1
|
Park S, Yi E, Jeon J, Oh J, Xu Z, Park SH. The Role of Bone Marrow Stromal Cell Antigen 2 (BST2) in the Migration of Dendritic Cells to Lymph Nodes. Int J Mol Sci 2024; 26:149. [PMID: 39796009 PMCID: PMC11720714 DOI: 10.3390/ijms26010149] [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: 11/20/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/30/2025] Open
Abstract
Bone marrow stromal antigen 2 (BST2) is a host-restriction factor that plays multiple roles in the antiviral defense of innate immune responses, including the inhibition of viral particle release from virus-infected cells. BST2 may also be involved in the endothelial adhesion and migration of monocytes, but its importance in the immune system is still unclear. Immune cell adhesion and migration are closely related to the initiation of immune responses. In this study, we found that the expressions of the lymph node homing marker chemokine receptor 7 (CCR7) and an adhesion molecule intercellular adhesion molecule 1 (ICAM-1) in conventional dendritic cells (cDCs) were associated with BST2 expression. Interestingly, Bst2-/- cDCs showed lower chemotactic ability, including velocity and accumulative distance toward chemokine ligand 19 (CCL19) gradient in vitro, compared to wild-type cDCs. Bst2-/- cDCs also showed reduced migration and reduced retention capacity in draining lymph nodes in vivo. As a result, Bst2-/- cDCs as antigen-presenting cells induced lower antigen-specific B cell and T cell responses compared to Bst2+/+ cDCs. Notably, mice administered the influenza vaccine via Bst2-/- cDCs exhibited substantially inefficient virus clearance compared to mice administered the Bst2+/+ cDCs vaccine. Therefore, we propose that BST2, which plays a critical role in the effective migration and retention of cDCs, is involved in the development of optimal immunological effects in draining lymph nodes.
Collapse
Affiliation(s)
| | | | | | | | | | - Se-Ho Park
- College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea; (S.P.); (E.Y.); (J.J.); (J.O.); (Z.X.)
| |
Collapse
|
2
|
Yuan Z, Shu L, Fu J, Yang P, Wang Y, Sun J, Zheng M, Liu Z, Yang J, Song J, Song S, Cai Z. Single-Cell RNA Sequencing Deconstructs the Distribution of Immune Cells Within Abdominal Aortic Aneurysms in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1986-2003. [PMID: 39051127 DOI: 10.1161/atvbaha.124.321129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Inflammation is a key component in the development of abdominal aortic aneurysm (AAA), yet insights into the roles of immune cells and their interactions in this process are limited. METHODS Using single-cell RNA transcriptomic analysis, we deconstructed the CD45+ cell population in elastase-induced murine AAA at the single-cell level. We isolated each group of immune cells from murine AAA tissue at different time points and divided them into several subtypes, listed the remarkable differentially expressed genes, explored the developmental trajectories of immune cells, and demonstrated the interactions among them. RESULTS Our findings reveal significant differences in several immune cell subsets, including macrophages, dendritic cells, and T cells, within the AAA microenvironment compared with the normal aorta. Especially, conventional dendritic cell type 1 exclusively existed in the AAA tissue rather than the normal aortas. Via CellChat analysis, we identified several intercellular communication pathways like visfatin, which targets monocyte differentiation and neutrophil extracellular trap-mediated interaction between neutrophils and dendritic cells, which might contribute to AAA development. Some of these pathways were validated in human AAA. CONCLUSIONS Despite the absence of external pathogenic stimuli, AAA tissues develop a complex inflammatory microenvironment involving numerous immune cells. In-depth studies of the inflammatory network shall provide new strategies for patients with AAA.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Single-Cell Analysis
- Disease Models, Animal
- Mice, Inbred C57BL
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/immunology
- Mice
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Humans
- Macrophages/metabolism
- Macrophages/immunology
- Male
- Transcriptome
- RNA-Seq
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Gene Expression Profiling/methods
- Pancreatic Elastase
- Cell Communication
Collapse
Affiliation(s)
- Zhen Yuan
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Li Shu
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jiantao Fu
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Peipei Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Yidong Wang
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jie Sun
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Zheng
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Liu
- Vascular Surgery (Z.L.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Zhejun Cai
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| |
Collapse
|
3
|
Ung T, Rutledge NS, Weiss AM, Esser-Kahn AP, Deak P. Cell-targeted vaccines: implications for adaptive immunity. Front Immunol 2023; 14:1221008. [PMID: 37662903 PMCID: PMC10468591 DOI: 10.3389/fimmu.2023.1221008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Recent advancements in immunology and chemistry have facilitated advancements in targeted vaccine technology. Targeting specific cell types, tissue locations, or receptors can allow for modulation of the adaptive immune response to vaccines. This review provides an overview of cellular targets of vaccines, suggests methods of targeting and downstream effects on immune responses, and summarizes general trends in the literature. Understanding the relationships between vaccine targets and subsequent adaptive immune responses is critical for effective vaccine design. This knowledge could facilitate design of more effective, disease-specialized vaccines.
Collapse
Affiliation(s)
- Trevor Ung
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Nakisha S. Rutledge
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Peter Deak
- Chemical and Biological Engineering Department, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Abstract
The critical role of conventional dendritic cells in physiological cross-priming of immune responses to tumors and pathogens is widely documented and beyond doubt. However, there is ample evidence that a wide range of other cell types can also acquire the capacity to cross-present. These include not only other myeloid cells such as plasmacytoid dendritic cells, macrophages and neutrophils, but also lymphoid populations, endothelial and epithelial cells and stromal cells including fibroblasts. The aim of this review is to provide an overview of the relevant literature that analyzes each report cited for the antigens and readouts used, mechanistic insight and in vivo experimentation addressing physiological relevance. As this analysis shows, many reports rely on the exceptionally sensitive recognition of an ovalbumin peptide by a transgenic T cell receptor, with results that therefore cannot always be extrapolated to physiological settings. Mechanistic studies remain basic in most cases but reveal that the cytosolic pathway is dominant across many cell types, while vacuolar processing is most encountered in macrophages. Studies addressing physiological relevance rigorously remain exceptional but suggest that cross-presentation by non-dendritic cells may have significant impact in anti-tumor immunity and autoimmunity.
Collapse
Affiliation(s)
- François-Xavier Mauvais
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France; Service de Physiologie - Explorations Fonctionnelles Pédiatriques, AP-HP, Hôpital Universitaire Robert Debré, F-75019 Paris, France.
| | - Peter van Endert
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France; Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France.
| |
Collapse
|
5
|
Identification and Analysis of Potential Immune-Related Biomarkers in Endometriosis. J Immunol Res 2023; 2023:2975581. [PMID: 36660246 PMCID: PMC9845045 DOI: 10.1155/2023/2975581] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 01/12/2023] Open
Abstract
Background Endometriosis is an inflammatory gynecological disease leading to deep pelvic pain, dyspareunia, and infertility. The pathophysiology of endometriosis is complex and depends on a variety of biological processes and pathways. Therefore, there is an urgent need to identify reliable biomarkers for early detection and accurate diagnosis to predict clinical outcomes and aid in the early intervention of endometriosis. We screened transcription factor- (TF-) immune-related gene (IRG) regulatory networks as potential biomarkers to reveal new molecular subgroups for the early diagnosis of endometriosis. Methods To explore potential therapeutic targets for endometriosis, the Gene Expression Omnibus (GEO), Immunology Database and Analysis Portal (ImmPort), and TF databases were used to obtain data related to the recognition of differentially expressed genes (DEGs), differentially expressed IRGs (DEIRGs), and differentially expressed TFs (DETFs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DETFs and DEIRGs. Then, DETFs and DEIRGs were further validated in the external datasets of GSE51981 and GSE1230103. Then, we used quantitative real-time polymerase chain reaction (qRT-PCR) to verify the hub genes. Simultaneously, the Pearson correlation analysis and protein-protein interaction (PPI) analyses were used to indicate the potential mechanisms of TF-IRGs at the molecular level and obtain hub IRGs. Finally, the receiver operating characteristic (ROC) curve analysis was used to assess the diagnostic value of the hub IRGs. Results We screened a total of 94 DETFs and 121 DEIRGs in endometriosis. Most downregulated DETFs showed decreased expression in the endometria of moderate/severe endometriosis patients. The top-ranked upregulated DEIRGs were upregulated in the endometra of infertile women. Functional analysis showed that DETFs and DEIRGs may be involved in the biological behaviors and pathways of endometriosis. The TF-IRG PPI network was successfully constructed. Compared with the control group, high C3, VCAM1, ITGB2, and C3AR1 expression had statistical significance in endometriosis among the hub DEIRGs. They also showed higher sensitivity and specificity by ROC analysis for the diagnosis of endometriosis. Finally, compared with controls, C3 and VCAM1 were highly expressed in endometriosis tissue samples. In addition, they also showed high specificity and sensitivity for diagnosing endometriosis. Conclusion Overall, we discovered the TF-IRG regulatory network and analyzed 4 hub IRGs that were closely related to endometriosis, which contributes to the diagnosis of endometriosis. Additionally, we verified that DETFs or DEIRGs were associated with the clinicopathological features of endometriosis, and external datasets also confirmed the hub IRGs. Finally, C3 and VCAM1 were highly expressed in endometriosis tissue samples compared with controls and may be potential biomarkers of endometriosis, which are helpful for the early diagnosis of endometriosis.
Collapse
|
6
|
Piacente F, Bottero M, Benzi A, Vigo T, Uccelli A, Bruzzone S, Ferrara G. Neuroprotective Potential of Dendritic Cells and Sirtuins in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23084352. [PMID: 35457169 PMCID: PMC9025744 DOI: 10.3390/ijms23084352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins’ role in DCs functions.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
- Correspondence: ; Tel.: +39-(0)10-353-8150
| | - Giovanni Ferrara
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| |
Collapse
|
7
|
Bone marrow stromal antigen 2 (BST-2) genetic variants influence expression levels and disease outcome in HIV-1 chronically infected patients. Retrovirology 2022; 19:3. [PMID: 35081977 PMCID: PMC8793201 DOI: 10.1186/s12977-022-00588-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022] Open
Abstract
Background Bone marrow stromal antigen 2 (BST-2) also known as Tetherin (CD317/HM1.24), is a host restriction factor that blocks the release of HIV-1 virions from infected cells. Previous studies reported that BST-2 genetic variants or single nucleotide polymorphims (SNPs) have a preventative role during HIV-1 infection. However, the influence of BST-2 SNPs on expression levels remains unknown. In this study, we investigated the influence of BST-2 SNPs on expression levels and disease outcome in HIV-1 subtype C chronically infected antiretroviral therapy naïve individuals. Results We quantified BST-2 mRNA levels in peripheral blood mononuclear cells (PBMCs), determined BST-2 protein expression on the surface of CD4+ T cells using flow cytometry and genotyped two intronic single nucleotide polymorphisms (SNPs) rs919267 and rs919266 together with one SNP rs9576 located in the 3’ untranslated region (UTR) of bst-2 gene using TaqMan assays from HIV-1 uninfected and infected participants. Subsequently, we determined the ability of plasma antibody levels to mediate antibody-dependent cellular phagocytosis (ADCP) using gp120 consensus C and p24 subtype B/C protein. Fc receptor-mediated NK cell degranulation was evaluated as a surrogate for ADCC activity using plasma from HIV-1 positive participants. BST-2 mRNA expression levels in PBMCs and protein levels on CD4+ T cells were lower in HIV-1 infected compared to uninfected participants (p = 0.075 and p < 0.001, respectively). rs919267CT (p = 0.042) and rs919267TT (p = 0.045) were associated with lower BST-2 mRNA expression levels compared to rs919267CC in HIV-1 uninfected participants. In HIV-1 infected participants, rs919267CT associated with lower CD4 counts, (p = 0.003), gp120-IgG1 (p = 0.040), gp120-IgG3 (p = 0.016) levels but higher viral loads (p = 0.001) while rs919267TT was associated with lower BST-2 mRNA levels (p = 0.046), CD4 counts (p = 0.001), gp120-IgG1 levels (p = 0.033) but higher plasma viral loads (p = 0.007). Conversely, rs9576CA was associated with higher BST-2 mRNA expression levels (p = 0.027), CD4 counts (p = 0.079), gp120-IgG1 (p = 0.009), gp120-IgG3 (p = 0.039) levels but with lower viral loads (p = 0.037). Conclusion Our findings show that bst-2 SNPs mediate BST-2 expression and disease outcome, correlate with gp120-IgG1, gp120-IgG3 levels but not p24-IgG levels, ADCC and ADCP activity. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12977-022-00588-2.
Collapse
|
8
|
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
|
9
|
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: 35] [Impact Index Per Article: 11.7] [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
|
10
|
Nakagawa T, Tanino T, Onishi M, Tofukuji S, Kanazawa T, Ishioka Y, Itoh T, Kugimiya A, Katayama K, Yamamoto T, Nagira M, Ishii KJ. S-540956, a CpG Oligonucleotide Annealed to a Complementary Strand With an Amphiphilic Chain Unit, Acts as a Potent Cancer Vaccine Adjuvant by Targeting Draining Lymph Nodes. Front Immunol 2022; 12:803090. [PMID: 35003132 PMCID: PMC8735836 DOI: 10.3389/fimmu.2021.803090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Robust induction of cancer-antigen-specific CD8+ T cells is essential for the success of cancer peptide vaccines, which are composed of a peptide derived from a cancer-specific antigen and an immune-potentiating adjuvant, such as a Toll-like receptor (TLR) agonist. Efficient delivery of a vaccine antigen and an adjuvant to antigen-presenting cells in the draining lymph nodes (LNs) holds key to maximize vaccine efficacy. Here, we developed S-540956, a novel TLR9-agonistic adjuvant consisting of B-type CpG ODN2006 (also known as CpG7909), annealed to its complementary sequence oligodeoxynucleotide (ODN) conjugated to a lipid; it could target both a cancer peptide antigen and a CpG-adjuvant in the draining LNs. S-540956 accumulation in the draining LNs and activation of plasmacytoid dendritic cells (pDCs) were significantly higher than that of ODN2006. Mechanistic analysis revealed that S-540956 enhanced the induction of MHC class I peptide-specific CD8+ T cell responses via TLR9 in a CD4+ T cell-independent manner. In mice, the therapeutic effect of S-540956-adjuvanted with a human papillomavirus (HPV)-E7 peptide vaccine against HPV-E7-expressing TC-1 tumors was significantly better than that of an ODN2006-adjuvanted vaccine. Our findings demonstrate a novel adjuvant discovery with the complementary strand conjugated to a lipid, which enabled draining LN targeting and increased ODN2006 accumulation in draining LNs, thereby enhancing the adjuvant effect. Our findings imply that S-540956 is a promising adjuvant for cancer peptide vaccines and has a high potential for applications in various vaccines, including recombinant protein vaccines.
Collapse
Affiliation(s)
- Takayuki Nakagawa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Tetsuya Tanino
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Motoyasu Onishi
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Soichi Tofukuji
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takayuki Kanazawa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Yukichi Ishioka
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takeshi Itoh
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akira Kugimiya
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Kazufumi Katayama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takuya Yamamoto
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research (CVAR), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Morio Nagira
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research (CVAR), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Laboratory of Mock-up Vaccine Project, Center for Vaccine and Adjuvant Research (CVAR), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
11
|
Yao Y, Fu C, Zhou L, Mi QS, Jiang A. DC-Derived Exosomes for Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13153667. [PMID: 34359569 PMCID: PMC8345209 DOI: 10.3390/cancers13153667] [Citation(s) in RCA: 64] [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: 06/14/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/18/2022] Open
Abstract
As the initiators of adaptive immune responses, DCs play a central role in regulating the balance between CD8 T cell immunity versus tolerance to tumor antigens. Exploiting their function to potentiate host anti-tumor immunity, DC-based vaccines have been one of most promising and widely used cancer immunotherapies. However, DC-based cancer vaccines have not achieved the promised success in clinical trials, with one of the major obstacles being tumor-mediated immunosuppression. A recent discovery on the critical role of type 1 conventional DCs (cDC1s) play in cross-priming tumor-specific CD8 T cells and determining the anti-tumor efficacy of cancer immunotherapies, however, has highlighted the need to further develop and refine DC-based vaccines either as monotherapies or in combination with other therapies. DC-derived exosomes (DCexos) have been heralded as a promising alternative to DC-based vaccines, as DCexos are more resistance to tumor-mediated suppression and DCexo vaccines have exhibited better anti-tumor efficacy in pre-clinical animal models. However, DCexo vaccines have only achieved limited clinical efficacy and failed to induce tumor-specific T cell responses in clinical trials. The lack of clinical efficacy might be partly due to the fact that all current clinical trials used peptide-loaded DCexos from monocyte-derived DCs. In this review, we will focus on the perspective of expanding current DCexo research to move DCexo cancer vaccines forward clinically to realize their potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Yi Yao
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (Y.Y.); (C.F.); (L.Z.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (Y.Y.); (C.F.); (L.Z.)
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (Y.Y.); (C.F.); (L.Z.)
- 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; (Y.Y.); (C.F.); (L.Z.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
- Correspondence: (Q.-S.M.); (A.J.); Tel.: +313-876-1017 (Q.-S.M.); +313-876-7292 (A.J.)
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (Y.Y.); (C.F.); (L.Z.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
- Correspondence: (Q.-S.M.); (A.J.); Tel.: +313-876-1017 (Q.-S.M.); +313-876-7292 (A.J.)
| |
Collapse
|
12
|
Hatscher L, Amon L, Heger L, Dudziak D. Inflammasomes in dendritic cells: Friend or foe? Immunol Lett 2021; 234:16-32. [PMID: 33848562 DOI: 10.1016/j.imlet.2021.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/14/2022]
Abstract
Inflammasomes are cytosolic multiprotein complexes that crucially contribute to host defense against pathogens but are also involved in the pathogenesis of autoinflammatory diseases. Inflammasome formation leads to activation of effector caspases (caspase-1, 4, 5, or 11), the proteolytic maturation of IL-1β and IL-18 as well as cleavage of the pore-forming protein Gasdermin D. Dendritic cells are major regulators of immune responses as they bridge innate and adaptive immunity. We here summarize the current knowledge on inflammasome expression and formation in murine bone marrow-, human monocyte-derived as well as murine and human primary dendritic cells. Further, we discuss both, the beneficial and detrimental, involvement of inflammasome activation in dendritic cells in cancer, infections, and autoimmune diseases. As inflammasome activation is typically accompanied by Gasdermin d-mediated pyroptosis, which is an inflammatory form of programmed cell death, inflammasome formation in dendritic cells seems ill-advised. Therefore, we propose that hyperactivation, which is inflammasome activation without the induction of pyroptosis, may be a general model of inflammasome activation in dendritic cells to enhance Th1, Th17 as well as cytotoxic T cell responses.
Collapse
Affiliation(s)
- Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany.
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany; Medical Immunology Campus Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Germany.
| |
Collapse
|
13
|
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
|
14
|
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: 78] [Impact Index Per Article: 15.6] [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
|
15
|
Shi H, Luo K, Huang W. Bone Marrow Stromal Antigen 2 is a Potential Unfavorable Prognostic Factor for High-Grade Glioma. Onco Targets Ther 2020; 13:8723-8734. [PMID: 32943880 PMCID: PMC7468947 DOI: 10.2147/ott.s258631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/07/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Bone marrow stromal antigen 2 (BST2) is considered as a transmembrane glycoprotein and plays essential roles in innate immunity. It has been recently reported that up-regulation of BST2 was associated with the development of breast carcinoma. However, the clinical significance of BST2 in glioma has not been identified. The purpose of the present study is to explore the expression pattern and the role of BST2 in the progression of high-grade glioma. METHODS Expression levels of BST2 were tested in glioma tissues by analyzing the GEO database and immunohistochemistry staining. The prognostic role of BST2 in glioma was evaluated through univariate and multivariate analyses. In vitro and in vivo assays were conducted to confirm the role of BST2 on promoting glioma proliferation. RESULTS The mRNA level of BST2 was higher in glioma tissues than that in nontumorous brain tissues. High protein level of BST2 was correlated with larger tumor size and advanced WHO grade. Glioma patients with a high BST2 level had worse overall survival. In addition, BST2 was defined as an independent risk factor for glioma prognosis. Cellular and xenograft studies revealed that BST2 can significantly promote glioma proliferation. CONCLUSION Our study revealed that a high BST2 expression level was closely related to the unfavorable clinical features and poor prognosis of high-grade glioma patients. BST2 may serve as an invaluable prognostic indicator and novel therapeutic target for glioma treatment considering its membrane localization.
Collapse
Affiliation(s)
- Haiping Shi
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| | - Ke Luo
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| | - Wei Huang
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| |
Collapse
|
16
|
Sun L, Zhang W, Zhao Y, Wang F, Liu S, Liu L, Zhao L, Lu W, Li M, Xu Y. Dendritic Cells and T Cells, Partners in Atherogenesis and the Translating Road Ahead. Front Immunol 2020; 11:1456. [PMID: 32849502 PMCID: PMC7403484 DOI: 10.3389/fimmu.2020.01456] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a chronic process associated with arterial inflammation, the accumulation of lipids, plaque formation in vessel walls, and thrombosis with late mortal complications such as myocardial infarction and ischemic stroke. Immune and inflammatory responses have significant effects on every phase of atherosclerosis. Increasing evidence has shown that both innate and adaptive “arms” of the immune system play important roles in regulating the progression of atherosclerosis. Accumulating evidence suggests that a unique type of innate immune cell, termed dendritic cells (DCs), play an important role as central instigators, whereas adaptive immune cells, called T lymphocytes, are crucial as active executors of the DC immunity in atherogenesis. These two important immune cell types work in pairs to establish pro-atherogenic or atheroprotective immune responses in vascular tissues. Therefore, understanding the role of DCs and T cells in atherosclerosis is extremely important. Here, in this review, we will present a complete overview, based on existing knowledge of these two cell types in the atherosclerotic microenvironment, and discuss some of the novel means of targeting DCs and T cells as therapeutic tactics for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Yanfang Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Shan Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Lei Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Lin Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Wei Lu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Minghui Li
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| |
Collapse
|
17
|
Targeting Dendritic Cells with Antigen-Delivering Antibodies for Amelioration of Autoimmunity in Animal Models of Multiple Sclerosis and Other Autoimmune Diseases. Antibodies (Basel) 2020; 9:antib9020023. [PMID: 32549343 PMCID: PMC7345927 DOI: 10.3390/antib9020023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
The specific targeting of dendritic cells (DCs) using antigen-delivering antibodies has been established to be a highly efficient protocol for the induction of tolerance and protection from autoimmune processes in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS), as well as in some other animal disease models. As the specific mechanisms of such induced tolerance are being investigated, the newly gained insights may also possibly help to design effective treatments for patients. Here we review approaches applied for the amelioration of autoimmunity in animal models based on antibody-mediated targeting of self-antigens to DCs. Further, we discuss relevant mechanisms of immunological tolerance that underlie such approaches, and we also offer some future perspectives for the application of similar methods in certain related disease settings such as transplantation.
Collapse
|
18
|
Xu X, Wang Y, Xue F, Guan E, Tian F, Xu J, Zhang H. BST2 Promotes Tumor Growth via Multiple Pathways in Hepatocellular Carcinoma. Cancer Invest 2020; 38:329-337. [PMID: 32427495 DOI: 10.1080/07357907.2020.1769125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Bone marrow stromal antigen 2 (BST2) is a transmembrane glycoprotein and plays an essential role in innate immunity. Here we firstly found that BST2 expression was significantly elevated in hepatocellular carcinoma (HCC) tissues. High BST2 was closely related to the larger tumor size and more tumor number. Moreover, HCC patients with higher expression of BST2 had poorer overall survival and BST2 was identified as an independent unfavorable prognosis factor. Finally, we demonstrated that BST2 can promote proliferation capacity of tumor cells. In conclusion, HCC patients with higher BST2 expression were more predisposed to poorer clinical symptoms and unfavorable prognosis.
Collapse
Affiliation(s)
- Xiaoguang Xu
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Yu Wang
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Fangxi Xue
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Encui Guan
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Feng Tian
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Jian Xu
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Hongjin Zhang
- Department of Endocrinology and Nephrology, The Third People's Hospital of Linyi, Shandong, Linyi, China
| |
Collapse
|
19
|
Ye Y, Gaugler B, Mohty M, Malard F. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin Transl Immunology 2020; 9:e1139. [PMID: 32489664 PMCID: PMC7248678 DOI: 10.1002/cti2.1139] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are a unique subset of dendritic cells specialised in secreting high levels of type I interferons. pDCs play a crucial role in antiviral immunity and have been implicated in the initiation and development of many autoimmune and inflammatory diseases. This review summarises the latest advances in recent years in several aspects of pDC biology, with special focus on pDC heterogeneity, pDC development via the lymphoid pathway, and newly identified proteins/pathways involved in pDC trafficking, nucleic acid sensing and interferon production. Finally, we also highlight the current understanding of pDC involvement in autoimmunity and alloreactivity, and opportunities for pDC‐targeting therapies in these diseases. These new insights have contributed to answers to several fundamental questions remaining in pDC biology and may pave the way to successful pDC‐targeting therapy in the future.
Collapse
Affiliation(s)
- Yishan Ye
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Bone Marrow Transplantation Center The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Béatrice Gaugler
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France
| | - Mohamad Mohty
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| | - Florent Malard
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| |
Collapse
|
20
|
Zadeh VR, Urata S, Sakaguchi M, Yasuda J. Human BST-2/tetherin inhibits Junin virus release from host cells and its inhibition is partially counteracted by viral nucleoprotein. J Gen Virol 2020; 101:573-586. [PMID: 32375950 DOI: 10.1099/jgv.0.001414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Bone marrow stromal cell antigen-2 (BST-2), also known as tetherin, is an interferon-inducible membrane-associated protein. It effectively targets enveloped viruses at the release step of progeny viruses from host cells, thereby restricting the further spread of viral infection. Junin virus (JUNV) is a member of Arenaviridae, which causes Argentine haemorrhagic fever that is associated with a high rate of mortality. In this study, we examined the effect of human BST-2 on the replication and propagation of JUNV. The production of JUNV Z-mediated virus-like particles (VLPs) was significantly inhibited by over-expression of BST-2. Electron microscopy analysis revealed that BST-2 functions by forming a physical link that directly retains VLPs on the cell surface. Infection using JUNV showed that infectious JUNV production was moderately inhibited by endogenous or exogenous BST-2. We also observed that JUNV infection triggers an intense interferon response, causing an upregulation of BST-2, in infected cells. However, the expression of cell surface BST-2 was reduced upon infection. Furthermore, the expression of JUNV nucleoprotein (NP) partially recovered VLP production from BST-2 restriction, suggesting that the NP functions as an antagonist against antiviral effect of BST-2. We further showed that JUNV NP also rescued the production of Ebola virus VP40-mediated VLP from BST-2 restriction as a broad spectrum BST-2 antagonist. To our knowledge, this is the first report showing that an arenavirus protein counteracts the antiviral function of BST-2.
Collapse
Affiliation(s)
- Vahid Rajabali Zadeh
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan.,Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Jiro Yasuda
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| |
Collapse
|
21
|
Shirai S, Shibuya M, Kawai A, Tamiya S, Munakata L, Omata D, Suzuki R, Aoshi T, Yoshioka Y. Lipid Nanoparticles Potentiate CpG-Oligodeoxynucleotide-Based Vaccine for Influenza Virus. Front Immunol 2020; 10:3018. [PMID: 31998305 PMCID: PMC6962196 DOI: 10.3389/fimmu.2019.03018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Current influenza vaccines are generally effective against highly similar (homologous) strains, but their effectiveness decreases markedly against antigenically mismatched (heterologous) strains. One way of developing a universal influenza vaccine with a broader spectrum of protection is to use appropriate vaccine adjuvants to improve a vaccine's effectiveness and change its immune properties. Oligodeoxynucleotides (ODNs) with unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODNs), which are Toll-like-receptor 9 (TLR9) agonists, are among the most promising adjuvants and are already being used in humans. However, the development of novel delivery vehicles to improve adjuvant effects in vivo is highly desirable. Here, we assessed the potential of lipid nanoparticles (LNPs) as CpG ODN delivery vehicles in mice to augment the vaccine adjuvant effects of CpG ODN and enhance the protective spectrum of conventional influenza split vaccine (SV). In vitro, compared with CpG ODN, LNPs containing CpG ODNs (LNP-CpGs) induced significantly greater production of cytokines such as IL-12 p40 and IFN-α by mouse dendritic cells (DCs) and significantly greater expression of the co-stimulatory molecules CD80 and CD86 on DCs. In addition, after subcutaneous administration in mice, compared with CpG ODN, LNP-CpGs enhanced the expression of CD80 and CD86 on plasmacytoid DCs in draining lymph nodes. LNP-CpGs given with SV from H1N1 influenza A virus improved T-cell responses and gave a stronger not only SV-specific but also heterologous-virus-strain-specific IgG2c response than CpG ODN. Furthermore, immunization with SV plus LNP-CpGs protected against not only homologous strain challenge but also heterologous and heterosubtypic strain challenge, whereas immunization with SV plus CpG ODNs protected against homologous strain challenge only. We therefore demonstrated that LNP-CpGs improved the adjuvant effects of CpG ODN and broadened the protective spectrum of SV against influenza virus. We expect that this strategy will be useful in developing adjuvant delivery vehicles and universal influenza vaccines.
Collapse
Affiliation(s)
- Seiki Shirai
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Meito Shibuya
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Atsushi Kawai
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shigeyuki Tamiya
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Taiki Aoshi
- Vaccine Dynamics Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
22
|
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: 50] [Impact Index Per Article: 8.3] [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
|
23
|
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: 22] [Impact Index Per Article: 3.7] [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
|
24
|
Garnier A, Laffont S, Garnier L, Kaba E, Deutsch U, Engelhardt B, Guéry J. CD49d/CD29‐integrin controls the accumulation of plasmacytoid dendritic cells into the CNS during neuroinflammation. Eur J Immunol 2019; 49:2030-2043. [DOI: 10.1002/eji.201948086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/28/2019] [Accepted: 07/16/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Arnaud Garnier
- Centre de Physiopathologie de Toulouse Purpan (CPTP) Université de Toulouse INSERM CNRS UPS Toulouse France
| | - Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP) Université de Toulouse INSERM CNRS UPS Toulouse France
| | - Laure Garnier
- Centre de Physiopathologie de Toulouse Purpan (CPTP) Université de Toulouse INSERM CNRS UPS Toulouse France
| | - Elisa Kaba
- Theodor Kocher Institute University of Bern Bern Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute University of Bern Bern Switzerland
| | | | - Jean‐Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP) Université de Toulouse INSERM CNRS UPS Toulouse France
| |
Collapse
|
25
|
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
|
26
|
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: 102] [Impact Index Per Article: 17.0] [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
|
27
|
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: 132] [Impact Index Per Article: 22.0] [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
|
28
|
Vaccination with a DNA vaccine encoding CD317-targeting HBs antigen elicits enhanced immunity in mice. Biochem Biophys Res Commun 2018; 504:865-870. [PMID: 30219230 DOI: 10.1016/j.bbrc.2018.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Conventional hepatitis B virus (HBV) vaccines fail to induce protective antibody titers in 5-10% of immune-competent vaccines. Therefore, safe and effective HBV vaccines are still clinically needed. METHODS In this study, we developed a plasmid DNA vaccine encoding CD317 single-chain fragment variable (α317scFv) linked with the hepatitis B surface antigen (HBsAg) and detected the humoral and cellular immune responses elicited by this vaccine in BALB/c mice. RESULTS Vaccination with this fusion DNA vaccine in BALB/c mice induced more robust antiviral T cell and antibody immunity against HBsAg. Compared with mice vaccinated with control vaccine encoding HBsAg, the level of serum-circulating anti-HBsAg antibody (HBsAb) was nearly double in fusion DNA-vaccinated mice. More interesting, splenic lymphocytes isolated from fusion DNA-vaccinated mice showed more potent proliferation and IFN-γ production after being re-stimulated with recombinant HBsAg in vitro. And not only that, the cytotoxicity of fusion DNA vaccine-sensitized splenocytes was ∼3-fold higher than that of controls. CONCLUSION Taken together, our results reveal that the fusion DNA vaccine can induce more effective immunological protection against HBV, and is a promising candidate for preventing HBV infection.
Collapse
|
29
|
Humbert M, Guery L, Brighouse D, Lemeille S, Hugues S. Intratumoral CpG-B Promotes Antitumoral Neutrophil, cDC, and T-cell Cooperation without Reprograming Tolerogenic pDC. Cancer Res 2018; 78:3280-3292. [PMID: 29588348 DOI: 10.1158/0008-5472.can-17-2549] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/13/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapies utilize distinct mechanisms to harness the power of the immune system to eradicate cancer cells. Therapeutic vaccines, aimed at inducing active immune responses against an existing cancer, are highly dependent on the immunological microenvironment, where many immune cell types display high levels of plasticity and, depending on the context, promote very different immunologic outcomes. Among them, plasmacytoid dendritic cells (pDC), known to be highly immunogenic upon inflammation, are maintained in a tolerogenic state by the tumor microenvironment. Here, we report that intratumoral (i.t.) injection of established solid tumors with CpG oligonucleotides-B (CpG-B) inhibits tumor growth. Interestingly, control of tumor growth was independent of tumor-associated pDC, which remained refractory to CpG-B stimulation and whose depletion did not alter the efficacy of the treatment. Instead, tumor growth inhibition subsequent to i.t. CpG-B injection depended on the recruitment of neutrophils into the milieu, resulting in the activation of conventional dendritic cells, subsequent increased antitumor T-cell priming in draining lymph nodes, and enhanced effector T-cell infiltration in the tumor microenvironment. These results reinforce the concept that i.t. delivery of TLR9 agonists alters the tumor microenvironment by improving the antitumor activity of both innate and adaptive immune cells.Significance: Intratumoral delivery of CpG-B disrupts the tolerogenic tumor microenvironment and inhibits tumor growth. Cancer Res; 78(12); 3280-92. ©2018 AACR.
Collapse
Affiliation(s)
- Marion Humbert
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Leslie Guery
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland.
| |
Collapse
|
30
|
Affandi AJ, Carvalheiro T, Radstake TRDJ, Marut W. Dendritic cells in systemic sclerosis: Advances from human and mice studies. Immunol Lett 2017; 195:18-29. [PMID: 29126878 DOI: 10.1016/j.imlet.2017.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is a complex heterogeneous fibrotic autoimmune disease with an unknown exact etiology, and characterized by three hallmarks: fibrosis, vasculopathy, and immune dysfunction. Dendritic cells (DCs) are specialized cells in pathogen sensing with high potency of antigen presentation and capable of releasing mediators to shape the immune response. Altered DCs distributions and their impaired functions may account for their role in breaking the immune tolerance and driving inflammation in SSc, and the direct contribution of DCs in promoting endothelial dysfunction and fibrotic process has only begun to be understood. Plasmacytoid dendritic cells in particular have been implicated due to their high production of type I interferon as well as other cytokines and chemokines, including the pro-inflammatory and anti-angiogenic CXCL4. Furthermore, a deeper understanding of human and mouse DC biology has clarified their identification and function in different tissues, and novel DC subsets have only recently been discovered. In this review, we highlight key findings and recent advances exploring DC role in the pathogenesis of SSc and other related autoimmune diseases, and consideration of their potential use as targeted therapy in SSc.
Collapse
Affiliation(s)
- Alsya J Affandi
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tiago Carvalheiro
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wioleta Marut
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Protective Role of BST2 Polymorphisms in Mother-to-Child Transmission of HIV-1 and Adult AIDS Progression. J Acquir Immune Defic Syndr 2017; 72:237-41. [PMID: 26885809 DOI: 10.1097/qai.0000000000000949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bone marrow stromal cell antigen-2 (BST-2)/Tetherin is a restriction factor that prevents Human immunodeficiency virus type 1 (HIV-1) release from infected cells and mediates pro-inflammatory cytokine production. This study investigated the risk conferred by single nucleotide polymorphisms (rs919266, rs9192677, and rs9576) at BST-2 coding gene (BST2) in HIV-1 mother-to-child transmission and in disease progression. Initially, 101 HIV-1+ pregnant women and 331 neonates exposed to HIV-1 from Zambia were enrolled. Additional BST2 single nucleotide polymorphism analyses were performed in 2 cohorts with acquired immunodeficiency syndrome (AIDS) progression: an adult Brazilian cohort (37 rapid, 30 chronic and 21 long-term non-progressors) and an Italian pediatric cohort (21 rapid and 67 slow progressors). The rs9576A allele was nominally associated with protection during breastfeeding (P = 0.019) and individuals carrying rs919266 GA showed slower progression to AIDS (P = 0.033). Despite the influence of rs919266 and rs9576 on BST2 expression being still undetermined, a preventive role by BST2 polymorphisms was found during HIV-1 infection.
Collapse
|
32
|
Lehmann CHK, Baranska A, Heidkamp GF, Heger L, Neubert K, Lühr JJ, Hoffmann A, Reimer KC, Brückner C, Beck S, Seeling M, Kießling M, Soulat D, Krug AB, Ravetch JV, Leusen JHW, Nimmerjahn F, Dudziak D. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J Exp Med 2017; 214:1509-1528. [PMID: 28389502 PMCID: PMC5413326 DOI: 10.1084/jem.20160951] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Lehmann et al. targeted antigens to Fcγ receptors expressed on various antigen-presenting cells. Induced CD4+ and CD8+ T cell responses were solely dependent on CD11b+ and CD8+ DC subsets, respectively, but independent of receptor intrinsic ITAM or ITIM signaling domains. Dendritic cells (DCs) are efficient antigen-presenting cells equipped with various cell surface receptors for the direct or indirect recognition of pathogenic microorganisms. Interestingly, not much is known about the specific expression pattern and function of the individual activating and inhibitory Fcγ receptors (FcγRs) on splenic DC subsets in vivo and how they contribute to the initiation of T cell responses. By targeting antigens to select activating and the inhibitory FcγR in vivo, we show that antigen uptake under steady-state conditions results in a short-term expansion of antigen-specific T cells, whereas under inflammatory conditions especially, the activating FcγRIV is able to induce superior CD4+ and CD8+ T cell responses. Of note, this effect was independent of FcγR intrinsic activating signaling pathways. Moreover, despite the expression of FcγRIV on both conventional splenic DC subsets, the induction of CD8+ T cell responses was largely dependent on CD11c+CD8+ DCs, whereas CD11c+CD8− DCs were critical for priming CD4+ T cell responses.
Collapse
Affiliation(s)
- Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany.,Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale-Centre National de la Recherche Scientifique, 13288 Marseille-Luminy, France
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kirsten Neubert
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alana Hoffmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Katharina C Reimer
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christin Brückner
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Simone Beck
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michaela Seeling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Melissa Kießling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany
| | - Jeffrey V Ravetch
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, 3584 Utrecht, Netherlands
| | - Falk Nimmerjahn
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
33
|
Ngu LN, Nji NN, Ambada GE, Sagnia B, Sake CN, Tchadji JC, Njambe Priso GD, Lissom A, Tchouangueu TF, Manga Tebit D, Waffo AB, Park CG, Steinman RM, Überla K, Nchinda GW. In vivo targeting of protein antigens to dendritic cells using anti-DEC-205 single chain antibody improves HIV Gag specific CD4 + T cell responses protecting from airway challenge with recombinant vaccinia-gag virus. IMMUNITY INFLAMMATION AND DISEASE 2017; 7:55-67. [PMID: 28474788 PMCID: PMC6485703 DOI: 10.1002/iid3.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
Abstract
Introduction Targeting antigens to dendritic cells (DCs) in vivo via a DC‐restricted endocytic receptor, DEC205, has been validated to enhance immunity in several vaccine platforms. Particularly atttractive is selected delivery of proteins to DCs in vivo because it enables proteins to be more immunogenic and provides a cheaper and effective way for repeated immunizations. Methods In this study, we tested the efficacy of a single chain antibody to DEC205 (scDEC) to deliver protein antigens selectively to DCs in vivo and to induce protective immunity. Results In comparison to soluble Ovalbumin (OVA) antigen, when recombinant scDEC:OVA protein was injected subcutaneously (s.c.) into mice, the OVA protein was selectively presented by DCs to both TCR transgenic CD8+ and CD4+ T cells approximately 500 and 100 times more efficient than soluble OVA, respectively, and could persist for seven days following s.c. injection of the scDEC205:OVA. Similarly selective targeting of HIV Gag P24 to DCs in vivo using scDEC‐Gag protein plus polyICLC vaccine resulted in strong, long lasting, polyfuntional CD4+ T cells in mice which were protective against airway challenge by a recombinant vaccinia‐gag virus. Conclusion Thus targeting protein antigens to DCs using scDEC can be used either alone or in combination with other strategies for effective immunization.
Collapse
Affiliation(s)
- Loveline N Ngu
- Department of Biochemistry, University of Yaounde, Yaounde, Cameroon.,Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for Research on The Prevention and Management of HIV/AIDS, Yaounde, Cameroon
| | - Nadesh N Nji
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for Research on The Prevention and Management of HIV/AIDS, Yaounde, Cameroon
| | - Georgia E Ambada
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for Research on The Prevention and Management of HIV/AIDS, Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaoundeone, Yaounde, Cameroon
| | - Bertrand Sagnia
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for Research on The Prevention and Management of HIV/AIDS, Yaounde, Cameroon
| | - Carol Ngane Sake
- Department of Microbiology, University of Yaoundeone, Yaounde, Cameroon
| | - Jules Colinc Tchadji
- Department of Animal Biology and Physiology, University of Yaoundeone, Yaounde, Cameroon
| | | | - Abel Lissom
- Department of Animal Biology and Physiology, University of Yaoundeone, Yaounde, Cameroon
| | | | - Denis Manga Tebit
- Myles Thaler Center for AIDS and Human Retrovirus Research Department of Microbiology, Immunology and Cancer Biology, Charlottesville, Virginia, USA
| | - Alain Bopda Waffo
- Department of Biological Sciences # 223 Alabama State University, Montgomery, Alabama, USA
| | - Chae Gyu Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York, USA
| | - Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York, USA
| | - Klaus Überla
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Godwin W Nchinda
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for Research on The Prevention and Management of HIV/AIDS, Yaounde, Cameroon.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York, USA
| |
Collapse
|
34
|
Shahjahan Miah SM, Erick TK, Emerich DF. Dendritic Cell-Based Cancer Therapies: Current Status and Future Directions. CELL THERAPY 2017. [DOI: 10.1007/978-3-319-57153-9_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Scott JL, Wirth JR, EuDaly JG, Gilkeson GS, Cunningham MA. Plasmacytoid dendritic cell distribution and maturation are altered in lupus prone mice prior to the onset of clinical disease. Clin Immunol 2016; 175:109-114. [PMID: 28041989 DOI: 10.1016/j.clim.2016.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 01/28/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) and their production of type I interferons (IFN) are key pathogenic mediators of systemic lupus erythematosus (SLE). Despite the key role of pDCs in SLE, the mechanism by which pDCs promote disease is not well understood. The first objective for this study was to assess the number and maturation state of pDCs in pre-disease NZM2410 lupus prone mice compared to control mice. Second, we sought to identify mechanisms responsible for the alteration in pDCs in NZM mice prior to onset of clinical disease. We compared the number and percent of pDCs in the spleens and bone marrow (BM) of pre-disease NZM24010 (NZM) mice to C57BL/6 (B6) control mice. In the spleens of pre-disease NZM mice, pDC percent and number were increased. This increase occurs in parallel with a decrease in BM pDC number and percent in the NZM mice. The decrease in BM pDC number suggests the increase in spleen pDCs is a result of altered pDC distribution and not increased production of pDCs in the BM. To determine if pDC developmental potential is altered in lupus prone mice, we cultured BM from NZM and B6 mice in vitro. We found a reduced percentage/number of pDCs developing from the BM of NZM mice compared to B6 mice, which further supports that the increase in pDC number is a result of altered pDC distribution rather than increased pDC production. To better characterize the pDC population, we compared the percentage of mature pDCs in the spleens and BM of NZM mice to controls. In the NZM mice, there is a dramatic reduction in the number of mature pDCs in the BM of NZM mice, suggesting that mature pDCs exit the BM at a higher rate/earlier maturation time compared to healthy mice. We conclude that pDCs contribution to disease pathogenesis in NZM mice may include the alteration of pDC distribution to increase the number of pDCs in the spleen prior to disease onset.
Collapse
Affiliation(s)
- Jennifer L Scott
- Department of Microbiology and Immunology, College of Graduate Studies, Medical University of South Carolina, 173 Ashley Avenue, BSB 203, Charleston, SC 29425, USA.
| | - Jena R Wirth
- Division of Rheumatology and Immunology, Department of Medicine, 96 Jonathan Lucas Street, Suite 816, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Jackie G EuDaly
- Division of Rheumatology and Immunology, Department of Medicine, 96 Jonathan Lucas Street, Suite 816, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Gary S Gilkeson
- Division of Rheumatology and Immunology, Department of Medicine, 96 Jonathan Lucas Street, Suite 816, Medical University of South Carolina, Charleston, SC 29425, USA; Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street, Charleston, SC 29401, USA.
| | - Melissa A Cunningham
- Division of Rheumatology and Immunology, Department of Medicine, 96 Jonathan Lucas Street, Suite 816, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
36
|
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
|
37
|
Dursun E, Endele M, Musumeci A, Failmezger H, Wang SH, Tresch A, Schroeder T, Krug AB. Continuous single cell imaging reveals sequential steps of plasmacytoid dendritic cell development from common dendritic cell progenitors. Sci Rep 2016; 6:37462. [PMID: 27892478 PMCID: PMC5124969 DOI: 10.1038/srep37462] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/31/2016] [Indexed: 12/18/2022] Open
Abstract
Functionally distinct plasmacytoid and conventional dendritic cells (pDC and cDC) shape innate and adaptive immunity. They are derived from common dendritic cell progenitors (CDPs) in the murine bone marrow, which give rise to CD11c+ MHCII− precursors with early commitment to DC subpopulations. In this study, we dissect pDC development from CDP into an ordered sequence of differentiation events by monitoring the expression of CD11c, MHC class II, Siglec H and CCR9 in CDP cultures by continuous single cell imaging and tracking. Analysis of CDP genealogies revealed a stepwise differentiation of CDPs into pDCs in a part of the CDP colonies. This developmental pathway involved an early CD11c+ SiglecH− pre-DC stage and a Siglec H+ CCR9low precursor stage, which was followed rapidly by upregulation of CCR9 indicating final pDC differentiation. In the majority of the remaining CDP pedigrees however the Siglec H+ CCR9low precursor state was maintained for several generations. Thus, although a fraction of CDPs transits through precursor stages rapidly to give rise to a first wave of pDCs, the majority of CDP progeny differentiate more slowly and give rise to longer lived precursor cells which are poised to differentiate on demand.
Collapse
Affiliation(s)
- Ezgi Dursun
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152 Martinsried, Germany
| | - Max Endele
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Andrea Musumeci
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152 Martinsried, Germany
| | - Henrik Failmezger
- Max-Planck-Institute for Plant Breeding Research, Carl-von-Linné-Weg 10, 50829 Cologne, Germany.,Department of Biology, University of Cologne, Zülpicher Str. 47, 50829 Cologne, Germany
| | - Shu-Hung Wang
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152 Martinsried, Germany
| | - Achim Tresch
- Max-Planck-Institute for Plant Breeding Research, Carl-von-Linné-Weg 10, 50829 Cologne, Germany.,Department of Biology, University of Cologne, Zülpicher Str. 47, 50829 Cologne, Germany
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152 Martinsried, Germany
| |
Collapse
|
38
|
Sie C, Korn T. Dendritic cells in central nervous system autoimmunity. Semin Immunopathol 2016; 39:99-111. [PMID: 27888330 DOI: 10.1007/s00281-016-0608-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/13/2016] [Indexed: 02/01/2023]
Abstract
Dendritic cells (DCs) operate at the intersection of the innate and adaptive immune systems. DCs can promote or inhibit adaptive immune responses against neuroantigens. While DC intrinsic properties, i.e., their maturation state or the subset they belong to, are important determinants of the outcome of an autoimmune reaction, tissue-specific cues might also be relevant for the function of DCs. Thus, a better understanding of the performance of distinct DC subsets in specific anatomical niches, not only in lymphoid tissue but also in non-lymphoid tissues such as the meninges, the choroid plexus, and the inflamed CNS parenchyma, will be instrumental for the design of immune intervention strategies to chronic inflammatory diseases that do not put at risk basic surveillance functions of the immune system in the CNS. Here, we will review modern concepts of DC biology in steady state and during autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Christopher Sie
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
39
|
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
|
40
|
Abstract
Atherosclerosis is a complex chronic disease. The accumulation of myeloid cells in the arterial intima, including macrophages and dendritic cells (DCs), is a feature of early stages of disease. For decades, it has been known that monocyte recruitment to the intima contributes to the burden of lesion macrophages. Yet, this paradigm may require reevaluation in light of recent advances in understanding of tissue macrophage ontogeny, their capacity for self-renewal, as well as observations that macrophages proliferate throughout atherogenesis and that self-renewal is critical for maintenance of macrophages in advanced lesions. The rate of atherosclerotic lesion formation is profoundly influenced by innate and adaptive immunity, which can be regulated locally within atherosclerotic lesions, as well as in secondary lymphoid organs, the bone marrow and the blood. DCs are important modulators of immunity. Advances in the past decade have cemented our understanding of DC subsets, functions, hematopoietic origin, gene expression patterns, transcription factors critical for differentiation, and provided new tools for study of DC biology. The functions of macrophages and DCs overlap to some extent, thus it is important to reassess the contributions of each of these myeloid cells taking into account strict criteria of cell identification, ontogeny, and determine whether their key roles are within atherosclerotic lesions or secondary lymphoid organs. This review will highlight key aspect of macrophage and DC biology, summarize how these cells participate in different stages of atherogenesis and comment on complexities, controversies, and gaps in knowledge in the field.
Collapse
Affiliation(s)
- Myron I. Cybulsky
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Cheolho Cheong
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| | - Clinton S. Robbins
- From the Division of Advanced Diagnostics, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada (M.I.C., C.S.R.); Departments of Laboratory Medicine and Pathobiology (M.I.C., C.S.R.) and Immunology (C.S.R.), University of Toronto, Toronto, Ontario, Canada; and Laboratory of Cellular Physiology and Immunology, Institut de Researches Cliniques de Montréal, Montréal, Québec, Canada (C.C.)
| |
Collapse
|
41
|
Mira JC, Szpila BE, Nacionales DC, Lopez MC, Gentile LF, Mathias BJ, Vanzant EL, Ungaro R, Holden D, Rosenthal MD, Rincon J, Verdugo PT, Larson SD, Moore FA, Brakenridge SC, Mohr AM, Baker HV, Moldawer LL, Efron PA. Patterns of gene expression among murine models of hemorrhagic shock/trauma and sepsis. Physiol Genomics 2015; 48:135-44. [PMID: 26578697 DOI: 10.1152/physiolgenomics.00072.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/13/2015] [Indexed: 01/22/2023] Open
Abstract
Controversy remains whether the leukocyte genomic response to trauma or sepsis is dependent upon the initiating stimulus. Previous work illustrated poor correlations between historical models of murine trauma and sepsis (i.e., trauma-hemorrhage and lipopolysaccharide injection, respectively). The aim of this study is to examine the early genomic response in improved murine models of sepsis [cecal ligation and puncture (CLP)] and trauma [polytrauma (PT)] with and without pneumonia (PT+Pp). Groups of naïve, CLP, PT, and PT+Pp mice were killed at 2 h, 1 or 3 days. Total leukocytes were isolated for genome-wide expression analysis, and genes that were found to differ from control (false discovery rate adjusted P < 0.001) were assessed for fold-change differences. Spearman correlations were also performed. For all time points combined (CLP, PT, PT+Pp), there were 10,426 total genes that were found to significantly differ from naïve controls. At 2 h, the transcriptomic changes between CLP and PT showed a positive correlation (rs) of 0.446 (P < 0.0001) but were less positive thereafter. Correlations were significantly improved when we limited the analysis to common genes whose expression differed by a 1.5 fold-change. Both pathway and upstream analyses revealed the activation of genes known to be associated with pathogen-associated and damage-associated molecular pattern signaling, and early activation patterns of expression were very similar between polytrauma and sepsis at the earliest time points. This study demonstrates that the early leukocyte genomic response to sepsis and trauma are very similar in mice.
Collapse
Affiliation(s)
- Juan C Mira
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Benjamin E Szpila
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Dina C Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Lori F Gentile
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Brittany J Mathias
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Erin L Vanzant
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Ricardo Ungaro
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - David Holden
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Martin D Rosenthal
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Jaimar Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Patrick T Verdugo
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Shawn D Larson
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Henry V Baker
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| |
Collapse
|
42
|
Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J Immunol Res 2015; 2015:785634. [PMID: 26583156 PMCID: PMC4637118 DOI: 10.1155/2015/785634] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 08/26/2015] [Accepted: 09/03/2015] [Indexed: 01/08/2023] Open
Abstract
In therapeutic antitumor vaccination, dendritic cells play the leading role since they decide if, how, when, and where a potent antitumor immune response will take place. Since the disentanglement of the complexity and merit of different antigen-presenting cell subtypes, antitumor immunotherapeutic research started to investigate the potential benefit of targeting these subtypes in situ. This review will discuss which antigen-presenting cell subtypes are at play and how they have been targeted and finally question the true meaning of targeting antitumor-based vaccines.
Collapse
|
43
|
Abstract
Plasmacytoid dendritic cells (pDCs) are a unique DC subset that specializes in the production of type I interferons (IFNs). pDCs promote antiviral immune responses and have been implicated in the pathogenesis of autoimmune diseases that are characterized by a type I IFN signature. However, pDCs can also induce tolerogenic immune responses. In this Review, we summarize recent progress in the field of pDC biology, focusing on the molecular mechanisms that regulate the development and functions of pDCs, the pathways involved in their sensing of pathogens and endogenous nucleic acids, their functions at mucosal sites, and their roles in infection, autoimmunity and cancer.
Collapse
|
44
|
Kato Y, Zaid A, Davey GM, Mueller SN, Nutt SL, Zotos D, Tarlinton DM, Shortman K, Lahoud MH, Heath WR, Caminschi I. Targeting Antigen to Clec9A Primes Follicular Th Cell Memory Responses Capable of Robust Recall. THE JOURNAL OF IMMUNOLOGY 2015; 195:1006-14. [DOI: 10.4049/jimmunol.1500767] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/26/2015] [Indexed: 11/19/2022]
|
45
|
Price JD, Tarbell KV. The Role of Dendritic Cell Subsets and Innate Immunity in the Pathogenesis of Type 1 Diabetes and Other Autoimmune Diseases. Front Immunol 2015; 6:288. [PMID: 26124756 PMCID: PMC4466467 DOI: 10.3389/fimmu.2015.00288] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/18/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are key antigen-presenting cells that have an important role in autoimmune pathogenesis. DCs control both steady-state T cell tolerance and activation of pathogenic responses. The balance between these two outcomes depends on several factors, including genetic susceptibility, environmental signals that stimulate varied innate responses, and which DC subset is presenting antigen. Although the specific DC phenotype can diverge depending on the tissue location and context, there are four main subsets identified in both mouse and human: conventional cDC1 and cDC2, plasmacytoid DCs, and monocyte-derived DCs. In this review, we will discuss the role of these subsets in autoimmune pathogenesis and regulation, as well as the genetic and environmental signals that influence their function. Specific topics to be addressed include impact of susceptibility loci on DC subsets, alterations in DC subset development, the role of infection- and host-derived innate inflammatory signals, and the role of the intestinal microbiota on DC phenotype. The effects of these various signals on disease progression and the relative effects of DC subset composition and maturation level of DCs will be examined. These areas will be explored using examples from several autoimmune diseases but will focus mainly on type 1 diabetes.
Collapse
Affiliation(s)
- Jeffrey D Price
- Diabetes, Endocrinology, and Obesity Branch, Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Kristin V Tarbell
- Diabetes, Endocrinology, and Obesity Branch, Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
46
|
Guéry L, Dubrot J, Lippens C, Brighouse D, Malinge P, Irla M, Pot C, Reith W, Waldburger JM, Hugues S. Ag-presenting CpG-activated pDCs prime Th17 cells that induce tumor regression. Cancer Res 2014; 74:6430-40. [PMID: 25252912 DOI: 10.1158/0008-5472.can-14-1149] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Plasmacytoid dendritic cells (pDC) rapidly and massively produce type I IFN and other inflammatory cytokines in response to foreign nucleic acids, thereby indirectly influencing T-cell responses. Moreover, antigen (Ag)-presenting pDCs directly regulate T-cell differentiation. Depending on the immune environment, pDCs exhibit either tolerogenic or immunogenic properties. Here, we show that CpG-activated pDCs promote efficient Th17 differentiation. Indeed, Th17 responses are defective in mice selectively lacking MHCII on pDCs upon antigenic challenge. Importantly, in those mice, the frequency of Th17 cells infiltrating solid tumors is impaired. As a result, the recruitment of infiltrating leukocytes in tumors, including tumor-specific cytotoxic T lymphocytes (CTL), is altered and results in increased tumor growth. Importantly, following immunization with tumor Ag and CpG-B, MHCII-restricted Ag presentation by pDCs promotes the differentiation of antitumor Th17 cells that induce intratumor CTL recruitment and subsequent regression of established tumors. Our results highlight a new role for Ag presenting activated pDCs in promoting the development of Th17 cells and impacting on antitumor immunity.
Collapse
Affiliation(s)
- Leslie Guéry
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | - Magali Irla
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Centre d'immunology de Marseille Luminy, Université de la Méditerranée, Marseille, France
| | - Caroline Pot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Jean-Marc Waldburger
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland.
| |
Collapse
|
47
|
Sehgal K, Dhodapkar KM, Dhodapkar MV. Targeting human dendritic cells in situ to improve vaccines. Immunol Lett 2014; 162:59-67. [PMID: 25072116 DOI: 10.1016/j.imlet.2014.07.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) provide a critical link between innate and adaptive immunity. The potent antigen presenting properties of DCs makes them a valuable target for the delivery of immunogenic cargo. Recent clinical studies describing in situ DC targeting with antibody-mediated targeting of DC receptor through DEC-205 provide new opportunities for the clinical application of DC-targeted vaccines. Further advances with nanoparticle vectors which can encapsulate antigens and adjuvants within the same compartment and be targeted against diverse DC subsets also represent an attractive strategy for targeting DCs. This review provides a brief summary of the rationale behind targeting dendritic cells in situ, the existing pre-clinical and clinical data on these vaccines and challenges faced by the next generation DC-targeted vaccines.
Collapse
Affiliation(s)
- Kartik Sehgal
- Department of Medicine, Yale University, New Haven, CT, United States
| | | | | |
Collapse
|
48
|
Neubert K, Lehmann CHK, Heger L, Baranska A, Staedtler AM, Buchholz VR, Yamazaki S, Heidkamp GF, Eissing N, Zebroski H, Nussenzweig MC, Nimmerjahn F, Dudziak D. Antigen delivery to CD11c+CD8- dendritic cells induces protective immune responses against experimental melanoma in mice in vivo. THE JOURNAL OF IMMUNOLOGY 2014; 192:5830-8. [PMID: 24829411 DOI: 10.4049/jimmunol.1300975] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are central modulators of immune responses and, therefore, interesting target cells for the induction of antitumor immune responses. Ag delivery to select DC subpopulations via targeting Abs to DC inhibitory receptor 2 (DCIR2, clone 33D1) or to DEC205 was shown to direct Ags specifically to CD11c(+)CD8(-) or CD11c(+)CD8(+) DCs, respectively, in vivo. In contrast to the increasing knowledge about the induction of immune responses by efficiently cross-presenting CD11c(+)CD8(+) DCs, little is known about the functional role of Ag-presenting CD11c(+)CD8(-) DCs with regard to the initiation of protective immune responses. In this study, we demonstrate that Ag targeting to the CD11c(+)CD8(-) DC subpopulation in the presence of stimulating anti-CD40 Ab and TLR3 ligand polyinosinic-polycytidylic acid induces protective responses against rapidly growing tumor cells in naive animals under preventive and therapeutic treatment regimens in vivo. Of note, this immunization protocol induced a mixed Th1/Th2-driven immune response, irrespective of which DC subpopulation initially presented the Ag. Our results provide important information about the role of CD11c(+)CD8(-) DCs, which have been considered to be less efficient at cross-presenting Ags, in the induction of protective antitumor immune responses.
Collapse
Affiliation(s)
- Kirsten Neubert
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Christian H K Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Anna Baranska
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Anna Maria Staedtler
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology, and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sayuri Yamazaki
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Gordon F Heidkamp
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Nathalie Eissing
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany
| | - Henry Zebroski
- Proteomics Resource Center, The Rockefeller University, New York 10065
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York 10065; and
| | - Falk Nimmerjahn
- Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91052 Erlangen, Germany;
| |
Collapse
|
49
|
Pickering S, Hué S, Kim EY, Reddy S, Wolinsky SM, Neil SJD. Preservation of tetherin and CD4 counter-activities in circulating Vpu alleles despite extensive sequence variation within HIV-1 infected individuals. PLoS Pathog 2014; 10:e1003895. [PMID: 24465210 PMCID: PMC3900648 DOI: 10.1371/journal.ppat.1003895] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 12/06/2013] [Indexed: 01/19/2023] Open
Abstract
The HIV-1 Vpu protein is expressed from a bi-cistronic message late in the viral life cycle. It functions during viral assembly to maximise infectious virus release by targeting CD4 for proteosomal degradation and counteracting the antiviral protein tetherin (BST2/CD317). Single genome analysis of vpu repertoires throughout infection in 14 individuals infected with HIV-1 clade B revealed extensive amino acid diversity of the Vpu protein. For the most part, this variation in Vpu increases over the course of infection and is associated with predicted epitopes of the individual's MHC class I haplotype, suggesting CD8+ T cell pressure is the major driver of Vpu sequence diversity within the host. Despite this variability, the Vpu functions of targeting CD4 and counteracting both physical virus restriction and NF-κB activation by tetherin are rigorously maintained throughout HIV-1 infection. Only a minority of circulating alleles bear lesions in either of these activities at any given time, suggesting functional Vpu mutants are heavily selected against even at later stages of infection. Comparison of Vpu proteins defective for one or several functions reveals novel determinants of CD4 downregulation, counteraction of tetherin restriction, and inhibition of NF-κB signalling. These data affirm the importance of Vpu functions for in vivo persistence of HIV-1 within infected individuals, not simply for transmission, and highlight its potential as a target for antiviral therapy. The accessory protein Vpu, encoded by HIV-1, performs at least two major roles in the virus life cycle, namely the degradation of newly synthesized CD4 molecules and the counteraction of a host antiviral protein, tetherin. These activities promote the release of infectious viruses from host cells, and recent evidence suggests that Vpu function has been crucial for the cross-species transmission of HIV-1 from chimpanzees, and its subsequent pandemic spread in humans. Here we studied the functional variation in Vpu in infected individuals. We found that the Vpu amino acid sequence can be highly variable within an individual, and that this variation is likely to result from host immune responses targeting antigens derived from Vpu. However, despite this variation, Vpu's major functions are preserved, with only a minority of circulating alleles showing defects throughout the course of infection. These data suggest that defective Vpu proteins are selected against within the infected individual, implying that Vpu functions are critical for HIV-1 replication throughout natural infection, not simply at transmission. Therefore Vpu may represent a novel target for antiviral therapy to augment current treatment strategies for HIV/AIDS.
Collapse
Affiliation(s)
- Suzanne Pickering
- Department of Infectious Disease, King's College School of Medicine, Guy's Hospital, London, United Kingdom
| | - Stephane Hué
- MRC Centre for Medical Molecular Virology, University College London, London, United Kingdom
| | - Eun-Young Kim
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Susheel Reddy
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Steven M. Wolinsky
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Stuart J. D. Neil
- Department of Infectious Disease, King's College School of Medicine, Guy's Hospital, London, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Chappell CP, Giltiay NV, Dresch C, Clark EA. Controlling immune responses by targeting antigens to dendritic cell subsets and B cells. Int Immunol 2013; 26:3-11. [PMID: 24285828 DOI: 10.1093/intimm/dxt059] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Delivering antigens in vivo by coupling them to mAbs specific for unique receptors on antigen-presenting cells (APCs) is a promising approach for modulating immune responses. Antigen delivery to receptors found on myeloid dendritic cell (DC) subsets, plasmacytoid DCs and B cells has shown them all to be viable targets to stimulate either the cellular or humoral arms of the immune system. It is now evident that antigen-targeting approaches can also be used to invoke antigen-specific inhibition of immune responses. The outcome of activation versus inhibition is determined by a combination of factors that include the choice of APC, the receptor that is targeted, whether to include an adjuvant and, if so, which adjuvant to employ. In addition to their use as a means to modulate immune responses, antigen-targeting systems are also a useful method to investigate the function of DC subsets and the early mechanistic events that underlie the initiation of both cellular and humoral immune responses. In this review, we focus on the literature surrounding the control of B-cell responses when antigen is delivered to various APC subsets.
Collapse
|