51
|
Guignabert C. Dendritic Cells in Pulmonary Hypertension: Foot Soldiers or Hidden Enemies? Am J Respir Cell Mol Biol 2020; 63:551-552. [PMID: 32804536 PMCID: PMC7605161 DOI: 10.1165/rcmb.2020-0330ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Christophe Guignabert
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France, and.,Institut National de la Santé et de la Recherche Médicale (INSERM) - Unité Mixte de Recherche en Santé (UMR_S) 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
52
|
Narimatsu A, Hattori T, Usui Y, Ueno H, Funaki T, Komatsu H, Nakagawa H, Akiba H, Goto H. Blockade of costimulatory CD27/CD70 pathway promotes corneal allograft survival. Exp Eye Res 2020; 199:108190. [PMID: 32798537 DOI: 10.1016/j.exer.2020.108190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/26/2020] [Accepted: 08/07/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine whether the CD27/CD70 pathway plays a significant role in corneal allograft rejection by investigating the effect of blocking the CD27/CD70 pathway by anti-CD70 antibody on corneal allograft survival. METHODS Orthotopic penetrating keratoplasty was performed using C57BL/6 donor grafts and BALB/c recipients. Expression of CD27 and CD70 on rejected cornea was examined by immunohistochemistry. Corneal transplant recipients received intraperitoneal injection of anti-CD70 antibody (FR70) or control rat IgG. Alloreactivity was measured by mixed lymphoid reaction (MLR) in recipients administered control rat IgG and those administered anti-CD70 antibody. Corneal expression of IFN-γ and IL-12 was also examined in both groups. Graft opacity was assessed over an 8-week period and graft survival was evaluated using Kaplan-Meier survival curves. Proportion of CD4+CD44+ memory T cells in lymph nodes was measured by flow cytometry. RESULTS CD4+CD27+ cells and CD11c+CD70+ cells were present in rejected cornea. Anti-CD70 antibody administration suppressed alloreactivity in corneal allograft recipients, and inhibited IFN-γ expression in recipient cornea (p < 0.05). Anti-CD70 antibody suppressed opacity score of recipient cornea and prolonged corneal allograft survival (p < 0.05). Proportion of CD4+CD44+ memory T cells in recipient lymph nodes was reduced by anti-CD70 antibody treatment. CONCLUSION The CD27/CD70 pathway plays a significant role in corneal allograft rejection by initiating alloreactive Th1 cells and preserving memory T cells. Anti-CD70 antibody administration prolongs corneal allograft survival indicating the potential therapeutic effect of CD27/CD70 pathway blockade on corneal allograft rejection.
Collapse
Affiliation(s)
- Akitomo Narimatsu
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takaaki Hattori
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hiroki Ueno
- Department of Immunology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toshinari Funaki
- Department of Ophthalmology, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8431, Japan
| | - Hiroyuki Komatsu
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hayate Nakagawa
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hisaya Akiba
- Department of Immunology, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8431, Japan
| | - Hiroshi Goto
- Department of Ophthalmology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
53
|
Sulczewski FB, Martino LA, Almeida BDS, Zaneti AB, Ferreira NS, Amorim KNDS, Yamamoto MM, Apostolico JDS, Rosa DS, Boscardin SB. Conventional type 1 dendritic cells induce T H 1, T H 1-like follicular helper T cells and regulatory T cells after antigen boost via DEC205 receptor. Eur J Immunol 2020; 50:1895-1911. [PMID: 32673408 DOI: 10.1002/eji.202048694] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/16/2020] [Indexed: 12/21/2022]
Abstract
Conventional dendritic cells (cDCs) are specialized in antigen presentation. In the mouse spleen, cDCs are classified in cDC1s and cDC2s, and express DEC205 and DCIR2 endocytic receptors, respectively. Monoclonal antibodies (mAbs) αDEC205 (αDEC) and αDCIR2 have been fused to different antigens to deliver them to cDC1s or cDC2s. We immunized mice with αDEC and αDCIR2 fused to an antigen using Poly(I:C) as adjuvant. The initial immune response was analyzed from days 3 to 6 after the immunization. We also studied the influence of a booster dose. Our results showed that antigen targeting to cDC1s promoted a pro-inflammatory TH 1 cell response. Antigen targeting to cDC2s induced TFH cells, GCs, and plasma cell differentiation. After boost, antigen targeting to cDC1s improved the TH 1 cell response and induced TH 1-like TFH cells that led to an increase in specific antibody titers and IgG class switch. Additionally, a population of regulatory T cells was also observed. Antigen targeting to cDC2s did not improve the specific antibody response after boost. Our results add new information on the immune response induced after the administration of a booster dose with αDEC and αDCIR2 fusion mAbs. These results may be useful for vaccine design using recombinant mAbs.
Collapse
Affiliation(s)
| | - Larissa Alves Martino
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bianca da Silva Almeida
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Arthur Baruel Zaneti
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Natália Soares Ferreira
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Márcio Massao Yamamoto
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Juliana de Souza Apostolico
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.,Instituto de Investigaçao em Imunologia (iii), INCT, Sao Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil.,Instituto de Investigaçao em Imunologia (iii), INCT, Sao Paulo, Brazil
| |
Collapse
|
54
|
Van Den Eeckhout B, Van Hoecke L, Burg E, Van Lint S, Peelman F, Kley N, Uzé G, Saelens X, Tavernier J, Gerlo S. Specific targeting of IL-1β activity to CD8 + T cells allows for safe use as a vaccine adjuvant. NPJ Vaccines 2020; 5:64. [PMID: 32714571 PMCID: PMC7378068 DOI: 10.1038/s41541-020-00211-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 06/19/2020] [Indexed: 11/08/2022] Open
Abstract
Annual administration and reformulation of influenza vaccines is required for protection against seasonal infections. However, the induction of strong and long-lasting T cells is critical to reach broad and potentially lifelong antiviral immunity. The NLRP3 inflammasome and its product interleukin-1β (IL-1β) are pivotal mediators of cellular immune responses to influenza, yet, overactivation of these systems leads to side effects, which hamper clinical applications. Here, we present a bypass around these toxicities by targeting the activity of IL-1β to CD8+ T cells. Using this approach, we demonstrate safe inclusion of IL-1β as an adjuvant in vaccination strategies, leading to full protection of mice against a high influenza virus challenge dose by raising potent T cell responses. In conclusion, this paper proposes a class of IL-1β-based vaccine adjuvants and also provides further insight in the mechanics of cellular immune responses driven by IL-1β.
Collapse
Affiliation(s)
- Bram Van Den Eeckhout
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lien Van Hoecke
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Elianne Burg
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sandra Van Lint
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Frank Peelman
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Niko Kley
- Orionis Biosciences Inc, Waltham, MA 02451 USA
| | - Gilles Uzé
- CNRS 5235, University of Montpellier, 34090 Montpellier, France
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Orionis Biosciences Inc, Waltham, MA 02451 USA
| | - Sarah Gerlo
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
55
|
Arroyo Hornero R, Georgiadis C, Hua P, Trzupek D, He LZ, Qasim W, Todd JA, Ferreira RC, Wood KJ, Issa F, Hester J. CD70 expression determines the therapeutic efficacy of expanded human regulatory T cells. Commun Biol 2020; 3:375. [PMID: 32665635 PMCID: PMC7360768 DOI: 10.1038/s42003-020-1097-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/17/2020] [Indexed: 12/27/2022] Open
Abstract
Regulatory T cells (Tregs) are critical mediators of immune homeostasis. The co-stimulatory molecule CD27 is a marker of highly suppressive Tregs, although the role of the CD27-CD70 receptor-ligand interaction in Tregs is not clear. Here we show that after prolonged in vitro stimulation, a significant proportion of human Tregs gain stable CD70 expression while losing CD27. The expression of CD70 in expanded Tregs is associated with a profound loss of regulatory function and an unusual ability to provide CD70-directed co-stimulation to TCR-activated conventional T cells. Genetic deletion of CD70 or its blockade prevents Tregs from delivering this co-stimulatory signal, thus maintaining their regulatory activity. High resolution targeted single-cell RNA sequencing of human peripheral blood confirms the presence of CD27-CD70+ Treg cells. These findings have important implications for Treg-based clinical studies where cells are expanded over extended periods in order to achieve sufficient treatment doses.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Christos Georgiadis
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Peng Hua
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Dominik Trzupek
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Li-Zhen He
- Celldex Therapeutics, Inc., Hampton, NJ, 08827, USA
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Ricardo C Ferreira
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Kathryn J Wood
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
56
|
Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12070663. [PMID: 32674488 PMCID: PMC7408110 DOI: 10.3390/pharmaceutics12070663] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
The onset of checkpoint inhibition revolutionized the treatment of cancer. However, studies from the last decade suggested that the sole enhancement of T cell functionality might not suffice to fight malignancies in all individuals. Dendritic cells (DCs) are not only part of the innate immune system, but also generals of adaptive immunity and they orchestrate the de novo induction of tolerogenic and immunogenic T cell responses. Thus, combinatorial approaches addressing DCs and T cells in parallel represent an attractive strategy to achieve higher response rates across patients. However, this requires profound knowledge about the dynamic interplay of DCs, T cells, other immune and tumor cells. Here, we summarize the DC subsets present in mice and men and highlight conserved and divergent characteristics between different subsets and species. Thereby, we supply a resource of the molecular players involved in key functional features of DCs ranging from their sentinel function, the translation of the sensed environment at the DC:T cell interface to the resulting specialized T cell effector modules, as well as the influence of the tumor microenvironment on the DC function. As of today, mostly monocyte derived dendritic cells (moDCs) are used in autologous cell therapies after tumor antigen loading. While showing encouraging results in a fraction of patients, the overall clinical response rate is still not optimal. By disentangling the general aspects of DC biology, we provide rationales for the design of next generation DC vaccines enabling to exploit and manipulate the described pathways for the purpose of cancer immunotherapy in vivo. Finally, we discuss how DC-based vaccines might synergize with checkpoint inhibition in the treatment of malignant diseases.
Collapse
|
57
|
Najmuddin SUFS, Amin ZM, Tan SW, Yeap SK, Kalyanasundram J, Ani MAC, Veerakumarasivam A, Chan SC, Chia SL, Yusoff K, Alitheen NB. Cytotoxicity study of the interleukin-12-expressing recombinant Newcastle disease virus strain, rAF-IL12, towards CT26 colon cancer cells in vitro and in vivo. Cancer Cell Int 2020; 20:278. [PMID: 32612457 PMCID: PMC7325054 DOI: 10.1186/s12935-020-01372-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/20/2020] [Indexed: 12/14/2022] Open
Abstract
Background Oncolytic viruses have emerged as an alternative therapeutic modality for cancer as they can replicate specifically in tumour cells and induce toxic effects leading to apoptosis. Despite the great potentials and promising results shown in multiple studies, it appears that their efficacy is still moderate and deemed as not sufficient in clinical studies. In addressing this issue, genetic/molecular engineering approach has paved its way to improve the therapeutic efficacy as observed in the case of herpes simplex virus (HSV) expressing granulocyte–macrophage colony-stimulating factor (GM-CSF). This study aimed to explore the cytotoxicity effects of recombinant NDV strain AF2240-i expressing interleukin-12 (rAF-IL12) against CT26 colon cancer cells. Methods The cytotoxicity effect of rAF-IL12 against CT26 colon cancer cell line was determined by MTT assay. Based on the IC50 value from the anti-proliferative assay, further downward assays such as Annexin V FITC and cell cycle progression were carried out and measured by flow cytometry. Then, the in vivo study was conducted where the rAF-IL12 viral injections were given at the intra-tumoral site of the CT26 tumour-burden mice. At the end of the experiment, serum biochemical, T cell immunophenotyping, serum cytokine, histopathology of tumour and organ section, TUNEL assay, and Nanostring gene expression analysis were performed. Results The rAF-IL12 induced apoptosis of CT26 colon cancer cells in vitro as revealed in the Annexin V FITC analysis and also arrested the cancer cells progression at G1 phase of the cell cycle analysis. On the other hand, the rAF-IL12 significantly (p < 0.05) inhibited the growth of CT26 tumour in Balb/c mice and had regulated the immune system by increasing the level of CD4 + , CD8 + , IL-2, IL-12, and IFN-γ. Furthermore, the expression level of apoptosis-related genes (bax and p53) was up-regulated as a result of the rAF-IL12 treatment. Additionally, the rAF-IL12 had also down-regulated the expression level of KRAS, BRAF, MAPK1, Notch1, CCL2, and VEGF oncogenes. Besides, rAF-IL12 intra-tumoral delivery was considered safe and not hazardous to the host as evidenced in pathophysiology of the normal tissues and organs of the mice as well as from the serum biochemistry profile of liver and kidney. Conclusions These results indicated that rAF-IL12 had better anti-tumoral and cytotoxicity effects compared to its parental wild-type, AF2240-i in combatting the CT26 colon cancer model.
Collapse
Affiliation(s)
| | - Zahiah Mohamed Amin
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Sheau Wei Tan
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Swee Keong Yeap
- Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, Sepang, Selangor Darul Ehsan Malaysia
| | - Jeevanathan Kalyanasundram
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Muhamad Alhapis Che Ani
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | | | - Soon Choy Chan
- School of Foundation Studies, Perdana University, Block B and D1, MAEPS Building, MARDI Complex, Jalan MAEPS Perdana, 43400 Serdang, Selangor Darul Ehsan Malaysia
| | - Suet Lin Chia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Khatijah Yusoff
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Malaysian Genome Institute, National Institute of Biotechnology, Kajang, Jalan Bangi, 43000 Selangor Darul Ehsan Malaysia
| | - Noorjahan Banu Alitheen
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| |
Collapse
|
58
|
Gu FF, Zhang K, Ma LL, Liu YY, Li C, Hu Y, Yang QF, Liang JY, Zeng YL, Wang Y, Liu L. The Superior Ability of Human BDCA3 + (CD141 +) Dendritic Cells (DCs) to Cross-Present Antigens Derived From Necrotic Lung Cancer Cells. Front Immunol 2020; 11:1267. [PMID: 32655564 PMCID: PMC7325999 DOI: 10.3389/fimmu.2020.01267] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/19/2020] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) play a key role in initiating and regulating the immune responses to pathogens, self-antigens, and cancers. Human blood DCs comprise a family of different subsets: plasmacytoid DCs (pDCs) and CD16+, CD1c/BDCA1+, and BDCA3+ (CD141+) myeloid DCs and possess different phenotypes and functional characteristics. Lung cancer is the most common cancer, with the highest morbidity and mortality in the world. However, which DC subset plays a leading role in the lung cancer immune responses is unclear. We reanalyzed C-type lectin domain family 9 member A (CLEC9A) and CD141 (THBD) gene expression profiles from the Cancer Genome Atlas (TCGA) database and performed the Kaplan-Meier survival analysis of overall survival for several cancers according to their expression levels. Next, we investigated the capacities of five human blood DC subsets to stimulate T cell proliferation and capture, process and (cross-) present tumor antigen. Human BDCA3+ (CD141+) DCs have a superior capacity to stimulate allogeneic CD4+T cells proliferation and induce superior Th1 response compared with other DC subsets. Interestingly, toll-like receptor (TLR) agonists have little effect on DCs to induce the proliferation of naïve CD4+ T cells, but contribute to their differentiation. Importantly, BDCA3+ (CD141+) DCs possess the most potent ability to cross-present human tumor antigen after their uptake of necrotic lung cancer cells despite their lower antigen uptake. These findings suggest that human BDCA3+ (CD141+) DCs are critical mediators of cytotoxic T lymphocyte responses against EGFR-positive lung cancer. Therefore, our findings may provide theoretical basis for the development of DC-based antitumor vaccines.
Collapse
Affiliation(s)
- Fei-Fei Gu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Li Ma
- Department of Oncology, Wuhan Brain Hospital, Wuhan, China
| | - Yang-Yang Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi-Fan Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Yan Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Lan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Analysis and Testing Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
59
|
Ashour D, Arampatzi P, Pavlovic V, Förstner KU, Kaisho T, Beilhack A, Erhard F, Lutz MB. IL-12 from endogenous cDC1, and not vaccine DC, is required for Th1 induction. JCI Insight 2020; 5:135143. [PMID: 32434994 PMCID: PMC7259537 DOI: 10.1172/jci.insight.135143] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Success of DC vaccines relies on the quality of antigen presentation, costimulation, lymph node migration, and the release of IL-12, in case of Th1 priming. Here, we provide evidence for interaction between the injected vaccine DCs with endogenous lymph node–resident DCs for Th1 induction. While migration of the injected DCs was essential for antigen delivery to the lymph node, the injected DCs contributed only partially to Th0 priming and were unable to instruct Th1 generation. Instead, we provide evidence that the lymph node–resident XCR1+ DCs are activated by the injected DCs to present the cognate antigen and release IL-12 for Th1 polarization. The timing of interactions in the draining lymph nodes appeared step-wise as (a) injected DCs with cognate T cells, (b) injected DCs with bystander DCs, and (c) bystander DCs with T cells. The transcriptome of the bystander DCs showed a downregulation of Treg- and Th2/Th9-inducing genes and self-antigen presentation, as well as upregulation of MHC class II and genes required for Th1 instruction. Together, these data show that injected mature lymph node migratory DCs direct T cell priming and bystander DC activation, but not Th1 polarization, which is mediated by endogenous IL-12p70+XCR1+ resident bystander DCs. Our results are of importance for clinical DC-based vaccinations against tumors where endogenous DCs may be functionally impaired by chemotherapy. Successful Th1 priming by DC vaccines in mice depends on IL-12 from endogenous and XCR1+ cDC1 population.
Collapse
Affiliation(s)
| | | | | | - Konrad U Förstner
- Core Unit Systems Medicine, University of Würzburg, Würzburg, Germany.,ZB MED, Information Centre for Life Sciences, Cologne, Germany.,TH Köln, University of Applied Sciences, Institute of Information Science, Cologne, Germany
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
60
|
Jafarzadeh L, Masoumi E, Fallah-Mehrjardi K, Mirzaei HR, Hadjati J. Prolonged Persistence of Chimeric Antigen Receptor (CAR) T Cell in Adoptive Cancer Immunotherapy: Challenges and Ways Forward. Front Immunol 2020; 11:702. [PMID: 32391013 PMCID: PMC7188834 DOI: 10.3389/fimmu.2020.00702] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/27/2020] [Indexed: 12/22/2022] Open
Abstract
CAR T cell qualities, such as persistence and functionality play important roles in determining the outcome of cancer immunotherapy. In spite of full functionality, it has been shown that poor persistence of CAR T cells can limit an effective antitumor immune response. Here, we outline specific strategies that can be employed to overcome intrinsic and extrinsic barriers to CAR T cell persistence. We also offer our viewpoint on how growing use of CAR T cells in various cancers may require modifications in the intrinsic and extrinsic survival signals of CAR T cells. We anticipate these amendments will additionally provide the rationales for generation of more persistent, and thereby, more effective CAR T cell treatments. CAR T cell qualities, such as persistence and functionality play important roles in determining the outcome of cancer immunotherapy. In spite of full functionality, it has been shown that poor persistence of CAR T cells can limit an effective antitumor immune response. Here, we outline specific strategies that can be employed to overcome intrinsic and extrinsic barriers to CAR T cell persistence. We also offer our viewpoint on how growing use of CAR T cells in various cancers may require modifications in the intrinsic and extrinsic survival signals of CAR T cells. We anticipate these amendments will additionally provide the rationales for generation of more persistent, and thereby, more effective CAR T cell treatments.
Collapse
Affiliation(s)
- Leila Jafarzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Masoumi
- Department of Medical Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Keyvan Fallah-Mehrjardi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
61
|
Clarke EC, Bradfute SB. The use of mice lacking type I or both type I and type II interferon responses in research on hemorrhagic fever viruses. Part 1: Potential effects on adaptive immunity and response to vaccination. Antiviral Res 2020; 174:104703. [DOI: 10.1016/j.antiviral.2019.104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022]
|
62
|
Imai J, Otani M, Sakai T. Distinct Subcellular Compartments of Dendritic Cells Used for Cross-Presentation. Int J Mol Sci 2019; 20:ijms20225606. [PMID: 31717517 PMCID: PMC6888166 DOI: 10.3390/ijms20225606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) present exogenous protein-derived peptides on major histocompatibility complex class I molecules to prime naïve CD8+ T cells. This DC specific ability, called cross-presentation (CP), is important for the activation of cell-mediated immunity and the induction of self-tolerance. Recent research revealed that endoplasmic reticulum-associated degradation (ERAD), which was first identified as a part of the unfolded protein response—a quality control system in the ER—plays a pivotal role in the processing of exogenous proteins in CP. Moreover, DCs express a variety of immuno-modulatory molecules and cytokines to regulate T cell activation in response to the environment. Although both CP and immuno-modulation are indispensable, contrasting ER conditions are required for their correct activity. Since ERAD substrates are unfolded proteins, their accumulation may result in ER stress, impaired cell homeostasis, and eventually apoptosis. In contrast, activation of the unfolded protein response should be inhibited for DCs to express immuno-modulatory molecules and cytokines. Here, we review recent advances on antigen CP, focusing on intracellular transport routes for exogenous antigens and distinctive subcellular compartments involved in ERAD.
Collapse
Affiliation(s)
- Jun Imai
- Correspondence: ; Tel.: +81-27-352-1180
| | | | | |
Collapse
|
63
|
Bourque J, Hawiger D. Immunomodulatory Bonds of the Partnership between Dendritic Cells and T Cells. Crit Rev Immunol 2019; 38:379-401. [PMID: 30792568 DOI: 10.1615/critrevimmunol.2018026790] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
By acquiring, processing, and presenting both foreign and self-antigens, dendritic cells (DCs) initiate T cell activation that is shaped through the immunomodulatory functions of a variety of cell-membrane-bound molecules including BTLA-HVEM, CD40-CD40L, CTLA-4-CD80/CD86, CD70-CD27, ICOS-ICOS-L, OX40-OX40L, and PD-L1-PD-1, as well as several key cytokines and enzymes such as interleukin-6 (IL-6), IL-12, IL-23, IL-27, transforming growth factor-beta 1 (TGF-β1), retinaldehyde dehydrogenase (Raldh), and indoleamine 2,3-dioxygenase (IDO). Some of these distinct immunomodulatory signals are mediated by specific subsets of DCs, therefore contributing to the functional specialization of DCs in the priming and regulation of immune responses. In addition to responding to the DC-mediated signals, T cells can reciprocally modulate the immunomodulatory capacities of DCs, further refining immune responses. Here, we review recent studies, particularly in experimental mouse systems, that have delineated the integrated mechanisms of crucial immunomodulatory pathways that enable specific populations of DCs and T cells to work intimately together as single functional units that are indispensable for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
64
|
Volckmar J, Knop L, Stegemann-Koniszewski S, Schulze K, Ebensen T, Guzmán CA, Bruder D. The STING activator c-di-AMP exerts superior adjuvant properties than the formulation poly(I:C)/CpG after subcutaneous vaccination with soluble protein antigen or DEC-205-mediated antigen targeting to dendritic cells. Vaccine 2019; 37:4963-4974. [PMID: 31320219 DOI: 10.1016/j.vaccine.2019.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/30/2019] [Accepted: 07/06/2019] [Indexed: 12/18/2022]
Abstract
Vaccination is the most efficient strategy to protect from infectious diseases and the induction of a protective immune response not only depends on the nature of the antigen, but is also influenced by the vaccination strategy and the co-administration of adjuvants. Therefore, the precise monitoring of adjuvant candidates and their immune modulatory properties is a crucial step in vaccine development. Here, one central aspect is the induction of appropriate humoral and cellular effector mechanisms. In our study we performed a direct comparison of two promising candidates in adjuvant development, the STING activator bis-(3,5)-cyclic dimeric adenosine monophosphate (c-di-AMP) and the Toll-like receptor ligand formulation poly(I:C)/CpG. These were evaluated in C57BL/6 mice using the model antigen ovalbumin (OVA) in subcutaneous vaccination with soluble protein as well as in a dendritic cell (DC) targeting approach (αDEC-OVA). Strikingly, c-di-AMP as compared to poly(I:C)/CpG resulted in significantly higher antigen-specific IgG antibody levels when used in immunization with soluble OVA as well as in antigen targeting to DC. In vaccination with soluble OVA, c-di-AMP induced a significantly stronger CTL, Th1 and IFNγ-producing CD8+ memory T cell response than poly(I:C)/CpG. The response was CTL and Th1 cell dominated, a profile shared by both adjuvants. In the context of targeting OVA to DC, c-di-AMP induced significantly increased Th1 and Th2 cell responses as compared to poly(I:C)/CpG. Interestingly, the Th1 response dominated the overall T cell response only when c-di-AMP was used, indicating a distinct modulatory property of c-di-AMP when the DC targeting immunization approach was exploited. Taken together, we describe superior properties of c-di-AMP as compared to poly(I:C)/CpG in subcutaneous vaccination with soluble antigen as well as antigen targeting to DC. This indicates exceptionally effective adjuvant properties for c-di-AMP and provides compelling evidence of its potential for further adjuvant development, especially also when using DC targeting approaches.
Collapse
Affiliation(s)
- Julia Volckmar
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Immune Regulation Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Laura Knop
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Immune Regulation Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Sabine Stegemann-Koniszewski
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Immune Regulation Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany; Experimental Pneumology, University Hospital for Pneumology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Immune Regulation Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany.
| |
Collapse
|
65
|
Remedios KA, Meyer L, Zirak B, Pauli ML, Truong HA, Boda D, Rosenblum MD. CD27 Promotes CD4 + Effector T Cell Survival in Response to Tissue Self-Antigen. THE JOURNAL OF IMMUNOLOGY 2019; 203:639-646. [PMID: 31209102 DOI: 10.4049/jimmunol.1900288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/29/2019] [Indexed: 11/19/2022]
Abstract
Signaling through CD27 plays a role in T cell activation and memory. However, it is currently unknown how this costimulatory receptor influences CD4+ effector T (Teff) cells in inflamed tissues. In the current study, we used a murine model of inducible self-antigen expression in the epidermis to elucidate the functional role of CD27 on autoreactive Teff cells. Expression of CD27 on Ag-specific Teff cells resulted in enhanced skin inflammation when compared with CD27-deficient Teff cells. CD27 signaling promoted the accumulation of IFN-γ and IL-2-producing T cells in skin draining lymph nodes in a cell-intrinsic fashion. Surprisingly, this costimulatory pathway had minimal effect on early T cell activation and proliferation. Instead, signaling through CD27 resulted in the progressive survival of Teff cells during the autoimmune response. Using BH3 profiling to assess mitochondrial cell priming, we found that CD27-deficient cells were equally as sensitive as CD27-sufficient cells to mitochondrial outer membrane polarization upon exposure to either BH3 activator or sensitizer peptides. In contrast, CD27-deficient Teff cells expressed higher levels of active caspase 8. Taken together, these results suggest that CD27 does not promote Teff cell survival by increasing expression of antiapoptotic BCL2 family members but instead acts by preferentially suppressing the cell-extrinsic apoptosis pathway, highlighting a previously unidentified role for CD27 in augmenting autoreactive Teff cell responses.
Collapse
Affiliation(s)
- Kelly A Remedios
- Department of Dermatology, University of California, San Francisco, CA 94143.,TRex Bio, Burlingame, CA 94010; and
| | - Lauren Meyer
- Department of Pediatrics, University of California, San Francisco, CA 94143
| | - Bahar Zirak
- Department of Dermatology, University of California, San Francisco, CA 94143
| | - Mariela L Pauli
- Department of Dermatology, University of California, San Francisco, CA 94143
| | | | - Devi Boda
- Department of Dermatology, University of California, San Francisco, CA 94143
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, CA 94143;
| |
Collapse
|
66
|
Noubade R, Majri-Morrison S, Tarbell KV. Beyond cDC1: Emerging Roles of DC Crosstalk in Cancer Immunity. Front Immunol 2019; 10:1014. [PMID: 31143179 PMCID: PMC6521804 DOI: 10.3389/fimmu.2019.01014] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/23/2019] [Indexed: 01/03/2023] Open
Abstract
Dendritic cells (DCs) efficiently process and present antigens to T cells, and by integrating environmental signals, link innate and adaptive immunity. DCs also control the balance between tolerance and immunity, and are required for T-cell mediated anti-tumor immunity. One subset of classical DCs, cDC1, are particularly important for eliciting CD8 T cells that can kill tumor cells. cDC1s are superior in antigen cross-presentation, a process of presenting exogenous antigens on MHC class I to activate CD8+ T cells. Tumor-associated cDC1s can transport tumor antigen to the draining lymph node and cross-present tumor antigens, resulting in priming and activation of cytotoxic T cells. Although cross-presenting cDC1s are critical for eliciting anti-tumor T cell responses, the role and importance of other DC subsets in anti-tumor immunity is not as well-characterized. Recent literature in other contexts suggests that critical crosstalk between DC subsets can significantly alter biological outcomes, and these DC interactions likely also contribute significantly to tumor-specific immune responses. Therefore, antigen presentation by cDC1s may be necessary but not sufficient for maximal immune responses against cancer. Here, we discuss recent advances in the understanding of DC subset interactions to maximize anti-tumor immunity, and propose that such interactions should be considered for the development of better DC-targeted immunotherapies.
Collapse
Affiliation(s)
- Rajkumar Noubade
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Sonia Majri-Morrison
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Kristin V Tarbell
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
67
|
Schluck M, Hammink R, Figdor CG, Verdoes M, Weiden J. Biomaterial-Based Activation and Expansion of Tumor-Specific T Cells. Front Immunol 2019; 10:931. [PMID: 31130945 PMCID: PMC6509561 DOI: 10.3389/fimmu.2019.00931] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022] Open
Abstract
Traditional tumor vaccination approaches mostly focus on activating dendritic cells (DCs) by providing them with a source of tumor antigens and/or adjuvants, which in turn activate tumor-reactive T cells. Novel biomaterial-based cancer immunotherapeutic strategies focus on directly activating and stimulating T cells through molecular cues presented on synthetic constructs with the aim of improving T cell survival, more precisely steer T cell activation and direct T cell differentiation. Synthetic artificial antigen presenting cells (aAPCs) decorated with T cell-activating ligands are being developed to induce robust tumor-specific T cell responses, essentially bypassing DCs. In this perspective, we approach these promising new technologies from an immunological angle, first by identifying the CD4+ and CD8+ T cell subtypes that are imperative for robust anti-cancer immunity and subsequently discussing the molecular cues needed to induce these cells types. We will elaborate on how biomaterials can be applied to stimulate T cells in vitro and in vivo to improve their survival, activation and function. Scaffold-based methods can also be used as delivery vehicles for adoptive transfer of T cells, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor expressing (CAR) T cells, while simultaneously stimulating these cells. Finally, we provide suggestions on how these insights could advance the field of biomaterial-based activation and expansion of tumor-specific T cells in the future.
Collapse
Affiliation(s)
- Marjolein Schluck
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
| | - Roel Hammink
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Jorieke Weiden
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
68
|
Zaneti AB, Yamamoto MM, Sulczewski FB, Almeida BDS, Souza HFS, Ferreira NS, Maeda DLNF, Sales NS, Rosa DS, Ferreira LCDS, Boscardin SB. Dendritic Cell Targeting Using a DNA Vaccine Induces Specific Antibodies and CD4 + T Cells to the Dengue Virus Envelope Protein Domain III. Front Immunol 2019; 10:59. [PMID: 30761131 PMCID: PMC6362411 DOI: 10.3389/fimmu.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/10/2019] [Indexed: 01/18/2023] Open
Abstract
Dengue fever has become a global threat, causing millions of infections every year. An effective vaccine against all four serotypes of dengue virus (DENV) has not been developed yet. Among the different vaccination strategies available today, DNA vaccines are safe and practical, but currently induce relatively weak immune responses in humans. In order to improve immunogenicity, antigens may be targeted to dendritic cells (DCs), the main antigen presenting cells and orchestrators of the adaptive immune response, inducing T and B cell activation. It was previously shown that a DNA vaccine encoding a fusion protein comprised of an antigen and a single-chain Fv antibody (scFv) specific for the DC endocytic receptor DEC205 induced strong immune responses to the targeted antigen. In this work, we evaluate this strategy to improve the immunogenicity of dengue virus (DENV) proteins. Plasmids encoding the scFv αDEC205, or an isotype control (scFv ISO), fused to the DENV2 envelope protein domain III (EDIII) were generated, and EDIII specific immune responses were evaluated in immunized mice. BALB/c mice were intramuscularly (i.m.) immunized three times with plasmid DNAs encoding either scDEC-EDIII or scISO-EDIII followed by electroporation. Analyses of the antibody responses indicated that EDIII fusion with scFv targeting the DEC205 receptor significantly enhanced serum anti-EDIII IgG titers that inhibited DENV2 infection. Similarly, mice immunized with the scDEC-EDIII plasmid developed a robust CD4+ T cell response to the targeted antigen, allowing the identification of two linear epitopes recognized by the BALB/c haplotype. Taken together, these results indicate that targeting DENV2 EDIII protein to DCs using a DNA vaccine encoding the scFv αDEC205 improves both antibody and CD4+ T cell responses. This strategy opens perspectives for the use of DNA vaccines that encode antigens targeted to DCs as a strategy to increase immunogenicity.
Collapse
Affiliation(s)
- Arthur Baruel Zaneti
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcio Massao Yamamoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Bianca da Silva Almeida
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Higo Fernando Santos Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natália Soares Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Natiely Silva Sales
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil.,Institute for Investigation in Immunology (iii)-INCTiii, São Paulo, Brazil
| | | | - Silvia Beatriz Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii)-INCTiii, São Paulo, Brazil
| |
Collapse
|
69
|
Dhaeze T, Tremblay L, Lachance C, Peelen E, Zandee S, Grasmuck C, Bourbonnière L, Larouche S, Ayrignac X, Rébillard RM, Poirier J, Lahav B, Duquette P, Girard M, Moumdjian R, Bouthillier A, Larochelle C, Prat A. CD70 defines a subset of proinflammatory and CNS-pathogenic T H1/T H17 lymphocytes and is overexpressed in multiple sclerosis. Cell Mol Immunol 2019; 16:652-665. [PMID: 30635649 DOI: 10.1038/s41423-018-0198-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/16/2018] [Indexed: 12/12/2022] Open
Abstract
CD70 is the unique ligand of CD27 and is expressed on immune cells only upon activation. Therefore, engagement of the costimulatory CD27/CD70 pathway is solely dependent on upregulation of CD70. However, the T cell-intrinsic effect and function of human CD70 remain underexplored. Herein, we describe that CD70 expression distinguishes proinflammatory CD4+ T lymphocytes that display an increased potential to migrate into the central nervous system (CNS). Upregulation of CD70 on CD4+ T lymphocytes is induced by TGF-β1 and TGF-β3, which promote a pathogenic phenotype. In addition, CD70 is associated with a TH1 and TH17 profile of lymphocytes and is important for T-bet and IFN-γ expression by both T helper subtypes. Moreover, adoptive transfer of CD70-/-CD4+ T lymphocytes induced less severe experimental autoimmune encephalomyelitis (EAE) disease than transfer of WT CD4+ T lymphocytes. CD70+CD4+ T lymphocytes are found in the CNS during acute autoimmune inflammation in humans and mice, highlighting CD70 as both an immune marker and an important costimulator of highly pathogenic proinflammatory TH1/TH17 lymphocytes infiltrating the CNS.
Collapse
Affiliation(s)
- Tessa Dhaeze
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Laurence Tremblay
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Catherine Lachance
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Evelyn Peelen
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Stephanie Zandee
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Camille Grasmuck
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Lyne Bourbonnière
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Sandra Larouche
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Xavier Ayrignac
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | - Rose-Marie Rébillard
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada
| | - Josée Poirier
- Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | - Boaz Lahav
- Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | - Pierre Duquette
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | - Marc Girard
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | | | | | - Catherine Larochelle
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Centre de Recherche du CHUM (CRCHUM), Montréal, QC, H2X0A9, Canada. .,Multiple Sclerosis Clinic, Division of Neurology, CHUM, Montréal, QC, H2X0A9, Canada.
| |
Collapse
|
70
|
Riccione KA, He LZ, Fecci PE, Norberg PK, Suryadevara CM, Swartz A, Healy P, Reap E, Keler T, Li QJ, Congdon KL, Sanchez-Perez L, Sampson JH. CD27 stimulation unveils the efficacy of linked class I/II peptide vaccines in poorly immunogenic tumors by orchestrating a coordinated CD4/CD8 T cell response. Oncoimmunology 2018; 7:e1502904. [PMID: 30524899 PMCID: PMC6279317 DOI: 10.1080/2162402x.2018.1502904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
Despite their promise, tumor-specific peptide vaccines have limited efficacy. CD27 is a costimulatory molecule expressed on CD4+ and CD8+ T cells that is important in immune activation. Here we determine if a novel CD27 agonist antibody (αhCD27) can enhance the antitumor T cell response and efficacy of peptide vaccines. We evaluated the effects of αhCD27 on the immunogenicity and antitumor efficacy of whole protein, class I-restricted, and class II-restricted peptide vaccines using a transgenic mouse expressing human CD27. We found that αhCD27 preferentially enhances the CD8+ T cell response in the setting of vaccines comprised of linked class I and II ovalbumin epitopes (SIINFEKL and TEWTSSNVMEERKIKV, respectively) compared to a peptide vaccine comprised solely of SIINFEKL, resulting in the antitumor efficacy of adjuvant αhCD27 against intracranial B16.OVA tumors when combined with vaccines containing linked class I/II ovalbumin epitopes. Indeed, we demonstrate that this efficacy is both CD8- and CD4-dependent and αhCD27 activity on ovalbumin-specific CD4+ T cells is necessary for its adjuvant effect. Importantly for clinical translation, a linked universal CD4+ helper epitope (tetanus P30) was sufficient to instill the efficacy of SIINFEKL peptide combined with αhCD27, eliminating the need for a tumor-specific class II-restricted peptide. This approach unveiled the efficacy of a class I-restricted peptide vaccine derived from the tumor-associated Trp2 antigen in mice bearing intracranial B16 tumors. CD27 agonist antibodies combined with peptide vaccines containing linked tumor-specific CD8+ epitopes and tumor-specific or universal CD4+ epitopes enhance the efficacy of active cancer immunotherapy.
Collapse
Affiliation(s)
- Katherine A. Riccione
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | | | - Peter E. Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Pamela K. Norberg
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Carter M. Suryadevara
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Adam Swartz
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Patrick Healy
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Elizabeth Reap
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | | | - Qi-Jing Li
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Kendra L. Congdon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - John H. Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
71
|
Ng S, Bennett NJ, Schulze J, Gao N, Rademacher C, Derda R. Genetically-encoded fragment-based discovery of glycopeptide ligands for DC-SIGN. Bioorg Med Chem 2018; 26:5368-5377. [PMID: 30344001 DOI: 10.1016/j.bmc.2018.08.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/18/2018] [Accepted: 08/27/2018] [Indexed: 11/19/2022]
Abstract
We have employed genetically-encoded fragment-based discovery to identify novel glycopeptides with affinity for the dendritic cell receptor DC-SIGN. Starting from libraries of 108 mannose-conjugated peptides, we identified glycopeptides that exhibited up to a 650-fold increase in multivalent binding affinity for DC-SIGN, which is also preserved in cells. Monovalently, our most potent glycopeptides have a similar potency to a Man3 oligosaccharide, representing a 15-fold increase in activity compared to mannose. These compounds represent the first examples of glycopeptide ligands that target the CRD of DC-SIGN. The natural framework of glycopeptide conjugates and the simplicity of orthogonal conjugation to make these glycopeptides anticipates a promising future for development of DC-SIGN-targeting moieties.
Collapse
Affiliation(s)
- Simon Ng
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | | | - Jessica Schulze
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam 14424, Germany
| | - Nan Gao
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam 14424, Germany
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
72
|
Bathke B, Pätzold J, Kassub R, Giessel R, Lämmermann K, Hinterberger M, Brinkmann K, Chaplin P, Suter M, Hochrein H, Lauterbach H. CD70 encoded by modified vaccinia virus Ankara enhances CD8 T-cell-dependent protective immunity in MHC class II-deficient mice. Immunology 2018; 154:285-297. [PMID: 29281850 PMCID: PMC5980220 DOI: 10.1111/imm.12884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 12/13/2022] Open
Abstract
The immunological outcome of infections and vaccinations is largely determined during the initial first days in which antigen-presenting cells instruct T cells to expand and differentiate into effector and memory cells. Besides the essential stimulation of the T-cell receptor complex a plethora of co-stimulatory signals not only ensures a proper T-cell activation but also instils phenotypic and functional characteristics in the T cells appropriate to fight off the invading pathogen. The tumour necrosis factor receptor/ligand pair CD27/CD70 gained a lot of attention because of its key role in regulating T-cell activation, survival, differentiation and maintenance, especially in the course of viral infections and cancer. We sought to investigate the role of CD70 co-stimulation for immune responses induced by the vaccine vector modified vaccinia virus Ankara-Bavarian Nordic® (MVA-BN® ). Short-term blockade of CD70 diminished systemic CD8 T-cell effector and memory responses in mice. The dependence on CD70 became even more apparent in the lungs of MHC class II-deficient mice. Importantly, genetically encoded CD70 in MVA-BN® not only increased CD8 T-cell responses in wild-type mice but also substituted for CD4 T-cell help. MHC class II-deficient mice that were immunized with recombinant MVA-CD70 were fully protected against a lethal virus infection, whereas MVA-BN® -immunized mice failed to control the virus. These data are in line with CD70 playing an important role for vaccine-induced CD8 T-cell responses and prove the potency of integrating co-stimulatory molecules into the MVA-BN® backbone.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mark Suter
- Vetsuisse Fakultät, Dekanat, Bereich Immunologie, Universität Zürich, Zurich, Switzerland
| | | | | |
Collapse
|
73
|
Antonialli R, Sulczewski FB, Amorim KNDS, Almeida BDS, Ferreira NS, Yamamoto MM, Soares IS, Ferreira LCDS, Rosa DS, Boscardin SB. CpG Oligodeoxinucleotides and Flagellin Modulate the Immune Response to Antigens Targeted to CD8α + and CD8α - Conventional Dendritic Cell Subsets. Front Immunol 2017; 8:1727. [PMID: 29255470 PMCID: PMC5723008 DOI: 10.3389/fimmu.2017.01727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells essential for the induction of adaptive immune responses. Their unprecedented ability to present antigens to T cells has made them excellent targets for vaccine development. In the last years, a new technology based on antigen delivery directly to different DC subsets through the use of hybrid monoclonal antibodies (mAbs) to DC surface receptors fused to antigens of interest opened new perspectives for the induction of robust immune responses. Normally, the hybrid mAbs are administered with adjuvants that induce DC maturation. In this work, we targeted an antigen to the CD8α+ or the CD8α− DC subsets in the presence of CpG oligodeoxinucleotides (ODN) or bacterial flagellin, using hybrid αDEC205 or αDCIR2 mAbs, respectively. We also accessed the role of toll-like receptors (TLRs) 5 and 9 signaling in the induction of specific humoral and cellular immune responses. Wild-type and TLR5 or TLR9 knockout mice were immunized with two doses of the hybrid αDEC205 or αDCIR2 mAbs, as well as with an isotype control, together with CpG ODN 1826 or flagellin. A chimeric antigen containing the Plasmodium vivax 19 kDa portion of the merozoite surface protein (MSP119) linked to the Pan-allelic DR epitope was fused to each mAb. Specific CD4+ T cell proliferation, cytokine, and antibody production were analyzed. We found that CpG ODN 1826 or flagellin were able to induce CD4+ T cell proliferation, CD4+ T cells producing pro-inflammatory cytokines, and specific antibodies when the antigen was targeted to the CD8α+ DC subset. On the other hand, antigen targeting to CD8α− DC subset promoted specific antibody responses and proliferation, but no detectable pro-inflammatory CD4+ T cell responses. Also, specific antibody responses after antigen targeting to CD8α+ or CD8α− DCs were reduced in the absence of TLR9 or TLR5 signaling, while CD4+ T cell proliferation was mainly affected after antigen targeting to CD8α+ DCs and in the absence of TLR9 signaling. These results extend our understanding of the modulation of specific immune responses induced by antigen targeting to DCs in the presence of different adjuvants. Such knowledge may be useful for the optimization of DC-based vaccines.
Collapse
Affiliation(s)
- Renan Antonialli
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Bianca da Silva Almeida
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natália Soares Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Márcio Massao Yamamoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene Silva Soares
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| |
Collapse
|
74
|
Minoda Y, Virshup I, Leal Rojas I, Haigh O, Wong Y, Miles JJ, Wells CA, Radford KJ. Human CD141 + Dendritic Cell and CD1c + Dendritic Cell Undergo Concordant Early Genetic Programming after Activation in Humanized Mice In Vivo. Front Immunol 2017; 8:1419. [PMID: 29163495 PMCID: PMC5670352 DOI: 10.3389/fimmu.2017.01419] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/12/2017] [Indexed: 12/24/2022] Open
Abstract
Human immune cell subsets develop in immunodeficient mice following reconstitution with human CD34+ hematopoietic stem cells. These "humanized" mice are useful models to study human immunology and human-tropic infections, autoimmunity, and cancer. However, some human immune cell subsets are unable to fully develop or acquire full functional capacity due to a lack of cross-reactivity of many growth factors and cytokines between species. Conventional dendritic cells (cDCs) in mice are categorized into cDC1, which mediate T helper (Th)1 and CD8+ T cell responses, and cDC2, which mediate Th2 and Th17 responses. The likely human equivalents are CD141+ DC and CD1c+ DC subsets for mouse cDC1 and cDC2, respectively, but the extent of any interspecies differences is poorly characterized. Here, we exploit the fact that human CD141+ DC and CD1c+ DC develop in humanized mice, to further explore their equivalency in vivo. Global transcriptome analysis of CD141+ DC and CD1c+ DC isolated from humanized mice demonstrated that they closely resemble those in human blood. Activation of DC subsets in vivo, with the TLR3 ligand poly I:C, and the TLR7/8 ligand R848 revealed that a core panel of genes consistent with DC maturation status were upregulated by both subsets. R848 specifically upregulated genes associated with Th17 responses by CD1c+ DC, while poly I:C upregulated IFN-λ genes specifically by CD141+ DC. MYCL expression, known to be essential for CD8+ T cell priming by mouse DC, was specifically induced in CD141+ DC after activation. Concomitantly, CD141+ DC were superior to CD1c+ DC in their ability to prime naïve antigen-specific CD8+ T cells. Thus, CD141+ DC and CD1c+ DC share a similar activation profiles in vivo but also have induce unique signatures that support specialized roles in CD8+ T cell priming and Th17 responses, respectively. In combination, these data demonstrate that humanized mice provide an attractive and tractable model to study human DC in vitro and in vivo.
Collapse
Affiliation(s)
- Yoshihito Minoda
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Isaac Virshup
- The Centre for Stem Cell Systems, Anatomy and Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Ingrid Leal Rojas
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Oscar Haigh
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Yide Wong
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Cairns, QLD, Australia
| | - John J Miles
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Cairns, QLD, Australia
| | - Christine A Wells
- The Centre for Stem Cell Systems, Anatomy and Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia.,Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Kristen J Radford
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
75
|
Galati D, Zanotta S. Hematologic neoplasms: Dendritic cells vaccines in motion. Clin Immunol 2017; 183:181-190. [PMID: 28870867 DOI: 10.1016/j.clim.2017.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/28/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) are bone-marrow-derived immune cells accounted for a key role in cancer vaccination as potent antigen-presenting cells within the immune system. Cancer microenvironment can modulate DCs maturation resulting in their accumulation into functional states associated with a reduced antitumor immune response. In this regard, a successful cancer vaccine needs to mount a potent antitumor immune response able to overcome the immunosuppressive tumor milieu. As a consequence, DCs-based approaches are a safe and promising strategy for improving the therapeutic efficacy in hematological malignancies, particularly in combinations with additional treatments. This review summarizes the most significant evidence about the immunotherapeutic strategies performed to target hematologic neoplasms including the tumoral associated antigens (TAA) pulsed on DCs, whole tumor cell vaccines or leukemia-derived DCs.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy.
| | - Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy
| |
Collapse
|
76
|
Abstract
Adjuvants have been deliberately added to vaccines since the 1920's when alum was discovered to boost antibody responses, leading to better protection. The first adjuvants were discovered by accident and were used in the safer but less immunogenic subunit vaccines, supposedly by providing an antigen depot to extend antigen presentation. Since that time, much has been discovered about how these adjuvants impact cells at the tissue site to activate innate immune responses, mobilize dendritic cells and drive adaptive immunity. New approaches to vaccine construction for infectious diseases that have so far not been well addressed by conventional vaccines often attempt to induce antibodies, polyfunctional CD4+ T cells and CD8+ CTLs. The discovery of pattern recognition receptors and ligands that drive desired T cell responses has led to development of novel adjuvant strategies using immunomodulatory agents to direct appropriate immune responses.
Collapse
Affiliation(s)
- Amy S McKee
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Philippa Marrack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, 1400, Jackson St., Denver, CO 80206, USA
| |
Collapse
|
77
|
Hahn AW, Gill DM, Pal SK, Agarwal N. The future of immune checkpoint cancer therapy after PD-1 and CTLA-4. Immunotherapy 2017; 9:681-692. [DOI: 10.2217/imt-2017-0024] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Andrew W Hahn
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84112 USA
| | - David M Gill
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84112 USA
| | - Sumanta K Pal
- Department of Oncology, City of Hope Cancer Center, Duarte, CA, 91010 USA
| | - Neeraj Agarwal
- Department of Oncology, Huntsman Cancer Institute, Salt Lake City, UT, 84112 USA
| |
Collapse
|
78
|
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: 60] [Impact Index Per Article: 7.5] [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
|
79
|
Lu E, Dang EV, McDonald JG, Cyster JG. Distinct oxysterol requirements for positioning naïve and activated dendritic cells in the spleen. Sci Immunol 2017; 2:2/10/eaal5237. [PMID: 28738017 DOI: 10.1126/sciimmunol.aal5237] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022]
Abstract
Correct positioning of dendritic cells (DCs) is critical for efficient pathogen encounter and antigen presentation. Epstein-Barr virus-induced gene 2 (EBI2) has been identified as a chemoattractant receptor required for naïve CD4+DCIR2+ DC positioning in response to 7α,25-hydroxycholesterol (7α,25-HC). We now provide evidence that a second EBI2 ligand, 7α,27-HC, is involved in splenic DCIR2+ DC positioning and homeostasis. Cyp27a1, the enzyme uniquely required for 7α,27-HC synthesis, is expressed by stromal cells in the region of naïve DC localization. After activation, DCIR2+ DCs move into the T cell zone. We find that EBI2 is rapidly up-regulated in DCIR2+ DCs under certain activation conditions, and positioning at the B-T zone interface depends on EBI2. Under conditions of type I interferon induction, EBI2 ligand levels are elevated, causing activated DCIR2+ DCs to disperse throughout the T zone. Last, we provide evidence that oxysterol metabolism by Batf3-dependent DCs is important for EBI2-dependent positioning of activated DCIR2+ DCs. This work indicates that 7α,27-HC functions as a guidance cue in vivo and reveals a multitiered role for EBI2 in DC positioning. Deficiency in this organizing system results in defective CD4+ T cell responses.
Collapse
Affiliation(s)
- Erick Lu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric V Dang
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
80
|
Li Y, Liu X, Duan W, Tian H, Zhu G, He H, Yao S, Yi S, Song W, Tang H. Batf3-dependent CD8α + Dendritic Cells Aggravates Atherosclerosis via Th1 Cell Induction and Enhanced CCL5 Expression in Plaque Macrophages. EBioMedicine 2017; 18:188-198. [PMID: 28411140 PMCID: PMC5405198 DOI: 10.1016/j.ebiom.2017.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 03/24/2017] [Accepted: 04/04/2017] [Indexed: 01/18/2023] Open
Abstract
Dendritic cells (DCs) play an important role in controlling T cell-mediated adaptive immunity in atherogenesis. However, the role of the basic leucine zipper transcription factor, ATF-like 3 (Batf3)-dependent CD8α+ DC subset in atherogenesis remains unclear. Here we show that Batf3-/-Apoe-/- mice, lacking CD8α+ DCs, exhibited a significant reduction in atherogenesis and T help 1 (Th1) cells compared with Apoe-/- controls. Then, we found that CD8α+ DCs preferentially induce Th1 cells via secreting interleukin-12 (IL-12), and that the expression of interferon-gamma (IFN-γ)or chemokine (C-C motif) ligand 5 (CCL5) in aorta were significantly decreased in Batf3-/-Apoe-/- mice. We further demonstrated that macrophages were the major CCL5-expressing cells in the plaque, which was significantly reduced in Batf3-/-Apoe-/- mice. Furthermore, we found CCL5 expression in macrophages was promoted by IFN-γ. Finally, we showed that Batf3-/-Apoe-/- mice displayed decreased infiltration of leukocytes in the plaque. Thus, CD8α+ DCs aggravated atherosclerosis, likely by inducing Th1 cell response, which promoted CCL5 expression in macrophages and increased infiltration of leukocytes and lesion inflammation.
Collapse
Affiliation(s)
- Yalin Li
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Xueyan Liu
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Wei Duan
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Shandong, China
| | - Guangming Zhu
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Hao He
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Shandong, China
| | - Shuying Yi
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Wengang Song
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China.
| | - Hua Tang
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China.
| |
Collapse
|
81
|
Bullock TN. Stimulating CD27 to quantitatively and qualitatively shape adaptive immunity to cancer. Curr Opin Immunol 2017; 45:82-88. [PMID: 28319731 DOI: 10.1016/j.coi.2017.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/09/2017] [Accepted: 02/16/2017] [Indexed: 12/22/2022]
Abstract
The capacity of the immune system to recognize and respond to tumors has been appreciated for over 100 years. However, clinical success has largely depended on the elucidation of the positive and negative regulators of effector cells after their activation via the antigen cell receptor. On the one hand, effector cells upregulate checkpoint molecules that are thought to play a role in limiting immunopathology. On the other, second and third waves of costimulation are often required to promote the expansion, survival and differentiation of effector cells. While it is clear that the immune system can be unleashed by blocking checkpoint molecules, this approach is most effective when pre-existing responses exist in patients' tumors. Thus, coordinating checkpoint blockade with costimulation could potentially expand the patient population that receives benefit from cancer immunotherapy. This review will discuss how the costimulatory molecule CD27 sculpts immunity and preclinical/clinical data indicating its potential for cancer immunotherapy and its clinical translation.
Collapse
Affiliation(s)
- Timothy Nj Bullock
- Department of Pathology and Human Immune Therapy Center, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
82
|
Apostólico JDS, Lunardelli VAS, Yamamoto MM, Souza HFS, Cunha-Neto E, Boscardin SB, Rosa DS. Dendritic Cell Targeting Effectively Boosts T Cell Responses Elicited by an HIV Multiepitope DNA Vaccine. Front Immunol 2017; 8:101. [PMID: 28223987 PMCID: PMC5295143 DOI: 10.3389/fimmu.2017.00101] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/20/2017] [Indexed: 11/13/2022] Open
Abstract
Despite several efforts in the last decades, an efficacious HIV-1 vaccine is still not available. Different approaches have been evaluated, such as recombinant proteins, viral vectors, DNA vaccines, and, most recently, dendritic cell (DC) targeting. This strategy is based on DC features that place them as central for induction of immunity. Targeting is accomplished by the use of chimeric monoclonal antibodies directed to DC surface receptors fused to the antigen of interest. In this work, we targeted eight promiscuous HIV-derived CD4+ T cell epitopes (HIVBr8) to the DEC205+ DCs by fusing the multiepitope immunogen to the heavy chain of αDEC205 (αDECHIVBr8), in the presence of the TLR3 agonist poly (I:C). In addition, we tested a DNA vaccine encoding the same epitopes using homologous or heterologous prime-boost regimens. Our results showed that mice immunized with αDECHIVBr8 presented higher CD4+ and CD8+ T cell responses when compared to mice that received the DNA vaccine (pVAXHIVBr8). In addition, pVAXHIVBr8 priming followed by αDECHIVBr8 boosting induced higher polyfunctional proliferative and cytokine-producing T cell responses to HIV-1 peptides than homologous DNA immunization or heterologous αDEC prime/DNA boost. Based on these results, we conclude that homologous prime-boost and heterologous boosting immunization strategies targeting CD4+ epitopes to DCs are effective to improve HIV-specific cellular immune responses when compared to standalone DNA immunization. Moreover, our results indicate that antigen targeting to DC is an efficient strategy to boost immunity against a multiepitope immunogen, especially in the context of DNA vaccination.
Collapse
Affiliation(s)
- Juliana de Souza Apostólico
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil; Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | | | - Marcio Massao Yamamoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - Higo Fernando Santos Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - Edecio Cunha-Neto
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil; Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil; Laboratory of Immunology, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil; Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil; Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| |
Collapse
|
83
|
Waisman A, Lukas D, Clausen BE, Yogev N. Dendritic cells as gatekeepers of tolerance. Semin Immunopathol 2017; 39:153-163. [PMID: 27456849 DOI: 10.1007/s00281-016-0583-z] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DC) are unique hematopoietic cells, linking innate and adaptive immune responses. In particular, they are considered as the most potent antigen presenting cells, governing both T cell immunity and tolerance. In view of their exceptional ability to present antigen and to interact with T cells, DC play distinct roles in shaping T cell development, differentiation and function. The outcome of the DC-T cell interaction is determined by the state of DC maturation, the type of DC subset, the cytokine microenvironment and the tissue location. Both regulatory T cells (Tregs) and DC are indispensable for maintaining central and peripheral tolerance. Over the past decade, accumulating data indicate that DC critically contribute to Treg differentiation and homeostasis.
Collapse
Affiliation(s)
- Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Dominika Lukas
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Microbiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
84
|
Lymphocyte Antigen 75 Polymorphisms Are Associated with Disease Susceptibility and Phenotype in Japanese Patients with Inflammatory Bowel Disease. DISEASE MARKERS 2016; 2016:6485343. [PMID: 27965521 PMCID: PMC5124651 DOI: 10.1155/2016/6485343] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/20/2016] [Indexed: 12/22/2022]
Abstract
Recent genome-wide association studies have rapidly improved our understanding of the molecular pathways leading to inflammatory bowel disease (IBD), which includes Crohn's disease (CD) and ulcerative colitis (UC). Although several reports have demonstrated that gene single nucleotide polymorphisms (SNPs) are associated with susceptibility to IBD, its precise genetic factors have not been fully clarified. Here, we performed an association analysis between lymphocyte antigen 75 (LY75) genetic variations and IBD susceptibility or phenotype. SNPs were genotyped in 51 CD patients, 94 UC patients, and 269 healthy controls of Japanese ethnicity. We detected a significant relationship with CD susceptibility for the rs16822581 LY75 SNP (P = 0.045). One haplotype (GT, P = 0.042) was also associated with CD susceptibility, while another carrying the opposite SNP (CA) was linked to an absence of surgical history for CD. Our findings confirm that LY75 is involved in CD susceptibility and may play a role in disease activity in the Japanese population.
Collapse
|
85
|
Tuettenberg A, Steinbrink K, Schuppan D. Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy. Nanomedicine (Lond) 2016; 11:2735-2751. [DOI: 10.2217/nnm-2016-0208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Macrophages, myeloid-derived suppressor cells and tolerogenic dendritic cells are central players of a heterogeneous myeloid cell population, with the ability to suppress innate and adaptive immune responses and thus to promote tumor growth. Their influx and local proliferation are mainly induced by the cancers themselves, and their numbers in the tumor microenvironment and the peripheral blood correlate with decreased survival. Therapeutic targeting these innate immune cells, either aiming at their elimination or polarization toward tumor suppressive cells is an attractive novel approach to control tumor progression and block metastasis. We review the current understanding of cancer immunology including immune surveillance and immune editing in the context of these prominent innate suppressor cells, and their targetability by nanoparticular immunotherapy with small molecules or siRNA.
Collapse
Affiliation(s)
- Andrea Tuettenberg
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology & Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
86
|
Abstract
Cancer heterogeneity, a hallmark enabling clonal survival and therapy resistance, is shaped by active immune responses. Antigen-specific T cells can control cancer, as revealed clinically by immunotherapeutics such as adoptive T-cell transfer and checkpoint blockade. The host immune system is thus a powerful tool that, if better harnessed, could significantly enhance the efficacy of cytotoxic therapy and improve outcomes for cancer sufferers. To realize this vision, however, a number of research frontiers must be tackled. These include developing strategies for neutralizing tumor-promoting inflammation, broadening T-cell repertoires (via vaccination), and elucidating the mechanisms by which immune cells organize tumor microenvironments to regulate T-cell activity. Such efforts will pave the way for identifying new targets for combination therapies that overcome resistance to current treatments and promote long-term cancer control.
Collapse
|
87
|
Abstract
Dendritic cells (DCs) lie at the heart of the innate immune system, specialised at recognising danger signals in many forms including foreign material, infection or tissue damage and initiating powerful adaptive immune and inflammatory responses. In barrier sites such as the lung, the instrumental role that DCs play at the interface between the environment and the host places them in a pivotal position in determining the severity of inflammatory disease. The past few years has seen a significant increase in our fundamental understanding of the subsets of DCs involved in pulmonary immunity, as well as the mechanisms by which they are activated and which they may use to coordinate downstream inflammation and pathology. In this review, we will summarise current understanding of the multi-faceted role that DCs play in the induction, maintenance and regulation of lung immunopathology, with an emphasis on allergic pulmonary disease.
Collapse
|
88
|
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
|
89
|
Affiliation(s)
- Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
90
|
Pennock ND, Kedl JD, Kedl RM. T Cell Vaccinology: Beyond the Reflection of Infectious Responses. Trends Immunol 2016; 37:170-180. [PMID: 26830540 PMCID: PMC4775298 DOI: 10.1016/j.it.2016.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 12/31/2015] [Accepted: 01/07/2016] [Indexed: 12/17/2022]
Abstract
Inducing sustained, robust CD8(+) T cell responses is necessary for therapeutic intervention in chronic infectious diseases and cancer. Unfortunately, most adjuvant formulations fail to induce substantial cellular immunity in humans. Attenuated acute infectious agents induce strong CD8(+) T cell immunity, and are thought to therefore represent a good road map for guiding the development of subunit vaccines capable of inducing the same. However, recent evidence suggests that this assumption may need reconsideration. Here we provide an overview of subunit vaccine history as it pertains to instigating T cell responses. We argue that in light of evidence demonstrating that T cell responses to vaccination differ from those induced by infectious challenge, research in pursuit of cellular immunity-inducing vaccine adjuvants should no longer follow only the infection paradigm.
Collapse
Affiliation(s)
- Nathan D Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Justin D Kedl
- Department of Immunology and Microbiology, University of Colorado Denver, Denver, CO, USA
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Denver, Denver, CO, USA.
| |
Collapse
|
91
|
Lutz MB. Induction of CD4(+) Regulatory and Polarized Effector/helper T Cells by Dendritic Cells. Immune Netw 2016; 16:13-25. [PMID: 26937228 PMCID: PMC4770096 DOI: 10.4110/in.2016.16.1.13] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are considered to play major roles during the induction of T cell immune responses as well as the maintenance of T cell tolerance. Naive CD4(+) T cells have been shown to respond with high plasticity to signals inducing their polarization into effector/helper or regulatory T cells. Data obtained from in vitro generated bone-marrow (BM)-derived DCs as well as genetic mouse models revealed an important but not exclusive role of DCs in shaping CD4(+) T cell responses. Besides the specialization of some conventional DC subsets for the induction of polarized immunity, also the maturation stage, activation of specialized transcription factors and the cytokine production of DCs have major impact on CD4(+) T cells. Since in vitro generated BM-DCs show a high diversity to shape CD4(+) T cells and their high similarity to monocyte-derived DCs in vivo, this review reports data mainly on BM-DCs in this process and only touches the roles of transcription factors or of DC subsets, which have been discussed elsewhere. Here, recent findings on 1) the conversion of naive into anergic and further into Foxp3(-) regulatory T cells (Treg) by immature DCs, 2) the role of RelB in steady state migratory DCs (ssmDCs) for conversion of naive T cells into Foxp3(+) Treg, 3) the DC maturation signature for polarized Th2 cell induction and 4) the DC source of IL-12 for Th1 induction are discussed.
Collapse
Affiliation(s)
- Manfred B Lutz
- Institute of Virology and Immunobiology, University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
92
|
Murakami R, Nakagawa Y, Shimizu M, Wakabayashi A, Negishi Y, Hiroi T, Okubo K, Takahashi H. Effects of Dendritic Cell Subset Manipulation on Airway Allergy in a Mouse Model. Int Arch Allergy Immunol 2016; 168:219-32. [PMID: 26855055 DOI: 10.1159/000443237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 12/09/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Two major distinct subsets of dendritic cells (DCs) are arranged to regulate immune responses: DEC-205+ DCs drive Th1 polarization and 33D1+ DCs establish Th2 dominancy. Th1 polarization can be achieved either by depletion of 33D1+ DCs with a 33D1-specific monoclonal antibody (mAb) or by activation of DEC-205+ DCs via intraperitoneal injection of α-galactosylceramide (α-GalCer). We studied the effect of 33D1+ DC depletion or DEC-205+ DC activation in vivo using an established mouse model of allergic rhinitis (AR). METHODS Mice were injected intraperitoneally with OVA plus alum and challenged 4 times with daily intranasal administration of OVA. Immediately after the last challenge, allergic symptoms such as sneezing and nasal rubbing as well as the number of cells in the bronchoalveolar lavage fluid (BALF) and nasal lavage fluid (NALF) were counted. The levels of serum OVA-specific IgG1, IgG2a, and IgE were also determined by ELISA. RESULTS The allergic symptom scores were significantly decreased in 33D1+ DC-depleted or DEC-205+ DC-activated AR mice. The levels of OVA-specific IgG1, IgG2a, and IgE, and the number of NALF cells, but not BALF cells, were reduced in 33D1+ DC-depleted but not in DEC-205+ DC-activated AR mice. Moreover, the activated DEC-205+ DCs suppressed histamine release from IgE-sensitized mast cells, probably through IL-12 secretion. CONCLUSIONS The manipulation of innate DC subsets may provide a new therapeutic strategy for controlling various allergic diseases by reducing histamine release from IgE-sensitized mast cells by driving the immune response towards Th1 dominancy via activation of DEC-205+ DCs in vivo.
Collapse
Affiliation(s)
- Ryosuke Murakami
- Departments of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
93
|
The CD27–CD70 pathway and pathogenesis of autoimmune disease. Semin Arthritis Rheum 2016; 45:496-501. [DOI: 10.1016/j.semarthrit.2015.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/27/2015] [Accepted: 08/05/2015] [Indexed: 11/19/2022]
|
94
|
Wadwa M, Klopfleisch R, Buer J, Westendorf AM. Targeting Antigens to Dec-205 on Dendritic Cells Induces Immune Protection in Experimental Colitis in Mice. Eur J Microbiol Immunol (Bp) 2016; 6:1-8. [PMID: 27141310 PMCID: PMC4838981 DOI: 10.1556/1886.2015.00048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022] Open
Abstract
The endocytotic c-type lectin receptor DEC-205 is highly expressed on immature dendritic cells. In previous studies, it was shown that antigen-targeting to DEC-205 is a useful tool for the induction of antigen-specific Foxp3+ regulatory T cells and thereby can prevent inflammatory processes. However, whether this approach is sufficient to mediate tolerance in mucosal tissues like the gut is unknown. In this study, we established a new mouse model in which the adoptive transfer of naive hemagglutinin (HA)-specific CD4+Foxp3– T cells into VILLIN-HA transgenic mice leads to severe colitis. To analyze if antigen-targeting to DEC-205 could protect against inflammation of the gut, VILLIN-HA transgenic mice were injected with an antibody–antigen complex consisting of the immunogenic HA110–120 peptide coupled to an α-DEC-205 antibody (DEC-HA) before adoptive T cell transfer. DEC-HA-treated mice showed significantly less signs of intestinal inflammation as was demonstrated by reduced loss of body weight and histopathology in the gut. Strikingly, abrogated intestinal inflammation was mediated via the conversion of naive HA-specific CD4+Foxp3– T cells into HA-specific CD4+Foxp3+ regulatory T cells. In this study, we provide evidence that antigen-targeting to DEC-205 can be utilized for the induction of tolerance in mucosal organs that are confronted with large numbers of exogenous antigens.
Collapse
Affiliation(s)
- Munisch Wadwa
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg Essen , Essen, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin , Berlin, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg Essen , Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg Essen , Essen, Germany
| |
Collapse
|
95
|
Cauwelaert ND, Desbien AL, Hudson TE, Pine SO, Reed SG, Coler RN, Orr MT. The TLR4 Agonist Vaccine Adjuvant, GLA-SE, Requires Canonical and Atypical Mechanisms of Action for TH1 Induction. PLoS One 2016; 11:e0146372. [PMID: 26731269 PMCID: PMC4701231 DOI: 10.1371/journal.pone.0146372] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
The Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) promotes strong TH1 and balanced IgG1/IgG2 responses to protein vaccine antigens. This enhanced immunity is sufficient to provide protection against many diseases including tuberculosis and leishmaniasis. To better characterize the adjuvant action it is important to understand how the different cytokines and transcription factors contribute to the initiation of immunity. In the present study using T-bet-/- and IL-12-/- mice and a blocking anti-IFNαR1 monoclonal antibody, we define mechanisms of adjuvant activity of GLA-SE. In accordance with previous studies of TLR4 agonist based adjuvants, we found that TH1 induction via GLA-SE was completely dependent upon T-bet, a key transcription factor for IFNγ production and TH1 differentiation. Consistent with this, deficiency of IL-12, a cytokine canonical to TH1 induction, ablated TH1 induction via GLA-SE. Finally we demonstrate that the innate immune response to GLA-SE, including rapid IFNγ production by memory CD8+ T cells and NK cells, was contingent on type I interferon, a cytokine group whose association with TH1 induction is contextual, and that they contributed to the adjuvant activity of GLA-SE.
Collapse
Affiliation(s)
| | - Anthony L. Desbien
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Thomas E. Hudson
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Samuel O. Pine
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Mark T. Orr
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
96
|
Greyer M, Whitney P, Stock A, Davey G, Tebartz C, Bachem A, Mintern J, Strugnell R, Turner S, Gebhardt T, O’Keeffe M, Heath W, Bedoui S. T Cell Help Amplifies Innate Signals in CD8 + DCs for Optimal CD8 + T Cell Priming. Cell Rep 2016; 14:586-597. [DOI: 10.1016/j.celrep.2015.12.058] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 11/02/2015] [Accepted: 12/10/2015] [Indexed: 12/29/2022] Open
|
97
|
van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy 2015; 7:655-67. [DOI: 10.2217/imt.15.32] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
98
|
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]
|
99
|
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
|
100
|
Flacher V, Tripp CH, Mairhofer DG, Steinman RM, Stoitzner P, Idoyaga J, Romani N. Murine Langerin+ dermal dendritic cells prime CD8+ T cells while Langerhans cells induce cross-tolerance. EMBO Mol Med 2015; 6:1191-204. [PMID: 25085878 PMCID: PMC4197865 DOI: 10.15252/emmm.201303283] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Skin dendritic cells (DCs) control the immunogenicity of cutaneously administered vaccines. Antigens targeted to DCs via the C-type lectin Langerin/CD207 are cross-presented to CD8+ T cells in vivo. We investigated the relative roles of Langerhans cells (LCs) and Langerin+ dermal DCs (dDCs) in different vaccination settings. Poly(I:C) and anti-CD40 agonist antibody promoted cytotoxic responses upon intradermal immunization with ovalbumin (OVA)-coupled anti-Langerin antibodies (Langerin/OVA). This correlated with CD70 upregulation in Langerin+ dDCs, but not LCs. In chimeric mice where Langerin targeting was restricted to dDCs, CD8+ T-cell memory was enhanced. Conversely, providing Langerin/OVA exclusively to LCs failed to prime cytotoxicity, despite initial antigen cross-presentation to CD8+ T cells. Langerin/OVA combined with imiquimod could not prime CD8+ T cells and resulted in poor cytotoxicity in subsequent responses. This tolerance induction required targeting and maturation of LCs. Altogether, Langerin+ dDCs prime long-lasting cytotoxic responses, while cross-presentation by LCs negatively influences CD8+ T-cell priming. Moreover, this highlights that DCs exposed to TLR agonists can still induce tolerance and supports the existence of qualitatively different DC maturation programs.
Collapse
Affiliation(s)
- Vincent Flacher
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - David G Mairhofer
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY, USA
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Juliana Idoyaga
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY, USA
| | - Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| |
Collapse
|