potential ways forward to improve DC therapy in a more fundamental way. Here, we discuss the continuous search for optimal in vitro conditions in order to generate clinical-grade DC with a potent immunogenic potential. For this, we explore the molecular and cellular mechanisms underlying adequate immune responses and focus on most favourable DC culture regimens and activation stimuli in humans. We envisage that by combining each of the features outlined in the current paper into a unified strategy, DC-based vaccines may advance to a higher level of effectiveness.
Collapse
|
43
|
Castillo EF, Schluns KS. Regulating the immune system via IL-15 transpresentation. Cytokine 2012; 59:479-90. [PMID: 22795955 DOI: 10.1016/j.cyto.2012.06.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 06/06/2012] [Accepted: 06/09/2012] [Indexed: 02/07/2023]
Abstract
Transpresentation has emerged as an important mechanism mediating IL-15 responses in a subset of lymphocytes during the steady state. In transpresentation, cell surface IL-15, bound to IL-15Rα is delivered to opposing lymphocytes during a cell-cell interaction. The events most dependent on IL-15 include the development and homeostasis of memory CD8 T cells, Natural Killer cells, invariant Natural Killer T cells, and intraepithelial lymphocytes. As lymphocyte development and homeostasis involve multiple steps and mechanisms, IL-15 transpresentation can have diverse roles throughout. Moreover, distinct stages of lymphocyte differentiation require IL-15 transpresented by different cells, which include both hematopoietic and non-hematopoietic cell types. Herein, we will describe the points where IL-15 transpresentation impacts these processes, the specific cells thought to drive IL-15 responses, as well as their role in the course of development and homeostasis.
Collapse
Affiliation(s)
- Eliseo F Castillo
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
44
|
The differential production of cytokines by human Langerhans cells and dermal CD14(+) DCs controls CTL priming. Blood 2012; 119:5742-9. [PMID: 22535664 DOI: 10.1182/blood-2011-08-371245] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We recently reported that human epidermal Langerhans cells (LCs) are more efficient than dermal CD14(+) DCs at priming naive CD8(+) T cells into potent CTLs. We hypothesized that distinctive dendritic cell (DC) cytokine expression profiles (ie, IL-15 produced by LCs and IL-10 expressed by dermal CD14(+) DCs) might explain the observed functional difference. Blocking IL-15 during CD8(+) T-cell priming reduced T-cell proliferation by ∼ 50%. These IL-15-deprived CD8(+) T cells did not acquire the phenotype of effector memory cells. They secreted less IL-2 and IFN-γ and expressed only low amounts of CD107a, granzymes and perforin, and reduced levels of the antiapoptotic protein Bcl-2. Confocal microscopy analysis showed that IL-15 is localized at the immunologic synapse of LCs and naive CD8(+) T cells. Conversely, blocking IL-10 during cocultures of dermal CD14(+) DCs and naive CD8(+) T cells enhanced the generation of effector CTLs, whereas addition of IL-10 to cultures of LCs and naive CD8(+) T cells inhibited their induction. TGF-β1 that is transcribed by dermal CD14(+) DCs further enhanced the inhibitory effect of IL-10. Thus, the respective production of IL-15 and IL-10 explains the contrasting effects of LCs and dermal CD14(+) DCs on CD8(+) T-cell priming.
Collapse
|
45
|
Abstract
Immunotherapy with dendritic cells (DCs), which have been manipulated ex vivo to become immunogenic or tolerogenic, has been tested in clinical trials for disease therapy. DCs are sentinels of the immune system, which after exposure to antigenic or inflammatory signals and crosstalk with effector CD4(+) T cells express high levels of costimulatory molecules and cytokines. Upregulation of either costimulatory molecules or cytokines promotes immunologic DCs, whereas their downregulation generates tolerogenic DCs (TDCs), which induce T regulatory cells (Tregs) and a state of tolerance. Immunogenic DCs are used for the therapy of infectious diseases such as HIV-1 and cancer, whereas tolerogenic DCs are used in treating various autoimmune diseases and in transplantation. DC vaccination is still at an early stage, and improvements are mainly needed in quality control of monitoring assays to generate clinical-grade DC products and to assess the effect of DC vaccination in future clinical trials. Here, we review the recent work in DC generation and monitoring approaches for DC-based trials with immunogenic or tolerogenic DCs.
Collapse
|
46
|
Human Langerhans cells use an IL-15R-α/IL-15/pSTAT5-dependent mechanism to break T-cell tolerance against the self-differentiation tumor antigen WT1. Blood 2012; 119:5182-90. [PMID: 22510877 DOI: 10.1182/blood-2011-09-382200] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human CD34(+) progenitor-derived Langerhans-type dendritic cells (LCs) are more potent stimulators of T-cell immunity against tumor and viral antigens in vitro than are monocyte-derived DCs (moDCs). The exact mechanisms have remained elusive until now, however. LCs synthesize the highest amounts of IL-15R-α mRNA and protein, which binds IL-15 for presentation to responder lymphocytes, thereby signaling the phosphorylation of signal transducer and activator of transcription 5 (pSTAT5). LCs electroporated with Wilms tumor 1 (WT1) mRNA achieve sufficiently sustained presentation of antigenic peptides, which together with IL-15R-α/IL-15, break tolerance against WT1 by stimulating robust autologous, WT1-specific cytolytic T-lymphocytes (CTLs). These CTLs develop from healthy persons after only 7 days' stimulation without exogenous cytokines and lyse MHC-restricted tumor targets, which include primary WT1(+) leukemic blasts. In contrast, moDCs require exogenous rhuIL-15 to phosphorylate STAT5 and attain stimulatory capacity comparable to LCs. LCs therefore provide a more potent costimulatory cytokine milieu for T-cell activation than do moDCs, thus accounting for their superior stimulation of MHC-restricted Ag-specific CTLs without need for exogenous cytokines. These data support the use of mRNA-electroporated LCs, or moDCs supplemented with exogenous rhuIL-15, as vaccines for cancer immunotherapy to break tolerance against self-differentiation antigens shared by tumors.
Collapse
|
47
|
Abstract
Cancer immunotherapy attempts to harness the power and specificity of the immune system to treat tumours. The molecular identification of human cancer-specific antigens has allowed the development of antigen-specific immunotherapy. In one approach, autologous antigen-specific T cells are expanded ex vivo and then re-infused into patients. Another approach is through vaccination; that is, the provision of an antigen together with an adjuvant to elicit therapeutic T cells in vivo. Owing to their properties, dendritic cells (DCs) are often called 'nature's adjuvants' and thus have become the natural agents for antigen delivery. After four decades of research, it is now clear that DCs are at the centre of the immune system owing to their ability to control both immune tolerance and immunity. Thus, DCs are an essential target in efforts to generate therapeutic immunity against cancer.
Collapse
Affiliation(s)
- Karolina Palucka
- Baylor Institute for Immunology Research, 3434 Live Oak Avenue, Dallas, Texas 75204, USA.
| | | |
Collapse
|
48
|
Induction of intestinal pro-inflammatory immune responses by lipoteichoic acid. JOURNAL OF INFLAMMATION-LONDON 2012; 9:7. [PMID: 22423982 PMCID: PMC3325164 DOI: 10.1186/1476-9255-9-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/16/2012] [Indexed: 02/02/2023]
Abstract
Background The cellular and molecular mechanisms of inflammatory bowel disease are not fully understood; however, data indicate that uncontrolled chronic inflammation induced by bacterial gene products, including lipoteichoic acid (LTA), may trigger colonic inflammation resulting in disease pathogenesis. LTA is a constituent glycolipid of Gram-positive bacteria that shares many inflammatory properties with lipopolysaccharide and plays a critical role in the pathogenesis of severe inflammatory responses via Toll-like receptor 2. Accordingly, we elucidate the role of LTA in immune stimulation and induced colitis in vivo. Methods To better understand the molecular mechanisms utilized by the intestinal microbiota and their gene products to induce or subvert inflammation, specifically the effect(s) of altered surface layer protein expression on the LTA-mediated pro-inflammatory response, the Lactobacillus acidophilus surface layer protein (Slp) genes encoding SlpB and SlpX were deleted resulting in a SlpB- and SlpX- mutant that continued to express SlpA (assigned as NCK2031). Results Our data show profound activation of dendritic cells by NCK2031, wild-type L. acidophilus (NCK56), and purified Staphylococcus aureus-LTA. In contrary to the LTA-deficient strain NCK2025, the LTA-expressing strains NCK2031 and NCK56, as well as S. aureus-LTA, induce pro-inflammatory innate and T cell immune responses in vivo. Additionally, neither NCK2031 nor S. aureus-LTA supplemented in drinking water protected mice from DSS-colitis, but instead, induced significant intestinal inflammation resulting in severe colitis and tissue destruction. Conclusions These findings suggest that directed alteration of two of the L. acidophilus NCFM-Slps did not ameliorate LTA-induced pro-inflammatory signals and subsequent colitis.
Collapse
|
49
|
Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst 2012; 104:599-613. [PMID: 22395641 DOI: 10.1093/jnci/djs033] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Concurrent with U.S. Food and Drug Administration (FDA) approval of the first therapeutic cancer vaccine, a wide spectrum of other cancer vaccine platforms that target a diverse range of tumor-associated antigens is currently being evaluated in randomized phase II and phase III trials. The profound influence of the tumor microenvironment and other immunosuppressive entities, however, can limit the effectiveness of these vaccines. Numerous strategies are currently being evaluated both preclinically and clinically to counteract these immunosuppressive entities, including the combined use of vaccines with immune checkpoint inhibitors, certain chemotherapeutics, small-molecule targeted therapies, and radiation. The potential influence of the appropriate patient population and clinical trial endpoint in vaccine therapy studies is discussed, as well as the potential importance of biomarkers in future directions of this field.
Collapse
Affiliation(s)
- Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr, Rm 8B09, Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Leukotriene C4 induces migration of human monocyte-derived dendritic cells without loss of immunostimulatory function. Blood 2012; 119:3113-22. [PMID: 22323449 DOI: 10.1182/blood-2011-10-385930] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Generation of human monocyte-derived dendritic cells (DCs) for cancer vaccination involves ex vivo maturation in the presence of proinflammatory cytokines and prostaglandin E(2) (PGE(2)). Although the inclusion of PGE(2) during maturation is imperative for the induction of DC migration, PGE(2) has unfavorable effects on the immunostimulatory capacity of these cells. Like PGE(2), leukotrienes (LTs) are potent mediators of DC migration. We therefore sought to characterize the migratory and immunologic properties of DCs that matured in the presence of LTB(4), LTC(4), LTD(4), and PGE(2). Here, we demonstrate that DCs matured in the presence of LTC(4), but not LTB(4) or LTD(4), are superior to PGE(2)-matured DCs in stimulating CD4(+) T-cell responses and in inducing antigen-specific cytotoxic T lymphocytes (CTLs) in vitro without concomitant induction or recruitment of regulatory T cells (Tregs). LTC(4)-matured DCs migrate efficiently through layers of extracellular matrix and secrete higher levels of immunostimulatory IL-12p70 while producing reduced levels of immune-inhibitory IL-10, IL12p40, indoleamine-2,3-dioxidase, and TIMP-1 (tissue inhibitor of matrix metalloproteinases). Intracellular calcium mobilization and receptor antagonist studies reveal that, in contrast to LTD(4), LTC(4) did not signal through CysLTR(1) in DCs. Collectively, our data suggest that LTC(4) represents a promising candidate to replace PGE(2) in DC maturation protocols for cancer vaccination.
Collapse
|