1
|
Lv D, Jiang H, Yang X, Li Y, Niu W, Zhang D. Advances in understanding of dendritic cell in the pathogenesis of acute kidney injury. Front Immunol 2024; 15:1294807. [PMID: 38433836 PMCID: PMC10904453 DOI: 10.3389/fimmu.2024.1294807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Acute kidney injury (AKI) is characterized by a rapid decline in renal function and is associated with a high morbidity and mortality rate. At present, the underlying mechanisms of AKI remain incompletely understood. Immune disorder is a prominent feature of AKI, and dendritic cells (DCs) play a pivotal role in orchestrating both innate and adaptive immune responses, including the induction of protective proinflammatory and tolerogenic immune reactions. Emerging evidence suggests that DCs play a critical role in the initiation and development of AKI. This paper aimed to conduct a comprehensive review and analysis of the role of DCs in the progression of AKI and elucidate the underlying molecular mechanism. The ultimate objective was to offer valuable insights and guidance for the treatment of AKI.
Collapse
Affiliation(s)
- Dongfang Lv
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huihui Jiang
- Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianzhen Yang
- Department of Urology, Afliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Li
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Engineering Laboratory of Urinary Organ and Functional Reconstruction of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Weipin Niu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Key Laboratory of Dominant Diseases of traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Denglu Zhang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Key Laboratory of Dominant Diseases of traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
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
|
3
|
Hasan A, Al-Ozairi E, Hassan NYM, Ali S, Ahmad R, Al-Shatti N, Alshemmari S, Al-Mulla F. Fatal COVID-19 is Associated with Reduced HLA-DR, CD123 or CD11c Expression on Circulating Dendritic Cells. J Inflamm Res 2022; 15:5665-5675. [PMID: 36238761 PMCID: PMC9553279 DOI: 10.2147/jir.s360207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/11/2022] [Indexed: 11/06/2022] Open
Abstract
Purpose Severe coronavirus disease 2019 (COVID-19) is linked to insufficient control of viral replication and excessive inflammation driven by an unbalanced immune response. Plasmacytoid dendritic cells (pDCs) are specialized in the rapid production of interferons in response to viral infections, and can also prime and activate T-cells. Conventional DCs (cDCs) are critical for the elimination of viral infections owing to their specialized ability to prime and activate T cells. We assessed the frequency and phenotype of pDCs and cDCs in survivors and non-survivors of COVID-19. Patients and methods Patients with COVID-19 were enrolled, and 22 were included in this study. Peripheral whole blood was obtained during the 2nd week of illness, stained with antibodies specific for lineage markers, human leukocyte antigen-DR isotype (HLA-DR), CD11c, and CD123, and analyzed by flow cytometry. Patients were followed-up during hospital admission and grouped into survivors (n=17) and non-survivors (n=5) of COVID-19. Results The ratio of pDCs to pre-cDCs was significantly lower (P=0.0005) in non-survivors compared to survivors. The frequency of pDCs was significantly higher than cDC2-like cells (P=0.0002) and pre-cDCs (P<0.0001) in survivors but not in non-survivors. HLA-DR expression level on pDCs and cDC2-like cells was lower in non-survivors compared to survivors (P=0.02 and P=0.058, respectively), and HLA-DR was inversely correlated with disease severity rating (pDCs: r= –0.47, P=0.027; cDC2-like cells: r= –0.45, P=0.037). CD123 expression level on pDCs was significantly lower (P=0.038) in non-survivors compared to survivors, and CD123 was inversely correlated with disease severity rating (r=–0.5, P=0.016). CD11c expression level on cDC2-like cells was significantly lower (P=0.03) in non-survivors compared to survivors, and CD11c was inversely correlated with disease severity rating (r=–0.47, P=0.025). Conclusion A lower frequency of pDCs compared to other circulating DCs, and lower expression levels of HLA-DR, CD123 or CD11c on DCs is associated with fatal COVID-19.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait,Correspondence: Amal Hasan, Department of Immunology and Microbiology; Dasman Diabetes Institute, Dasman, Kuwait, Tel +965 2224 2999 Ext. 4312, Fax +965 2249 2406, Email
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait,Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Nosiba Y M Hassan
- Department of Internal Medicine, Jaber Al-Ahmad Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Shamsha Ali
- Special Services Facility, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Nada Al-Shatti
- Immunology & HLA Laboratory, Kuwait Cancer Control Center, Ministry of Health, Kuwait City, Kuwait
| | - Salem Alshemmari
- Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait,Department of Hematology, Kuwait Cancer Control Center, Ministry of Health, Kuwait City, Kuwit
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| |
Collapse
|
4
|
Wu R, Murphy KM. DCs at the center of help: Origins and evolution of the three-cell-type hypothesis. J Exp Med 2022; 219:e20211519. [PMID: 35543702 PMCID: PMC9098650 DOI: 10.1084/jem.20211519] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/06/2022] Open
Abstract
Last year was the 10th anniversary of Ralph Steinman's Nobel Prize awarded for his discovery of dendritic cells (DCs), while next year brings the 50th anniversary of that discovery. Current models of anti-viral and anti-tumor immunity rest solidly on Steinman's discovery of DCs, but also rely on two seemingly unrelated phenomena, also reported in the mid-1970s: the discoveries of "help" for cytolytic T cell responses by Cantor and Boyse in 1974 and "cross-priming" by Bevan in 1976. Decades of subsequent work, controversy, and conceptual changes have gradually merged these three discoveries into current models of cell-mediated immunity against viruses and tumors.
Collapse
Affiliation(s)
- Renee Wu
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
5
|
Doan TA, Forward T, Tamburini BAJ. Trafficking and retention of protein antigens across systems and immune cell types. Cell Mol Life Sci 2022; 79:275. [PMID: 35505125 PMCID: PMC9063628 DOI: 10.1007/s00018-022-04303-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/05/2022]
Abstract
In response to infection or vaccination, the immune system initially responds non-specifically to the foreign insult (innate) and then develops a specific response to the foreign antigen (adaptive). The programming of the immune response is shaped by the dispersal and delivery of antigens. The antigen size, innate immune activation and location of the insult all determine how antigens are handled. In this review we outline which specific cell types are required for antigen trafficking, which processes require active compared to passive transport, the ability of specific cell types to retain antigens and the viruses (human immunodeficiency virus, influenza and Sendai virus, vesicular stomatitis virus, vaccinia virus) and pattern recognition receptor activation that can initiate antigen retention. Both where the protein antigen is localized and how long it remains are critically important in shaping protective immune responses. Therefore, understanding antigen trafficking and retention is necessary to understand the type and magnitude of the immune response and essential for the development of novel vaccine and therapeutic targets.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA.,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA. .,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA. .,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
6
|
Dalod M, Scheu S. Dendritic cell functions in vivo: a user's guide to current and next generation mutant mouse models. Eur J Immunol 2022; 52:1712-1749. [PMID: 35099816 DOI: 10.1002/eji.202149513] [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: 11/19/2021] [Accepted: 01/14/2022] [Indexed: 11/11/2022]
Abstract
Dendritic cells (DCs) do not just excel in antigen presentation. They orchestrate information transfer from innate to adaptive immunity, by sensing and integrating a variety of danger signals, and translating them to naïve T cells, to mount specifically tailored immune responses. This is accomplished by distinct DC types specialized in different functions and because each DC is functionally plastic, assuming different activation states depending on the input signals received. Mouse models hold the key to untangle this complexity and determine which DC types and activation states contribute to which functions. Here, we aim to provide comprehensive information for selecting the most appropriate mutant mouse strains to address specific research questions on DCs, considering three in vivo experimental approaches: (i) interrogating the roles of DC types through their depletion; (ii) determining the underlying mechanisms by specific genetic manipulations; (iii) deciphering the spatiotemporal dynamics of DC responses. We summarize the advantages, caveats, suggested use and perspectives for a variety of mutant mouse strains, discussing in more detail the most widely used or accurate models. Finally, we discuss innovative strategies to improve targeting specificity, for the next generation mutant mouse models, and briefly address how humanized mouse models can accelerate translation into the clinic. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marc Dalod
- CNRS, Inserm, Aix Marseille Univ, Centre d'Immunologie de Marseille-Luminy (CIML), Turing Center for Living Systems, Marseille, France
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
7
|
Bourque J, Hawiger D. Applications of Antibody-Based Antigen Delivery Targeted to Dendritic Cells In Vivo. Antibodies (Basel) 2022; 11:antib11010008. [PMID: 35225867 PMCID: PMC8884005 DOI: 10.3390/antib11010008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Recombinant immunoglobulins, derived from monoclonal antibodies recognizing the defined surface epitopes expressed on dendritic cells, have been employed for the past two decades to deliver antigens to dendritic cells in vivo, serving as critical tools for the investigation of the corresponding T cell responses. These approaches originated with the development of the recombinant chimeric antibody against a multilectin receptor, DEC-205, which is present on subsets of murine and human conventional dendritic cells. Following the widespread application of antigen targeting through DEC-205, similar approaches then utilized other epitopes as entry points for antigens delivered by specific antibodies to multiple types of dendritic cells. Overall, these antigen-delivery methodologies helped to reveal the mechanisms underlying tolerogenic and immunogenic T cell responses orchestrated by dendritic cells. Here, we discuss the relevant experimental strategies as well as their future perspectives, including their translational relevance.
Collapse
Affiliation(s)
| | - Daniel Hawiger
- Correspondence: ; Tel.: +1-314-977-8875; Fax: +1-314-977-8717
| |
Collapse
|
8
|
Fu C, Zhou L, Mi QS, Jiang A. Plasmacytoid Dendritic Cells and Cancer Immunotherapy. Cells 2022; 11:222. [PMID: 35053338 PMCID: PMC8773673 DOI: 10.3390/cells11020222] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/02/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite largely disappointing clinical trials of dendritic cell (DC)-based vaccines, recent studies have shown that DC-mediated cross-priming plays a critical role in generating anti-tumor CD8 T cell immunity and regulating anti-tumor efficacy of immunotherapies. These new findings thus support further development and refinement of DC-based vaccines as mono-immunotherapy or combinational immunotherapies. One exciting development is recent clinical studies with naturally circulating DCs including plasmacytoid DCs (pDCs). pDC vaccines were particularly intriguing, as pDCs are generally presumed to play a negative role in regulating T cell responses in tumors. Similarly, DC-derived exosomes (DCexos) have been heralded as cell-free therapeutic cancer vaccines that are potentially superior to DC vaccines in overcoming tumor-mediated immunosuppression, although DCexo clinical trials have not led to expected clinical outcomes. Using a pDC-targeted vaccine model, we have recently reported that pDCs required type 1 conventional DCs (cDC1s) for optimal cross-priming by transferring antigens through pDC-derived exosomes (pDCexos), which also cross-prime CD8 T cells in a bystander cDC-dependent manner. Thus, pDCexos could combine the advantages of both cDC1s and pDCs as cancer vaccines to achieve better anti-tumor efficacy. In this review, we will focus on the pDC-based cancer vaccines and discuss potential clinical application of pDCexos in cancer immunotherapy.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| |
Collapse
|
9
|
Macri C, Morgan H, Villadangos JA, Mintern JD. Regulation of dendritic cell function by Fc-γ-receptors and the neonatal Fc receptor. Mol Immunol 2021; 139:193-201. [PMID: 34560415 DOI: 10.1016/j.molimm.2021.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023]
Abstract
Dendritic cells (DCs) express receptors to sense pathogens and/or tissue damage and to communicate with other immune cells. Among those receptors, Fc receptors (FcRs) are triggered by the Fc region of antibodies produced during adaptive immunity. In this review, the role of FcγR and neonatal Fc receptor (FcRn) in DC immunity will be discussed. Their expression in DC subsets and impact on antigen uptake and presentation, DC maturation and polarisation of T cell responses will be described. Lastly, we will discuss the importance of FcR-mediated DC function in the context of immunity during viral infection, inflammatory disease, cancer and immunotherapy.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Huw Morgan
- ACRF Translational Research Laboratory, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, 3050, Australia; Department of Medicine, University of Melbourne, Parkville, Melbourne, Victoria, 3010, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
10
|
Luu T, Cheung JF, Baccon J, Waldner H. Priming of myelin-specific T cells in the absence of dendritic cells results in accelerated development of Experimental Autoimmune Encephalomyelitis. PLoS One 2021; 16:e0250340. [PMID: 33891644 PMCID: PMC8064509 DOI: 10.1371/journal.pone.0250340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an established animal model of multiple sclerosis (MS). Inflammatory CD4+ T cell responses directed against CNS antigens, including myelin proteolipid protein (PLP), are key mediators of EAE. Dendritic cells (DCs) are critical for the induction of T cell responses against infectious agents. However, the importance of DCs in priming self-reactive CD4+ T cells in autoimmune disease such as MS has been unclear. To determine the requirement of DCs in PLP-specific CD4+ T cell responses and EAE, we genetically deleted CD11c+ DCs in PLP T cell receptor (TCR) transgenic SJL mice constitutively. DC deficiency did not impair the development, selection or the pathogenic function of PLP-specific CD4+ T cells in these mice, and resulted in accelerated spontaneous EAE compared to DC sufficient controls. In addition, using a genetic approach to ablate DCs conditionally in SJL mice, we show that CD11c+ DCs were dispensable for presenting exogenous or endogenous myelin antigen to PLP-specific T cells and for promoting pro-inflammatory T cell responses and severe EAE. Our findings demonstrate that constitutive or conditional ablation of CD11c+ DCs diminished self-tolerance to PLP autoantigen. They further show that in the absence of DCs, non-DCs can efficiently present CNS myelin antigens such as PLP to self-reactive T cells, resulting in accelerated onset of spontaneous or induced EAE.
Collapse
Affiliation(s)
- Thaiphi Luu
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Julie F. Cheung
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Jennifer Baccon
- Department of Pathology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Hanspeter Waldner
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
11
|
Fu C, Zhou L, Mi QS, Jiang A. DC-Based Vaccines for Cancer Immunotherapy. Vaccines (Basel) 2020; 8:vaccines8040706. [PMID: 33255895 PMCID: PMC7712957 DOI: 10.3390/vaccines8040706] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
As the sentinels of the immune system, dendritic cells (DCs) play a critical role in initiating and regulating antigen-specific immune responses. Cross-priming, a process that DCs activate CD8 T cells by cross-presenting exogenous antigens onto their MHCI (Major Histocompatibility Complex class I), plays a critical role in mediating CD8 T cell immunity as well as tolerance. Current DC vaccines have remained largely unsuccessful despite their ability to potentiate both effector and memory CD8 T cell responses. There are two major hurdles for the success of DC-based vaccines: tumor-mediated immunosuppression and the functional limitation of the commonly used monocyte-derived dendritic cells (MoDCs). Due to their resistance to tumor-mediated suppression as inert vesicles, DC-derived exosomes (DCexos) have garnered much interest as cell-free therapeutic agents. However, current DCexo clinical trials have shown limited clinical benefits and failed to generate antigen-specific T cell responses. Another exciting development is the use of naturally circulating DCs instead of in vitro cultured DCs, as clinical trials with both human blood cDC2s (type 2 conventional DCs) and plasmacytoid DCs (pDCs) have shown promising results. pDC vaccines were particularly encouraging, especially in light of promising data from a recent clinical trial using a human pDC cell line, despite pDCs being considered tolerogenic and playing a suppressive role in tumors. However, how pDCs generate anti-tumor CD8 T cell immunity remains poorly understood, thus hindering their clinical advance. Using a pDC-targeted vaccine model, we have recently reported that while pDC-targeted vaccines led to strong cross-priming and durable CD8 T cell immunity, cross-presenting pDCs required cDCs to achieve cross-priming in vivo by transferring antigens to cDCs. Antigen transfer from pDCs to bystander cDCs was mediated by pDC-derived exosomes (pDCexos), which similarly required cDCs for cross-priming of antigen-specific CD8 T cells. pDCexos thus represent a new addition in our arsenal of DC-based cancer vaccines that would potentially combine the advantage of pDCs and DCexos.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
- Correspondence: ; Tel.: +1-716-400-2536
| |
Collapse
|
12
|
Plasmacytoid dendritic cells cross-prime naive CD8 T cells by transferring antigen to conventional dendritic cells through exosomes. Proc Natl Acad Sci U S A 2020; 117:23730-23741. [PMID: 32879009 DOI: 10.1073/pnas.2002345117] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although plasmacytoid dendritic cells (pDCs) have been shown to play a critical role in generating viral immunity and promoting tolerance to suppress antitumor immunity, whether and how pDCs cross-prime CD8 T cells in vivo remain controversial. Using a pDC-targeted vaccine model to deliver antigens specifically to pDCs, we have demonstrated that pDC-targeted vaccination led to strong cross-priming and durable CD8 T cell immunity. Surprisingly, cross-presenting pDCs required conventional DCs (cDCs) to achieve cross-priming in vivo by transferring antigens to cDCs. Taking advantage of an in vitro system where only pDCs had access to antigens, we further demonstrated that cross-presenting pDCs were unable to efficiently prime CD8 T cells by themselves, but conferred antigen-naive cDCs the capability of cross-priming CD8 T cells by transferring antigens to cDCs. Although both cDC1s and cDC2s exhibited similar efficiency in acquiring antigens from pDCs, cDC1s but not cDC2s were required for cross-priming upon pDC-targeted vaccination, suggesting that cDC1s played a critical role in pDC-mediated cross-priming independent of their function in antigen presentation. Antigen transfer from pDCs to cDCs was mediated by previously unreported pDC-derived exosomes (pDCexos), that were also produced by pDCs under various conditions. Importantly, all these pDCexos primed naive antigen-specific CD8 T cells only in the presence of bystander cDCs, similarly to cross-presenting pDCs, thus identifying pDCexo-mediated antigen transfer to cDCs as a mechanism for pDCs to achieve cross-priming. In summary, our data suggest that pDCs employ a unique mechanism of pDCexo-mediated antigen transfer to cDCs for cross-priming.
Collapse
|
13
|
Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12070663. [PMID: 32674488 PMCID: PMC7408110 DOI: 10.3390/pharmaceutics12070663] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
The onset of checkpoint inhibition revolutionized the treatment of cancer. However, studies from the last decade suggested that the sole enhancement of T cell functionality might not suffice to fight malignancies in all individuals. Dendritic cells (DCs) are not only part of the innate immune system, but also generals of adaptive immunity and they orchestrate the de novo induction of tolerogenic and immunogenic T cell responses. Thus, combinatorial approaches addressing DCs and T cells in parallel represent an attractive strategy to achieve higher response rates across patients. However, this requires profound knowledge about the dynamic interplay of DCs, T cells, other immune and tumor cells. Here, we summarize the DC subsets present in mice and men and highlight conserved and divergent characteristics between different subsets and species. Thereby, we supply a resource of the molecular players involved in key functional features of DCs ranging from their sentinel function, the translation of the sensed environment at the DC:T cell interface to the resulting specialized T cell effector modules, as well as the influence of the tumor microenvironment on the DC function. As of today, mostly monocyte derived dendritic cells (moDCs) are used in autologous cell therapies after tumor antigen loading. While showing encouraging results in a fraction of patients, the overall clinical response rate is still not optimal. By disentangling the general aspects of DC biology, we provide rationales for the design of next generation DC vaccines enabling to exploit and manipulate the described pathways for the purpose of cancer immunotherapy in vivo. Finally, we discuss how DC-based vaccines might synergize with checkpoint inhibition in the treatment of malignant diseases.
Collapse
|
14
|
Eckert N, Permanyer M, Yu K, Werth K, Förster R. Chemokines and other mediators in the development and functional organization of lymph nodes. Immunol Rev 2020; 289:62-83. [PMID: 30977201 DOI: 10.1111/imr.12746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022]
Abstract
Secondary lymphoid organs like lymph nodes (LNs) are the main inductive sites for adaptive immune responses. Lymphocytes are constantly entering LNs, scanning the environment for their cognate antigen and get replenished by incoming cells after a certain period of time. As only a minor percentage of lymphocytes recognizes cognate antigen, this mechanism of permanent recirculation ensures fast and effective immune responses when necessary. Thus, homing, positioning, and activation as well as egress require precise regulation within LNs. In this review we discuss the mediators, including chemokines, cytokines, growth factors, and others that are involved in the formation of the LN anlage and subsequent functional organization of LNs. We highlight very recent findings in the fields of LN development, steady-state migration in LNs, and the intranodal processes during an adaptive immune response.
Collapse
Affiliation(s)
- Nadine Eckert
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kai Yu
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
15
|
Amon L, Lehmann CHK, Baranska A, Schoen J, Heger L, Dudziak D. Transcriptional control of dendritic cell development and functions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:55-151. [PMID: 31759434 DOI: 10.1016/bs.ircmb.2019.10.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are major regulators of adaptive immunity, as they are not only capable to induce efficient immune responses, but are also crucial to maintain peripheral tolerance and thereby inhibit autoimmune reactions. DCs bridge the innate and the adaptive immune system by presenting peptides of self and foreign antigens as peptide MHC complexes to T cells. These properties render DCs as interesting target cells for immunomodulatory therapies in cancer, but also autoimmune diseases. Several subsets of DCs with special properties and functions have been described. Recent achievements in understanding transcriptional programs on single cell level, together with the generation of new murine models targeting specific DC subsets, advanced our current understanding of DC development and function. Thus, DCs arise from precursor cells in the bone marrow with distinct progenitor cell populations splitting the monocyte populations and macrophage populations from the DC lineage, which upon lineage commitment can be separated into conventional cDC1, cDC2, and plasmacytoid DCs (pDCs). The DC populations harbor intrinsic programs enabling them to react for specific pathogens in dependency on the DC subset, and thereby orchestrate T cell immune responses. Similarities, but also varieties, between human and murine DC subpopulations are challenging, and will require further investigation of human specimens under consideration of the influence of the tissue micromilieu and DC subset localization in the future.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Janina Schoen
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
16
|
Leylek R, Idoyaga J. The versatile plasmacytoid dendritic cell: Function, heterogeneity, and plasticity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:177-211. [PMID: 31759431 DOI: 10.1016/bs.ircmb.2019.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their identification as the natural interferon-producing cell two decades ago, plasmacytoid dendritic cells (pDCs) have been attributed diverse functions in the immune response. Their most well characterized function is innate, i.e., their rapid and robust production of type-I interferon (IFN-I) in response to viruses. However, pDCs have also been implicated in antigen presentation, activation of adaptive immune responses and immunoregulation. The mechanisms by which pDCs enact these diverse functions are poorly understood. One central debate is whether these functions are carried out by different pDC subpopulations or by plasticity in the pDC compartment. This chapter summarizes the latest reports regarding pDC function, heterogeneity, cell conversion and environmentally influenced plasticity, as well as the role of pDCs in infection, autoimmunity and cancer.
Collapse
Affiliation(s)
- Rebecca Leylek
- Department of Microbiology and Immunology, and Immunology Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, and Immunology Program, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
17
|
Gulubova M. Myeloid and Plasmacytoid Dendritic Cells and Cancer - New Insights. Open Access Maced J Med Sci 2019; 7:3324-3340. [PMID: 31949539 PMCID: PMC6953922 DOI: 10.3889/oamjms.2019.735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) use effective mechanisms to combat antigens and to bring about adaptive immune responses through their ability to stimulate näive T cells. At present, four major cell types are categorised as DCs: Classical or conventional (cDCs), Plasmacytoid (pDCs), Langerhans cells (LCs), and monocyte-derived DCs (Mo-DCs). It was suggested that pDCs, CD1c+ DCs and CD141+ DCs in humans are equivalent to mouse pDCs, CD11b+ DCs and CD8α+ DCs, respectively. Human CD141+ DCs compared to mouse CD8α+ DCs have remarkable functional and transcriptomic similarities. Characteristic markers, transcription factors, toll-like receptors, T helpers (Th) polarisation, cytokines, etc. of DCs are discussed in this review. Major histocompatibility complex (MHC) I and II antigen presentation, cross-presentation and Th polarisation are defined, and the dual role of DCs in the tumour is discussed. Human DCs are the main immune cells that orchestrate the immune response in the tumour microenvironment.
Collapse
Affiliation(s)
- Maya Gulubova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
18
|
Iberg CA, Hawiger D. Advancing immunomodulation by in vivo antigen delivery to DEC-205 and other cell surface molecules using recombinant chimeric antibodies. Int Immunopharmacol 2019; 73:575-580. [PMID: 31228685 DOI: 10.1016/j.intimp.2019.05.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/26/2022]
Abstract
A targeted delivery of defined antigens in vivo allows for the probing of relevant functions of the immune system. Recombinant chimeric antibodies, produced by genetically modifying original monoclonal antibodies specific for molecules expressed on dendritic cells and other immune cells, have paved the way for the development of such strategies and have become reliable tools for achieving a specific immunomodulation. These antibodies have proven important in both basic research and clinical applications, extending data obtained in disease models of autoimmunity and cancer. Here we will describe the advances gained from the experimental and therapeutic strategies based on the targeting of the specific antigens by recombinant chimeric antibodies to the multilectin receptor DEC-205 and other cell surface molecules.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Doisy Research Center, 1205 Carr Lane, St. Louis, MO 63104, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Doisy Research Center, 1205 Carr Lane, St. Louis, MO 63104, USA.
| |
Collapse
|
19
|
Ali S, Mann-Nüttel R, Schulze A, Richter L, Alferink J, Scheu S. Sources of Type I Interferons in Infectious Immunity: Plasmacytoid Dendritic Cells Not Always in the Driver's Seat. Front Immunol 2019; 10:778. [PMID: 31031767 PMCID: PMC6473462 DOI: 10.3389/fimmu.2019.00778] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/25/2019] [Indexed: 12/28/2022] Open
Abstract
Type I Interferons (IFNs) are hallmark cytokines produced in immune responses to all classes of pathogens. Type I IFNs can influence dendritic cell (DC) activation, maturation, migration, and survival, but also directly enhance natural killer (NK) and T/B cell activity, thus orchestrating various innate and adaptive immune effector functions. Therefore, type I IFNs have long been considered essential in the host defense against virus infections. More recently, it has become clear that depending on the type of virus and the course of infection, production of type I IFN can also lead to immunopathology or immunosuppression. Similarly, in bacterial infections type I IFN production is often associated with detrimental effects for the host. Although most cells in the body are thought to be able to produce type I IFN, plasmacytoid DCs (pDCs) have been termed the natural "IFN producing cells" due to their unique molecular adaptations to nucleic acid sensing and ability to produce high amounts of type I IFN. Findings from mouse reporter strains and depletion experiments in in vivo infection models have brought new insights and established that the role of pDCs in type I IFN production in vivo is less important than assumed. Production of type I IFN, especially the early synthesized IFNβ, is rather realized by a variety of cell types and cannot be mainly attributed to pDCs. Indeed, the cell populations responsible for type I IFN production vary with the type of pathogen, its tissue tropism, and the route of infection. In this review, we summarize recent findings from in vivo models on the cellular source of type I IFN in different infectious settings, ranging from virus, bacteria, and fungi to eukaryotic parasites. The implications from these findings for the development of new vaccination and therapeutic designs targeting the respectively defined cell types are discussed.
Collapse
Affiliation(s)
- Shafaqat Ali
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
- Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
| | - Ritu Mann-Nüttel
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Anja Schulze
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Lisa Richter
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Judith Alferink
- Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
20
|
Cancel JC, Crozat K, Dalod M, Mattiuz R. Are Conventional Type 1 Dendritic Cells Critical for Protective Antitumor Immunity and How? Front Immunol 2019; 10:9. [PMID: 30809220 PMCID: PMC6379659 DOI: 10.3389/fimmu.2019.00009] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are endowed with a unique potency to prime T cells, as well as to orchestrate their expansion, functional polarization and effector activity in non-lymphoid tissues or in their draining lymph nodes. The concept of harnessing DC immunogenicity to induce protective responses in cancer patients was put forward about 25 years ago and has led to a multitude of DC-based vaccine trials. However, until very recently, objective clinical responses were below expectations. Conventional type 1 DCs (cDC1) excel in the activation of cytotoxic lymphocytes including CD8+ T cells (CTLs), natural killer (NK) cells, and NKT cells, which are all critical effector cell types in antitumor immunity. Efforts to investigate whether cDC1 might orchestrate immune defenses against cancer are ongoing, thanks to the recent blossoming of tools allowing their manipulation in vivo. Here we are reporting on these studies. We discuss the mouse models used to genetically deplete or manipulate cDC1, and their main caveats. We present current knowledge on the role of cDC1 in the spontaneous immune rejection of tumors engrafted in syngeneic mouse recipients, as a surrogate model to cancer immunosurveillance, and how this process is promoted by type I interferon (IFN-I) effects on cDC1. We also discuss cDC1 implication in promoting the protective effects of immunotherapies in mouse preclinical models, especially for adoptive cell transfer (ACT) and immune checkpoint blockers (ICB). We elaborate on how to improve this process by in vivo reprogramming of certain cDC1 functions with off-the-shelf compounds. We also summarize and discuss basic research and clinical data supporting the hypothesis that the protective antitumor functions of cDC1 inferred from mouse preclinical models are conserved in humans. This analysis supports potential applicability to cancer patients of the cDC1-targeting adjuvant immunotherapies showing promising results in mouse models. Nonetheless, further investigations on cDC1 and their implications in anti-cancer mechanisms are needed to determine whether they are the missing key that will ultimately help switching cold tumors into therapeutically responsive hot tumors, and how precisely they mediate their protective effects.
Collapse
Affiliation(s)
- Jean-Charles Cancel
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Karine Crozat
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Marc Dalod
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Raphaël Mattiuz
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| |
Collapse
|
21
|
Ruedl C, Jung S. DTR-mediated conditional cell ablation-Progress and challenges. Eur J Immunol 2019; 48:1114-1119. [PMID: 29974950 DOI: 10.1002/eji.201847527] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/28/2018] [Indexed: 12/14/2022]
Abstract
Cell ablation is a valuable complement to mutagenesis for experimentally defining specific cell functions in physiology and pathophysiology in small animal models. One of the most popular ablation strategies involves transgenic expression of a primate diphtheria toxin receptor (DTR) on murine cells that are otherwise resistant to the bacterial exotoxin. The efforts of many laboratories using the DTR approach over the years have yielded numerous valuable insights into specific cell functions. Here, we will discuss the technical aspects of the DTR approach, including the strengths, pitfalls, and future strategies to overcome the shortcomings, highlighting a recent paper published in the European Journal of Immunology [El Hachem et al. Eur. J. Immunol. 2018 https://doi.org/10.1002/eji.201747351]. A particular focus will be given to the application of DTR approach to decipher in vivo functions of the murine myeloid cell compartment.
Collapse
Affiliation(s)
- Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
22
|
Mitchell D, Chintala S, Dey M. Plasmacytoid dendritic cell in immunity and cancer. J Neuroimmunol 2018; 322:63-73. [PMID: 30049538 DOI: 10.1016/j.jneuroim.2018.06.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/29/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) comprise a subset of dendritic cells characterized by their ability to produce large amount of type I interferon (IFN-I/α). Originally recognized for their role in modulating immune responses to viral stimulation, growing interest has been directed toward their contribution to tumorigenesis. Under normal conditions, Toll-like receptor (TLR)-activated pDCs exhibit robust IFN-α production and promote both innate and adaptive immune responses. In cancer, however, pDCs demonstrate an impaired response to TLR7/9 activation, decreased or absent IFN-α production and contribute to the establishment of an immunosuppressive tumor microenvironment. In addition to IFN-α production, pDCs can also act as antigen presenting cells (APCs) and regulate immune responses to various antigens. The significant role played by pDCs in regulating both the innate and adaptive components of the immune system makes them a critical player in cancer immunology. In this review, we discuss the development and function of pDCs as well as their role in innate and adaptive immunity. Finally, we summarize pDC contribution to cancer pathogenesis, with a special focus on primary malignant brain tumor, their significance in the era of immunotherapy and suggest potential strategies for pDC-targeted therapy.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, IU Simon Cancer Center, Indiana University, Indiana, USA
| | - Sreenivasulu Chintala
- Department of Neurosurgery, IU Simon Cancer Center, Indiana University, Indiana, USA
| | - Mahua Dey
- Department of Neurosurgery, IU Simon Cancer Center, Indiana University, Indiana, USA.
| |
Collapse
|
23
|
Ho NI, Camps MGM, de Haas EFE, Ossendorp F. Sustained cross-presentation capacity of murine splenic dendritic cell subsets in vivo. Eur J Immunol 2018; 48:1164-1173. [PMID: 29676785 PMCID: PMC6055716 DOI: 10.1002/eji.201747372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/22/2018] [Accepted: 04/17/2018] [Indexed: 11/16/2022]
Abstract
An exclusive feature of dendritic cells (DCs) is their ability to cross‐present exogenous antigens in MHC class I molecules. We analyzed the fate of protein antigen in antigen presenting cell (APC) subsets after uptake of naturally formed antigen‐antibody complexes in vivo. We observed that murine splenic DC subsets were able to present antigen in vivo for at least a week. After ex vivo isolation of four APC subsets, the presence of antigen in the storage compartments was visualized by confocal microscopy. Although all APC subsets stored antigen for many days, their ability and kinetics in antigen presentation was remarkably different. CD8α+ DCs showed sustained MHC class I‐peptide specific CD8+ T‐cell activation for more than 4 days. CD8α− DCs also presented antigenic peptides in MHC class I but presentation decreased after 48 h. In contrast, only the CD8α− DCs were able to present antigen in MHC class II to specific CD4+ T cells. Plasmacytoid DCs and macrophages were unable to activate any of the two T‐cell types despite detectable antigen uptake. These results indicate that naturally occurring DC subsets have functional antigen storage capacity for prolonged T‐cell activation and have distinct roles in antigen presentation to specific T cells in vivo.
Collapse
Affiliation(s)
- Nataschja I Ho
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel G M Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Edwin F E de Haas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Abstract
Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
25
|
Paterka M, Voss JO, Werr J, Reuter E, Franck S, Leuenberger T, Herz J, Radbruch H, Bopp T, Siffrin V, Zipp F. Dendritic cells tip the balance towards induction of regulatory T cells upon priming in experimental autoimmune encephalomyelitis. J Autoimmun 2017; 76:108-114. [DOI: 10.1016/j.jaut.2016.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 12/21/2022]
|
26
|
Lippens C, Duraes FV, Dubrot J, Brighouse D, Lacroix M, Irla M, Aubry-Lachainaye JP, Reith W, Mandl JN, Hugues S. IDO-orchestrated crosstalk between pDCs and Tregs inhibits autoimmunity. J Autoimmun 2016; 75:39-49. [PMID: 27470005 PMCID: PMC5127883 DOI: 10.1016/j.jaut.2016.07.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 01/21/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) have been shown to both mediate and prevent autoimmunity, and the regulation of their immunogenic versus tolerogenic functions remains incompletely understood. Here we demonstrate that, compared to other cells, pDCs are the major expressors of Indoleamine-2,3-dioxygenase (IDO) in steady-state lymph nodes (LNs). IDO expression by LN pDCs was closely dependent on MHCII-mediated, antigen-dependent, interactions with Treg. We further established that IDO production by pDCs was necessary to confer suppressive function to Tregs. During EAE development, IDO expression by pDCs was required for the generation of Tregs capable of dampening the priming of encephalitogenic T cell and disease severity. Thus, we describe a novel crosstalk between pDCs and Tregs: Tregs shape tolerogenic functions of pDCs prior to inflammation, such that pDCs in turn, promote Treg suppressive functions during autoimmunity.
Collapse
MESH Headings
- Animals
- Autoimmunity/genetics
- Autoimmunity/immunology
- Cells, Cultured
- Coculture Techniques
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Flow Cytometry
- Gene Expression Regulation, Enzymologic
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Lymph Nodes/enzymology
- Lymph Nodes/immunology
- Mice, Inbred C57BL
- Mice, Transgenic
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Carla Lippens
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Fernanda V Duraes
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Mathilde Lacroix
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Magali Irla
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | | | - Walter Reith
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Judith N Mandl
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland.
| |
Collapse
|
27
|
Meng C, Wang X, Xu Z, Hu M, Liu J, Pan Z, Chen X, Jiao X. Murine Flt3 ligand-generated plasmacytoid and conventional dendritic cells display functional differentiation in activation, inflammation, and antigen presentation during BCG infection in vitro. In Vitro Cell Dev Biol Anim 2016; 53:67-76. [PMID: 27496194 DOI: 10.1007/s11626-016-0076-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/22/2016] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are composed of distinct subsets. Their immunologic functions (especially in pathogenic infection, such as with mycobacteria) are poorly understood, largely because of their rarity and difficulty of preparation. We used the murine Fms-like tyrosine kinase 3 (Flt3) ligand to generate conventional DCs (FL-cDCs) and plasmacytoid DCs (FL-pDCs) and further evaluated their immunological responses to bacillus Calmette-Guérin (BCG) infection in vitro. BCG cells were observed inside both FL-cDCs and FL-pDCs by confocal microscopy, as confirmed by flow cytometric analysis showing a low infection rate of approximately 6 %, which was similar to in vivo results. The CD40, CD80, CD86, and MHC-II proteins were significantly upregulated in both FL-cDCs and -pDCs beginning at 4 h post-BCG exposure. FL-pDCs secreted TNF-α and IL-6 earlier and at significantly higher levels in the first 12 h following infection, but demonstrated delayed and weak activation and maturation compared to FL-cDCs. Although both subsets proved capable of presenting a mycobacterial antigen, FL-pDCs exhibited weaker activity in this respect than did FL-cDCs. In summary, the existence of FL-generated cDCs and pDCs imply functional differentiation in activation, inflammation, and antigen presentation, although both cells types participated extensively in the immune response to BCG infection.
Collapse
Affiliation(s)
- Chuang Meng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoyan Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Maozhi Hu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.,Testing Center, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
28
|
Ding Z, Dahlin JS, Xu H, Heyman B. IgE-mediated enhancement of CD4(+) T cell responses requires antigen presentation by CD8α(-) conventional dendritic cells. Sci Rep 2016; 6:28290. [PMID: 27306570 PMCID: PMC4910288 DOI: 10.1038/srep28290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/01/2016] [Indexed: 12/20/2022] Open
Abstract
IgE, forming an immune complex with small proteins, can enhance the specific antibody and CD4(+) T cell responses in vivo. The effects require the presence of CD23 (Fcε-receptor II)(+) B cells, which capture IgE-complexed antigens (Ag) in the circulation and transport them to splenic B cell follicles. In addition, also CD11c(+) cells, which do not express CD23, are required for IgE-mediated enhancement of T cell responses. This suggests that some type of dendritic cell obtains IgE-Ag complexes from B cells and presents antigenic peptides to T cells. To elucidate the nature of this dendritic cell, mice were immunized with ovalbumin (OVA)-specific IgE and OVA, and different populations of CD11c(+) cells, obtained from the spleens four hours after immunization, were tested for their ability to present OVA. CD8α(-) conventional dendritic cells (cDCs) were much more efficient in inducing specific CD4(+) T cell proliferation ex vivo than were CD8α(+) cDCs or plasmacytoid dendritic cells. Thus, IgE-Ag complexes administered intravenously are rapidly transported to the spleen by recirculating B cells where they are delivered to CD8α(-) cDCs which induce proliferation of CD4(+) T cells.
Collapse
Affiliation(s)
- Zhoujie Ding
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Joakim S. Dahlin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hui Xu
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Toivonen R, Kong L, Rasool O, Lund RJ, Lahesmaa R, Hänninen A. Activation of Plasmacytoid Dendritic Cells in Colon-Draining Lymph Nodes duringCitrobacter rodentiumInfection Involves Pathogen-Sensing and Inflammatory Pathways Distinct from Conventional Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:4750-9. [DOI: 10.4049/jimmunol.1600235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/28/2016] [Indexed: 12/23/2022]
|
30
|
Direct Delivery of Antigens to Dendritic Cells via Antibodies Specific for Endocytic Receptors as a Promising Strategy for Future Therapies. Vaccines (Basel) 2016; 4:vaccines4020008. [PMID: 27043640 PMCID: PMC4931625 DOI: 10.3390/vaccines4020008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are the most potent professional antigen presenting cells and are therefore indispensable for the control of immunity. The technique of antibody mediated antigen targeting to DC subsets has been the basis of intense research for more than a decade. Many murine studies have utilized this approach of antigen delivery to various kinds of endocytic receptors of DCs both in vitro and in vivo. Today, it is widely accepted that different DC subsets are important for the induction of select immune responses. Nevertheless, many questions still remain to be answered, such as the actual influence of the targeted receptor on the initiation of the immune response to the delivered antigen. Further efforts to better understand the induction of antigen-specific immune responses will support the transfer of this knowledge into novel treatment strategies for human diseases. In this review, we will discuss the state-of-the-art aspects of the basic principles of antibody mediated antigen targeting approaches. A table will also provide a broad overview of the latest studies using antigen targeting including addressed DC subset, targeted receptors, outcome, and applied coupling techniques.
Collapse
|
31
|
Carbon nanotubes' surface chemistry determines their potency as vaccine nanocarriers in vitro and in vivo. J Control Release 2016; 225:205-16. [PMID: 26802552 PMCID: PMC4778609 DOI: 10.1016/j.jconrel.2016.01.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/16/2016] [Accepted: 01/18/2016] [Indexed: 02/07/2023]
Abstract
Carbon nanotubes (CNTs) have shown marked capabilities in enhancing antigen delivery to antigen presenting cells. However, proper understanding of how altering the physical properties of CNTs may influence antigen uptake by antigen presenting cells, such as dendritic cells (DCs), has not been established yet. We hypothesized that altering the physical properties of multi-walled CNTs (MWNTs)-antigen conjugates, e.g. length and surface charge, can affect the internalization of MWNT-antigen by DCs, hence the induced immune response potency. For this purpose, pristine MWNTs (p-MWNTs) were exposed to various chemical reactions to modify their physical properties then conjugated to ovalbumin (OVA), a model antigen. The yielded MWNTs-OVA conjugates were long MWNT-OVA (~386nm), bearing net positive charge (5.8mV), or short MWNTs-OVA (~122nm) of increasing negative charges (-23.4, -35.8 or -39mV). Compared to the short MWNTs-OVA bearing high negative charges, short MWNT-OVA with the lowest negative charge demonstrated better cellular uptake and OVA-specific immune response both in vitro and in vivo. However, long positively-charged MWNT-OVA showed limited cellular uptake and OVA specific immune response in contrast to short MWNT-OVA displaying the least negative charge. We suggest that reduction in charge negativity of MWNT-antigen conjugate enhances cellular uptake and thus the elicited immune response intensity. Nevertheless, length of MWNT-antigen conjugate might also affect the cellular uptake and immune response potency; highlighting the importance of physical properties as a consideration in designing a MWNT-based vaccine delivery system.
Collapse
|
32
|
Abstract
The study of the intestinal dendritic cell (DC) compartment, its homeostasis, regulation, and response to challenges calls for the investigation within the physiological tissue context comprising the unique anatomic constellation of the epithelial single cell layer and the luminal microbiota, as well as neighboring immune and nonimmune cells. Here we provide protocols we developed that use a combination of conditional cell ablation, conditional compartment mutagenesis, and adoptive precursor transfers to study DC and other intestinal mononuclear phagocytes in in vivo context. We will highlight pitfalls and strengths of these approaches.
Collapse
Affiliation(s)
- Caterina Curato
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Biana Bernshtein
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tegest Aychek
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Steffen Jung
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
33
|
Redundant Function of Plasmacytoid and Conventional Dendritic Cells Is Required To Survive a Natural Virus Infection. J Virol 2015. [PMID: 26202250 DOI: 10.1128/jvi.01024-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Viruses that spread systemically from a peripheral site of infection cause morbidity and mortality in the human population. Innate myeloid cells, including monocytes, macrophages, monocyte-derived dendritic cells (mo-DC), and dendritic cells (DC), respond early during viral infection to control viral replication, reducing virus spread from the peripheral site. Ectromelia virus (ECTV), an orthopoxvirus that naturally infects the mouse, spreads systemically from the peripheral site of infection and results in death of susceptible mice. While phagocytic cells have a requisite role in the response to ECTV, the requirement for individual myeloid cell populations during acute immune responses to peripheral viral infection is unclear. In this study, a variety of myeloid-specific depletion methods were used to dissect the roles of individual myeloid cell subsets in the survival of ECTV infection. We showed that DC are the primary producers of type I interferons (T1-IFN), requisite cytokines for survival, following ECTV infection. DC, but not macrophages, monocytes, or granulocytes, were required for control of the virus and survival of mice following ECTV infection. Depletion of either plasmacytoid DC (pDC) alone or the lymphoid-resident DC subset (CD8α(+) DC) alone did not confer lethal susceptibility to ECTV. However, the function of at least one of the pDC or CD8α(+) DC subsets is required for survival of ECTV infection, as mice depleted of both populations were susceptible to ECTV challenge. The presence of at least one of these DC subsets is sufficient for cytokine production that reduces ECTV replication and virus spread, facilitating survival following infection. IMPORTANCE Prior to the eradication of variola virus, the orthopoxvirus that causes smallpox, one-third of infected people succumbed to the disease. Following successful eradication of smallpox, vaccination rates with the smallpox vaccine have significantly dropped. There is now an increasing incidence of zoonotic orthopoxvirus infections for which there are no effective treatments. Moreover, the safety of the smallpox vaccine is of great concern, as complications may arise, resulting in morbidity. Like many viruses that cause significant human diseases, orthopoxviruses spread from a peripheral site of infection to become systemic. This study elucidates the early requirement for innate immune cells in controlling a peripheral infection with ECTV, the causative agent of mousepox. We report that there is redundancy in the function of two innate immune cell subsets in controlling virus spread early during infection. The viral control mediated by these cell subsets presents a potential target for therapies and rational vaccine design.
Collapse
|
34
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
35
|
Schuster P, Thomann S, Werner M, Vollmer J, Schmidt B. A subset of human plasmacytoid dendritic cells expresses CD8α upon exposure to herpes simplex virus type 1. Front Microbiol 2015; 6:557. [PMID: 26082771 PMCID: PMC4451679 DOI: 10.3389/fmicb.2015.00557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/20/2015] [Indexed: 11/13/2022] Open
Abstract
Classical and plasmacytoid dendritic cells (DC) play important roles in the defense against murine and human infections with herpes simplex virus (HSV). So far, CD8α expression has only been reported for murine DC. CD8α+ DC have prominent cross-presenting activities, which are enhanced by murine CD8α+ PDC. The human orthologue of murine CD8α+ DC, the CD141 (BDCA3)+ DC, mainly cross-present after TLR3 ligation. We report here the serendipitous finding that a subset of human PDC upregulates CD8α upon HSV-1 stimulation, as shown by gene array and flow cytometry analyses. CD8α, not CD8ß, was expressed upon exposure. Markers of activation, migration, and costimulation were upregulated on CD8α-expressing human PDC. In these cells, increased cytokine and chemokine levels were detected that enhance development and function of T, B, and NK cells, and recruit immature DC, monocytes, and Th1 cells, respectively. Altogether, human CD8α+ PDC exhibit a highly activated phenotype and appear to recruit other immune cells to the site of inflammation. Further studies will show whether CD8α-expressing PDC contribute to antigen cross-presentation, which may be important for immune defenses against HSV infections in vitro and in vivo.
Collapse
Affiliation(s)
- Philipp Schuster
- Institute of Medical Microbiology and Hygiene, University of Regensburg , Regensburg, Germany ; Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Sabrina Thomann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Maren Werner
- Institute of Medical Microbiology and Hygiene, University of Regensburg , Regensburg, Germany
| | | | - Barbara Schmidt
- Institute of Medical Microbiology and Hygiene, University of Regensburg , Regensburg, Germany ; Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| |
Collapse
|
36
|
Dey M, Chang AL, Miska J, Wainwright DA, Ahmed AU, Balyasnikova IV, Pytel P, Han Y, Tobias A, Zhang L, Qiao J, Lesniak MS. Dendritic Cell-Based Vaccines that Utilize Myeloid Rather than Plasmacytoid Cells Offer a Superior Survival Advantage in Malignant Glioma. THE JOURNAL OF IMMUNOLOGY 2015; 195:367-76. [PMID: 26026061 DOI: 10.4049/jimmunol.1401607] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 05/02/2015] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DCs) are professional APCs that are traditionally divided into two distinct subsets, myeloid DC (mDCs) and plasmacytoid DC (pDCs). pDCs are known for their ability to secrete large amounts of IFN-α. Apart from IFN-α production, pDCs can also process Ag and induce T cell immunity or tolerance. In several solid tumors, pDCs have been shown to play a critical role in promoting tumor immunosuppression. We investigated the role of pDCs in the process of glioma progression in the syngeneic murine model of glioma. We show that glioma-infiltrating pDCs are the major APC in glioma and are deficient in IFN-α secretion (p < 0.05). pDC depletion leads to increased survival of the mice bearing intracranial tumor by decreasing the number of regulatory T cells (Tregs) and by decreasing the suppressive capabilities of Tregs. We subsequently compared the ability of mDCs and pDCs to generate effective antiglioma immunity in a GL261-OVA mouse model of glioma. Our data suggest that mature pDCs and mDCs isolated from naive mice can be effectively activated and loaded with SIINFEKL Ag in vitro. Upon intradermal injection in the hindleg, a fraction of both types of DCs migrate to the brain and lymph nodes. Compared to mice vaccinated with pDC or control mice, mice vaccinated with mDCs generate a robust Th1 type immune response, characterized by high frequency of CD4(+)T-bet(+) T cells and CD8(+)SIINFEKEL(+) T cells. This robust antitumor T cell response results in tumor eradication and long-term survival in 60% of the animals (p < 0.001).
Collapse
Affiliation(s)
- Mahua Dey
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Alan L Chang
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Jason Miska
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Derek A Wainwright
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Atique U Ahmed
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Irina V Balyasnikova
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Peter Pytel
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Yu Han
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Alex Tobias
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Lingjiao Zhang
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Jian Qiao
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| | - Maciej S Lesniak
- Brain Tumor Center, University of Chicago Pritzker School of Medicine, Chicago, IL 60637; and
| |
Collapse
|
37
|
Ghirelli C, Reyal F, Jeanmougin M, Zollinger R, Sirven P, Michea P, Caux C, Bendriss-Vermare N, Donnadieu MH, Caly M, Fourchotte V, Vincent-Salomon A, Sigal-Zafrani B, Sastre-Garau X, Soumelis V. Breast Cancer Cell-Derived GM-CSF Licenses Regulatory Th2 Induction by Plasmacytoid Predendritic Cells in Aggressive Disease Subtypes. Cancer Res 2015; 75:2775-87. [PMID: 25977333 DOI: 10.1158/0008-5472.can-14-2386] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 03/27/2015] [Indexed: 11/16/2022]
Abstract
Reciprocal interactions between tumor cells and their microenvironment vitally impact tumor progression. In this study, we show that GM-CSF produced by primary breast tumor cells induced the activation of plasmacytoid predendritic cells (pDC), a cell type critical to anti-viral immunity. pDC that expressed the GM-CSF receptor were increased in breast tumors compared with noninvolved adjacent breast tissue. Tumor-activated pDC acquired naïve CD4(+) T-cell stimulatory capacity and promoted a regulatory Th2 response. Finally, the concomitant increase of GM-CSF and pDC was significantly associated with relatively more aggressive breast cancer subtypes. Our results characterize the first tumor-derived factor that can activate pDC to promote a regulatory Th2 response, with implications for therapeutic targeting of a tumor-immune axis of growing recognition in its significance to cancer.
Collapse
Affiliation(s)
- Cristina Ghirelli
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Fabien Reyal
- UMR144 CNRS, Institut Curie, Paris, France. Institut Curie, Department of Surgical Oncology, Paris, France
| | - Marine Jeanmougin
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Raphaël Zollinger
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Philémon Sirven
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Paula Michea
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Christophe Caux
- Université Lyon 1, Lyon, France. INSERM U1052/CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France. Centre Léon Bérard, Lyon, France
| | - Nathalie Bendriss-Vermare
- Université Lyon 1, Lyon, France. INSERM U1052/CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France. Centre Léon Bérard, Lyon, France
| | - Marie-Hélène Donnadieu
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France
| | - Martial Caly
- Institut Curie, Department of Pathology, Paris, France
| | | | | | | | | | - Vassili Soumelis
- INSERM U932, Institut Curie, Paris, France. Institut Curie, Department of Immunology, Paris, France. Center of Clinical Investigations, Curie-IGR, Paris-Villejuif, France.
| |
Collapse
|
38
|
Dahlgren MW, Gustafsson-Hedberg T, Livingston M, Cucak H, Alsén S, Yrlid U, Johansson-Lindbom B. T follicular helper, but not Th1, cell differentiation in the absence of conventional dendritic cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5187-99. [PMID: 25917099 DOI: 10.4049/jimmunol.1401938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/18/2015] [Indexed: 11/19/2022]
Abstract
Development of long-lived humoral immunity is dependent on CXCR5-expressing T follicular helper (Tfh) cells, which develop concomitantly to effector Th cells that support cellular immunity. Conventional dendritic cells (cDCs) are critical APCs for initial priming of naive CD4(+) T cells but, importantly, also provide accessory signals that govern effector Th cell commitment. To define the accessory role of cDCs during the concurrent development of Tfh and effector Th1 cells, we performed high-dose Ag immunization in conjunction with the Th1-biased adjuvant polyinosinic:polycytidylic acid (pI:C). In the absence of cDCs, pI:C failed to induce Th1 cell commitment and IgG2c production. However, cDC depletion did not impair Tfh cell differentiation or germinal center formation, and long-lived IgG1 responses of unaltered affinity developed in mice lacking cDCs at the time point for immunization. Thus, cDCs are required for the pI:C-driven Th1 cell fate commitment but have no crucial accessory function in relation to Tfh cell differentiation.
Collapse
Affiliation(s)
| | - Tobias Gustafsson-Hedberg
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Megan Livingston
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Helena Cucak
- Immunology Section, Lund University, 221 84 Lund, Sweden; and
| | - Samuel Alsén
- Immunology Section, Lund University, 221 84 Lund, Sweden; and Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | |
Collapse
|
39
|
Aychek T, Mildner A, Yona S, Kim KW, Lampl N, Reich-Zeliger S, Boon L, Yogev N, Waisman A, Cua DJ, Jung S. IL-23-mediated mononuclear phagocyte crosstalk protects mice from Citrobacter rodentium-induced colon immunopathology. Nat Commun 2015; 6:6525. [PMID: 25761673 PMCID: PMC4382688 DOI: 10.1038/ncomms7525] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 02/07/2023] Open
Abstract
Gut homeostasis and mucosal immune defense rely on the differential contributions of dendritic cells (DC) and macrophages. Here we show that colonic CX3CR1(+) mononuclear phagocytes are critical inducers of the innate response to Citrobacter rodentium infection. Specifically, the absence of IL-23 expression in macrophages or CD11b(+) DC results in the impairment of IL-22 production and in acute lethality. Highlighting immunopathology as a death cause, infected animals are rescued by the neutralization of IL-12 or IFNγ. Moreover, mice are also protected when the CD103(+) CD11b(-) DC compartment is rendered deficient for IL-12 production. We show that IL-12 production by colonic CD103(+) CD11b(-) DC is repressed by IL-23. Collectively, in addition to its role in inducing IL-22 production, macrophage-derived or CD103(-) CD11b(+) DC-derived IL-23 is required to negatively control the otherwise deleterious production of IL-12 by CD103(+) CD11b(-) DC. Impairment of this critical mononuclear phagocyte crosstalk results in the generation of IFNγ-producing former TH17 cells and fatal immunopathology.
Collapse
Affiliation(s)
- Tegest Aychek
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Mildner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Simon Yona
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ki-Wook Kim
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nardy Lampl
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Louis Boon
- Bioceros, Yalelaan 46, 3584 CM Utrecht, The Netherlands
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Daniel J. Cua
- Merck Research Laboratories, 901 South California Avenue, Palo Alto, California 94304-1104, USA
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
40
|
Rogers NM, Ferenbach DA, Isenberg JS, Thomson AW, Hughes J. Dendritic cells and macrophages in the kidney: a spectrum of good and evil. Nat Rev Nephrol 2014; 10:625-43. [PMID: 25266210 PMCID: PMC4922410 DOI: 10.1038/nrneph.2014.170] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Renal dendritic cells (DCs) and macrophages represent a constitutive, extensive and contiguous network of innate immune cells that provide sentinel and immune-intelligence activity; they induce and regulate inflammatory responses to freely filtered antigenic material and protect the kidney from infection. Tissue-resident or infiltrating DCs and macrophages are key factors in the initiation and propagation of renal disease, as well as essential contributors to subsequent tissue regeneration, regardless of the aetiological and pathogenetic mechanisms. The identification, and functional and phenotypic distinction of these cell types is complex and incompletely understood, and the same is true of their interplay and relationships with effector and regulatory cells of the adaptive immune system. In this Review, we discuss the common and distinct characteristics of DCs and macrophages, as well as key advances that have identified the renal-specific functions of these important phagocytic, antigen-presenting cells, and their roles in potentiating or mitigating intrinsic kidney disease. We also identify remaining issues that are of priority for further investigation, and highlight the prospects for translational and therapeutic application of the knowledge acquired.
Collapse
Affiliation(s)
- Natasha M Rogers
- Vascular Medicine Institute and Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, W1544 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - David A Ferenbach
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jeffrey S Isenberg
- Vascular Medicine Institute and Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, W1544 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Angus W Thomson
- Vascular Medicine Institute and Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, W1544 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
41
|
Sage AP, Murphy D, Maffia P, Masters LM, Sabir SR, Baker LL, Cambrook H, Finigan AJ, Ait-Oufella H, Grassia G, Harrison JE, Ludewig B, Reith W, Hansson GK, Reizis B, Hugues S, Mallat Z. MHC Class II-restricted antigen presentation by plasmacytoid dendritic cells drives proatherogenic T cell immunity. Circulation 2014; 130:1363-73. [PMID: 25223984 DOI: 10.1161/circulationaha.114.011090] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.
Collapse
Affiliation(s)
- Andrew P Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Deirdre Murphy
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Pasquale Maffia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Leanne M Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Suleman R Sabir
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Lauren L Baker
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Helen Cambrook
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Alison J Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Hafid Ait-Oufella
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Gianluca Grassia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - James E Harrison
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Burkhard Ludewig
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Walter Reith
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Göran K Hansson
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Boris Reizis
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Stéphanie Hugues
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.).
| |
Collapse
|
42
|
Abstract
Classical dendritic cells (cDCs) form a critical interface between innate and adaptive immunity. As myeloid immune cell sentinels, cDCs are specialized in the sensing of pathogen challenges and cancer. They translate the latter for T cells into peptide form. Moreover, cDCs provide additional critical information on the original antigen context to trigger a diverse spectrum of appropriate protective responses. Here we review recent progress in our understanding of cDC subsets in mice. We will discuss cDC subset ontogeny and transcription factor dependencies, as well as emerging functional specializations within the cDC compartment in lymphoid and nonlymphoid tissues.
Collapse
Affiliation(s)
- Alexander Mildner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
43
|
Narusawa M, Inoue H, Sakamoto C, Matsumura Y, Takahashi A, Inoue T, Watanabe A, Miyamoto S, Miura Y, Hijikata Y, Tanaka Y, Inoue M, Takayama K, Okazaki T, Hasegawa M, Nakanishi Y, Tani K. TLR7 ligand augments GM-CSF-initiated antitumor immunity through activation of plasmacytoid dendritic cells. Cancer Immunol Res 2014; 2:568-80. [PMID: 24830413 DOI: 10.1158/2326-6066.cir-13-0143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vaccination with irradiated granulocyte macrophage colony-stimulating factor (GM-CSF)-transduced autologous tumor cells (GVAX) has been shown to induce therapeutic antitumor immunity. However, its effectiveness is limited. We therefore attempted to improve the antitumor effect by identifying little-known key pathways in GM-CSF-sensitized dendritic cells (GM-DC) in tumor-draining lymph nodes (TDLN). We initially confirmed that syngeneic mice subcutaneously injected with poorly immunogenic Lewis lung carcinoma (LLC) cells transduced with Sendai virus encoding GM-CSF (LLC/SeV/GM) remarkably rejected the tumor growth. Using cDNA microarrays, we found that expression levels of type I interferon (IFN)-related genes, predominantly expressed in plasmacytoid DCs (pDC), were significantly upregulated in TDLN-derived GM-DCs and focused on pDCs. Indeed, mouse experiments demonstrated that the effective induction of GM-CSF-induced antitumor immunity observed in immunocompetent mice treated with LLC/SeV/GM cells was significantly attenuated when pDC-depleted or IFNα receptor knockout (IFNAR(-/-)) mice were used. Importantly, in both LLC and CT26 colon cancer-bearing mice, the combinational use of imiquimod with autologous GVAX therapy overcame the refractoriness to GVAX monotherapy accompanied by tolerability. Mechanistically, mice treated with the combined vaccination displayed increased expression levels of CD86, CD9, and Siglec-H, which correlate with an antitumor phenotype, in pDCs, but decreased the ratio of CD4(+)CD25(+)FoxP3(+) regulatory T cells in TDLNs. Collectively, these findings indicate that the additional use of imiquimod to activate pDCs with type I IFN production, as a positive regulator of T-cell priming, could enhance the immunologic antitumor effects of GVAX therapy, shedding promising light on the understanding and treatment of GM-CSF-based cancer immunotherapy.
Collapse
Affiliation(s)
- Megumi Narusawa
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Hiroyuki Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Chika Sakamoto
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yumiko Matsumura
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Atsushi Takahashi
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Tomoko Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Ayumi Watanabe
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Shohei Miyamoto
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoshie Miura
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yasuki Hijikata
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoshihiro Tanaka
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Makoto Inoue
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Koichi Takayama
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Toshihiko Okazaki
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Mamoru Hasegawa
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Yoichi Nakanishi
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| | - Kenzaburo Tani
- Authors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, JapanAuthors' Affiliations: Department of Molecular Genetics, Medical Institute of Bioregulation; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences; Department of Advanced Cell and Molecular Therapy and Center for Clinical and Translational Research, Kyushu University Hospital, Kyushu University, Fukuoka; and DNAVEC Corporation, Tsukuba, Japan
| |
Collapse
|
44
|
Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5:140. [PMID: 24744762 PMCID: PMC3978348 DOI: 10.3389/fimmu.2014.00140] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 03/19/2014] [Indexed: 12/23/2022] Open
Abstract
The ability of dendritic cells (DCs) to cross-present tumor antigens has long been a focus of interest to physicians, as well as basic scientists, that aim to establish efficient cell-based cancer immune therapy. A prerequisite for exploiting this pathway for therapeutic purposes is a better understanding of the mechanisms that underlie the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses when initiated by DCs via cross-presentation. The ability of humans DC to perform cross-presentation is of utmost interest, as this cell type is a main target for cell-based immunotherapy in humans. The outcome of a cross-presentation event is guided by the nature of the antigen, the form of antigen uptake, and the subpopulation of DCs that performs presentation. Generally, CD8α+ DCs are considered to be the most potent cross-presenting DCs. This paradigm, however, only applies to soluble antigens. During adaptive immune responses, immune complexes form when antibodies interact with their specific epitopes on soluble antigens. Immunoglobulin G (IgG) immune complexes target Fc-gamma receptors on DCs to shuttle exogenous antigens efficiently into the cross-presentation pathway. This receptor-mediated cross-presentation pathway is a well-described route for the induction of strong CD8+ T cell responses. IgG-mediated cross-presentation is intriguing because it permits the CD8− DCs, which are commonly considered to be weak cross-presenters, to efficiently cross-present. Engaging multiple DC subtypes for cross-presentation might be a superior strategy to boost CTL responses in vivo. We here summarize our current understanding of how DCs use IgG-complexed antigens for the efficient induction of CTL responses. Because of its importance for human cell therapy, we also review the recent advances in the characterization of cross-presentation properties of human DC subsets.
Collapse
Affiliation(s)
- Barbara Platzer
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Madeleine Stout
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
45
|
Lim H, Kim YU, Yun K, Drouin SM, Chung Y. Distinct regulation of Th2 and Th17 responses to allergens by pulmonary antigen presenting cells in vivo. Immunol Lett 2013; 156:140-8. [DOI: 10.1016/j.imlet.2013.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/02/2013] [Accepted: 10/14/2013] [Indexed: 01/13/2023]
|
46
|
Obata T, Shibata N, Goto Y, Ishikawa I, Sato S, Kunisawa J, Kiyono H. Critical role of dendritic cells in T cell retention in the interfollicular region of Peyer's patches. THE JOURNAL OF IMMUNOLOGY 2013; 191:942-8. [PMID: 23772027 DOI: 10.4049/jimmunol.1200636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peyer's patches (PPs) simultaneously initiate active and quiescent immune responses in the gut. The immunological function is achieved by the rigid regulation of cell distribution and trafficking, but how the cell distribution is maintained remains to be elucidated. In this study, we show that binding of stromal cell-derived lymphoid chemokines to conventional dendritic cells (cDCs) is essential for the retention of naive CD4(+) T cells in the interfollicular region (IFR) of PPs. Transitory depletion of CD11c(high) cDCs in mice rapidly impaired the IFR structure in the PPs without affecting B cell follicles or germinal centers, lymphoid chemokine production from stromal cells, or the immigration of naive T cells into the IFRs of PPs. The cDC-orchestrated retention of naive T cells was mediated by heparinase-sensitive molecules that were expressed on cDCs and bound the lymphoid chemokine CCL21 produced from stromal cells. These data collectively reveal that interactions among cDCs, stromal cells, and naive T cells are necessary for the formation of IFRs in the PPs.
Collapse
Affiliation(s)
- Takashi Obata
- Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Elftman MD, Gonzalez-Hernandez MB, Kamada N, Perkins C, Henderson KS, Núñez G, Wobus CE. Multiple effects of dendritic cell depletion on murine norovirus infection. J Gen Virol 2013; 94:1761-1768. [PMID: 23636823 DOI: 10.1099/vir.0.052134-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) are permissive to murine norovirus (MNV) infection in vitro and in vivo. However, their roles during infection in vivo are not well defined. To determine the role of DCs during infection, conventional DCs were depleted from CD11c-DTR mice and infected with a persistent MNV strain. Viral titres in the intestine and secondary lymphoid organs were determined at early time points during infection, and anti-MNV antibody responses were analysed later during infection. Depletion of conventional DCs resulted in increased viral loads in intestinal tissues, impaired generation of antibody responses, and a failure of MNV to efficiently infect lymphoid tissues. These data suggest that DCs play multiple roles in MNV pathogenesis, in both innate immunity and the efficient generation of adaptive immune responses against MNV, as well as by promoting the dissemination of MNV to secondary lymphoid tissues. This is the first study to probe the roles of DCs in controlling and/or facilitating a norovirus infection in vivo and provides the basis for further studies aimed at defining mechanisms by which DCs control MNV replication and promote viral dissemination.
Collapse
Affiliation(s)
- Michael D Elftman
- Department of Biomedical and Diagnostic Sciences, University of Detroit Mercy School of Dentistry, Detroit, Michigan 48208, USA.,Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Mariam B Gonzalez-Hernandez
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Nobuhiko Kamada
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Cheryl Perkins
- Molecular Diagnostics Research, Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts 01887, USA
| | - Kenneth S Henderson
- Molecular Diagnostics Research, Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts 01887, USA
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
48
|
Dynamic imaging reveals promiscuous crosspresentation of blood-borne antigens to naive CD8+ T cells in the bone marrow. Blood 2013; 122:193-208. [PMID: 23637125 DOI: 10.1182/blood-2012-01-401265] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bone marrow (BM) hosts memory lymphocytes and supports secondary immune responses against blood-borne antigens, but it is unsettled whether primary responses occur there and which cells present the antigen. We used 2-photon microscopy in the BM of live mice to study these questions. Naïve CD8(+) T cells crawled rapidly at steady state but arrested immediately upon sensing antigenic peptides. Following infusion of soluble protein, various cell types were imaged ingesting the antigen, while antigen-specific T cells decelerated, clustered, upregulated CD69, and were observed dividing in situ to yield effector cells. Unlike in the spleen, T-cell responses persisted when BM-resident dendritic cells (DCs) were ablated but failed when all phagocytic cells were depleted. Potential antigen-presenting cells included monocytes and macrophages but not B cells. Collectively, our results suggest that the BM supports crosspresentation of blood-borne antigens similar to the spleen; uniquely, alongside DCs, other myeloid cells participate in crosspresentation.
Collapse
|
49
|
Ng D, Gommerman JL. The Regulation of Immune Responses by DC Derived Type I IFN. Front Immunol 2013; 4:94. [PMID: 23626590 PMCID: PMC3631742 DOI: 10.3389/fimmu.2013.00094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/08/2013] [Indexed: 11/13/2022] Open
Abstract
Our immune system bears the tremendous task of mounting effective anti-microbial responses whilst maintaining immunoregulatory functions to avoid autoimmunity. In order to quickly respond to pathogens, Dendritic cells (DC) are armed with pattern recognition receptors (PRRs), allowing them to recognize highly conserved pathogen-associated molecular patterns (PAMPs) that are uniquely expressed by invading microbes. PRR activation can trigger DCs to release the pleiotropic cytokine, Type I interferons (IFN), which facilitates various biological functions in different immune cell types. In this review, we will discuss the classical PRR-induced Type I IFN response in DCs as well as describe a novel mechanism for Type I IFN induction by the tumor-necrosis factor receptor superfamily (TNFRSF) members, TNFR-1 and lymphotoxin-β receptor (LTβR). While PRR activation during viral infection, produces large amounts of Type I IFN in a relative short period of time, TNFRSF-induced Type I IFN expression is modest with gradual kinetics. Type I IFN can exert pro-inflammatory effects, but in some cases it also facilitates immune-regulatory functions. Therefore, DCs are important regulators of immune responses by carefully modulating Type I IFN expression.
Collapse
Affiliation(s)
- Dennis Ng
- Department of Immunology, University of Toronto Toronto, ON, Canada
| | | |
Collapse
|
50
|
Nierkens S, Tel J, Janssen E, Adema GJ. Antigen cross-presentation by dendritic cell subsets: one general or all sergeants? Trends Immunol 2013; 34:361-70. [PMID: 23540650 DOI: 10.1016/j.it.2013.02.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/17/2013] [Accepted: 02/21/2013] [Indexed: 12/27/2022]
Abstract
Antigen cross-presentation describes the process through which dendritic cells (DCs) acquire exogenous antigens for presentation on MHC class I molecules. The ability to cross-present has been thought of as a feature of specialized DC subsets. Emerging data, however, suggest that the cross-presenting ability of each DC subset is tuned by and dependent on several factors, such as DC location and activation status, and the type of antigen and inflammatory signals. Thus, we argue that capacity of cross-presentation is not an exclusive trait of one or several distinct DC subtypes, but rather a common feature of the DC family in both mice and humans. Understanding DC subset activation and antigen-presentation pathways might yield improved tools and targets to exploit the unique cross-presenting capacity of DCs in immunotherapy.
Collapse
Affiliation(s)
- Stefan Nierkens
- Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Tumor Immunology Laboratory, Geert Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|