51
|
Plasmacytoid dendritic cells cross-prime naive CD8 T cells by transferring antigen to conventional dendritic cells through exosomes. Proc Natl Acad Sci U S A 2020; 117:23730-23741. [PMID: 32879009 DOI: 10.1073/pnas.2002345117] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although plasmacytoid dendritic cells (pDCs) have been shown to play a critical role in generating viral immunity and promoting tolerance to suppress antitumor immunity, whether and how pDCs cross-prime CD8 T cells in vivo remain controversial. Using a pDC-targeted vaccine model to deliver antigens specifically to pDCs, we have demonstrated that pDC-targeted vaccination led to strong cross-priming and durable CD8 T cell immunity. Surprisingly, cross-presenting pDCs required conventional DCs (cDCs) to achieve cross-priming in vivo by transferring antigens to cDCs. Taking advantage of an in vitro system where only pDCs had access to antigens, we further demonstrated that cross-presenting pDCs were unable to efficiently prime CD8 T cells by themselves, but conferred antigen-naive cDCs the capability of cross-priming CD8 T cells by transferring antigens to cDCs. Although both cDC1s and cDC2s exhibited similar efficiency in acquiring antigens from pDCs, cDC1s but not cDC2s were required for cross-priming upon pDC-targeted vaccination, suggesting that cDC1s played a critical role in pDC-mediated cross-priming independent of their function in antigen presentation. Antigen transfer from pDCs to cDCs was mediated by previously unreported pDC-derived exosomes (pDCexos), that were also produced by pDCs under various conditions. Importantly, all these pDCexos primed naive antigen-specific CD8 T cells only in the presence of bystander cDCs, similarly to cross-presenting pDCs, thus identifying pDCexo-mediated antigen transfer to cDCs as a mechanism for pDCs to achieve cross-priming. In summary, our data suggest that pDCs employ a unique mechanism of pDCexo-mediated antigen transfer to cDCs for cross-priming.
Collapse
|
52
|
Sukswai N, Jung HR, Amr SS, Ng SB, Sheikh SS, Lyapichev K, El Hussein S, Loghavi S, Agbay RLMC, Miranda RN, Medeiros LJ, Khoury JD. Immunopathology of Kikuchi-Fujimoto disease: A reappraisal using novel immunohistochemistry markers. Histopathology 2020; 77:262-274. [PMID: 31854007 DOI: 10.1111/his.14050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/10/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
AIMS Kikuchi-Fujimoto disease (KFD) is a self-limited disease characterised by destruction of the lymph node parenchyma. Few studies have assessed the immunohistological features of KFD, and most employed limited antibody panels that lacked many of the novel immunohistochemistry markers currently available. METHODS AND RESULTS We used immunohistochemistry to reappraise the microanatomical distribution of plasmacytoid dendritic cells (pDCs), follicular helper T cells and cytotoxic T cells, B cells, follicular dendritic cell (FDC) meshworks, and histiocytes in lymph nodes involved by KFD. The study group consisted of 138 KFD patients (89 women; 64.5%) with a median age of 27 years (range, 3-50 years). Cervical lymph nodes were most commonly involved, in 108 (78.3%) patients. The numbers of pDCs were increased, predominantly around and within apoptotic areas and the paracortex, and tapering off within xanthomatous areas. pDCs formed sizeable tight clusters, most notably around apoptotic/necrotic areas. T cells consisted mostly of CD8-positive cells with predominant expression of T-cell receptor-β. There were notable increases in the numbers of CD8-positive T cells within lymphoid follicles, and their numbers correlated with alterations in FDC meshworks (P < 0.001). The number of follicular helper T cells was decreased within distorted FDC meshworks. CD21 highlighted frequent distortion of FDC meshworks, even in lymph node tissue that was distant from apoptotic/necrotic areas. Distorted FDC meshworks spanned all morphological patterns, and FDC meshwork characteristics (intact; distorted; remnant/nearly absent) correlated with morphological patterns (P < 0.01). CONCLUSIONS The immunohistological landscape of KFD is complex and characterised by increased numbers of pDCs that frequently cluster around apoptotic/necrotic foci, increased numbers of cytotoxic T cells, and substantial distortion of FDC meshworks.
Collapse
Affiliation(s)
- Narittee Sukswai
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
- Department of Pathology, Chulalongkorn University, Bangkok, Thailand
| | - Hye Ra Jung
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
- Department of Pathology, Keimyung University, Dongsan Medical Center, Seoul, South Korea
| | - Samir S Amr
- Department of Pathology, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
| | - Siok Bian Ng
- Department of Pathology, National University Hospital, Singapore
| | - Salwa S Sheikh
- Department of Pathology, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
| | - Kirill Lyapichev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| | - Siba El Hussein
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| | - Rose Lou Marie C Agbay
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
- Department of Pathology, The Medical City Hospital, Manila, Philippines
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| | - Joseph D Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TV, USA
| |
Collapse
|
53
|
Chen HJ, Tas SW, de Winther MPJ. Type-I interferons in atherosclerosis. J Exp Med 2020; 217:132613. [PMID: 31821440 PMCID: PMC7037237 DOI: 10.1084/jem.20190459] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Chen et al. review the effects of type-I IFNs and the potential of anti–type-I IFN therapies in atherosclerosis. The contribution of dyslipidemia and inflammation in atherosclerosis is well established. Along with effective lipid-lowering treatments, the recent success of clinical trials with anti-inflammatory therapies and the accelerated atherosclerosis in many autoimmune diseases suggest that targeting inflammation may open new avenues for the prevention and the treatment for cardiovascular diseases (CVDs). In the past decades, studies have widened the role of type-I interferons (IFNs) in disease, from antivirus defense to autoimmune responses and immuno-metabolic syndromes. While elevated type-I IFN level in serum is associated with CVD incidence in patients with interferonopathies, experimental data have attested that type-I IFNs affect plaque-residing macrophages, potentiate foam cell and extracellular trap formation, induce endothelial dysfunction, alter the phenotypes of dendritic cells and T and B lymphocytes, and lead to exacerbated atherosclerosis outcomes. In this review, we discuss the production and the effects of type-I IFNs in different atherosclerosis-associated cell types from molecular biology studies, animal models, and clinical observations, and the potential of new therapies against type-I IFN signaling for atherosclerosis.
Collapse
Affiliation(s)
- Hung-Jen Chen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
54
|
Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, Chen J. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res 2020; 159:104980. [PMID: 32504832 DOI: 10.1016/j.phrs.2020.104980] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs), as specialized antigen-presenting cells, are essential for the initiation of specific T cell responses in innate antitumor immunity and, in certain cases, support humoral responses to inhibit tumor development. Mounting evidence suggests that the DC system displays a broad spectrum of dysfunctional status in the tumor microenvironment (TME), which ultimately affects antitumor immune responses. DC-based therapy can restore the function of DCs in the TME, thus showing a promising potential in tumor therapy. In this review, we provide an overview of the DC deficiency caused by various factors in the TME and discuss proposed strategies to reverse DC deficiency and the applications of novel combinatorial DC-based therapy for immune normalization of the tumor.
Collapse
Affiliation(s)
- Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wan Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | | | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
55
|
Fang Y, Zhang X, Lu C, Yin Y, Hu X, Xu W, Liu Y, Wang H. Cytosolic mtDNA released from pneumolysin-damaged mitochondria triggers IFN-β production in epithelial cells. Can J Microbiol 2020; 66:435-445. [PMID: 32191844 DOI: 10.1139/cjm-2019-0481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pneumolysin (Ply) is a major virulence factor of Streptococcus pneumoniae. Ply-induced interferon-β (IFN-β) expression in host macrophages has been shown to be due to the accumulation of mitochondrial deoxyribonucleic acid (mtDNA) in the cytoplasm during S. pneumoniae infection. Our findings extend this work to show human bronchial epithelial cells that reside at the interface of inflammatory injury, BEAS-2B, adapt to local cues by altering mitochondrial states and releasing excess mtDNA. The results in this research showed that purified Ply induced the expression of IFN-β in human epithelial cells, which was accompanied by mitochondrial damage both in vivo and in vitro. The observations also were supported by the increased mtDNA concentrations in the bronchial lavage fluid of mice infected with S. pneumoniae. In summary, our study demonstrated that Ply triggered the production of IFN-β in epithelial cells, and this response was mediated by mtDNA released from Ply-damaged mitochondria. It displayed an impressive modulation of IFN-β response to S. pneumoniae in epithelial cells.
Collapse
Affiliation(s)
- Yuting Fang
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuemei Zhang
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chang Lu
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yibing Yin
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuexue Hu
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Wenchun Xu
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yusi Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Hong Wang
- School of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|
56
|
Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma. Cells 2020; 9:E417. [PMID: 32054102 PMCID: PMC7072514 DOI: 10.3390/cells9020417] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Francesca Consoli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology, University of Brescia at ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
57
|
Li T, Wu J, Zhu S, Zang G, Li S, Lv X, Yue W, Qiao Y, Cui J, Shao Y, Zhang J, Liu YJ, Chen J. A Novel C Type CpG Oligodeoxynucleotide Exhibits Immunostimulatory Activity In Vitro and Enhances Antitumor Effect In Vivo. Front Pharmacol 2020; 11:8. [PMID: 32116691 PMCID: PMC7015978 DOI: 10.3389/fphar.2020.00008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 01/03/2020] [Indexed: 11/13/2022] Open
Abstract
Background C type CpG oligodeoxynucleotides (CpG-C ODNs), possessing the features of both A type and B type CpG ODNs, exert a variety of immunostimulatory activities and have been demonstrated as an effective antitumor immunotherapy. Based on the structural characteristics, we designed 20 potential ODNs with the aim of synthesizing an optimal, novel CpG-C ODN specific to human and murine Toll-like receptor 9 (TLR9). We also sought to investigate the in vitro immunostimulatory and in vivo antitumor effects of the novel CpG-C ODN. Methods Twenty potential CpG-C ODNs were screened for their ability to secrete interferon (IFN)-α, and interleukin (IL)-6 and tumor necrosis factor (TNF)-α production for the three most promising sequences were assayed in human peripheral blood mononuclear cells (PBMCs) by enzyme-linked immunosorbent assay (ELISA) or cytometric bead array assay. The functions of human and mouse B cells, and cytokine production in mice induced by the most promising sequence, HP06T07, were determined by flow cytometry and ELISA. Growth and morphology of tumor tissues in in vivo murine models inoculated with CT26 cells were analyzed by a growth inhibition assay and immunohistochemistry, respectively. Results Among the 20 designed ODNs, HP06T07 significantly induced IFN-α, IL-6, and TNF-α secretion, and promoted B-cell activation and proliferation in a dose-dependent manner in human PBMCs and mouse splenocytes in vitro. Intratumoral injection of HP06T07 notably suppressed tumor growth and prolonged survival in the CT26 subcutaneous mouse model in a dose-dependent manner. HP06T07 administered nine times at 2-day intervals (I2) eradicated tumor growth at both primary and distant sites of CT26 tumors. HP06T07 restrained tumor growth by increasing the infiltration of T cells, NK cells, and plasmacytoid dendritic cells (pDCs). Conclusions HP06T07, a novel CpG-C ODN, shows potent immunostimulatory activity in vitro and suppresses tumor growth in the CT26 subcutaneous mouse model.
Collapse
Affiliation(s)
- Tete Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Guoxia Zang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shuang Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xinping Lv
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wenjun Yue
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yuan Qiao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yan Shao
- Changchun Huapu Biotechnology Co., Ltd., Changchun, China
| | - Jun Zhang
- Changchun Huapu Biotechnology Co., Ltd., Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
58
|
Zeng S, Xiao Z, Wang Q, Guo Y, He Y, Zhu Q, Zou Y. Strategies to achieve immune tolerance in allogeneic solid organ transplantation. Transpl Immunol 2020; 58:101250. [DOI: 10.1016/j.trim.2019.101250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/19/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
|
59
|
Chen C, Chencheng Z, Cuiying L, Xiaokun G. Plasmacytoid Dendritic Cells Protect Against Middle Cerebral Artery Occlusion Induced Brain Injury by Priming Regulatory T Cells. Front Cell Neurosci 2020; 14:8. [PMID: 32076400 PMCID: PMC7006436 DOI: 10.3389/fncel.2020.00008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) play an anti-inflammatory effect to protect against ischemic stroke. Plasmacytoid dendritic cells (pDCs) can induce regulatory T cells tolerance in sterile-inflammation conditions. However, whether and how pDCs-mediated Tregs response play a part in the pathology of ischemic stroke remains unclear. In this study, we showed that pDCs were increased in the brain of middle cerebral artery occlusion (MCAO) mice. Depletion of pDCs with 120G8 exacerbated MCAO-induced brain injury, peripheral pro-inflammation response and decreased the systemic Tregs in mice. Furthermore, the data of mixed lymphocyte reaction (MLR) in vitro demonstrate that splenic pDCs from MCAO mice can significantly promote Tregs proliferation, accompanying with the increased expression of indoleamine 2,3-dioxygenase 1 (IDO1) on pDCs. Taken together, the findings here suggested that under the pathologic state of stroke, pDCs protect against MCAO-induced brain injury by priming Tregs, illustrating that pDCs represented as a therapeutic target for the prevention of ischemic brain injury.
Collapse
Affiliation(s)
- Chen Chen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhang Chencheng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liu Cuiying
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Geng Xiaokun
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
60
|
Liu J, Hu Y, Guo Q, Yu X, Shao L, Zhang C. Enhanced Anti-melanoma Efficacy of a Pim-3-Targeting Bifunctional Small Hairpin RNA via Single-Stranded RNA-Mediated Activation of Plasmacytoid Dendritic Cells. Front Immunol 2019; 10:2721. [PMID: 31849942 PMCID: PMC6902031 DOI: 10.3389/fimmu.2019.02721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/06/2019] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the most serious type of skin cancer. The immunosuppressive tumor microenvironment and aberrant expression of some proto-oncogenes are the main cause of melanoma development. We have constructed a single-stranded RNA (ssRNA)–Pim-3–small hairpin RNA (shRNA) dual-function vector, which activates the toll-like receptor (TLR)7 to stimulate the antitumor immune response through ssRNA fragments and simultaneously silences the proto-oncogene Pim-3 to intensify apoptosis of the tumor cells via shRNA. Here, we found that therapy with the ssRNA-Pim-3-shRNA dual-function vector not only promotes the apoptosis and inhibits the proliferation of B16F10 melanoma cells by inhibiting the expression of Pim-3 but also enhances the activation of CD8+ T cells and natural killer (NK) cells and simultaneously reduces the proportion of intratumoral regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Together, these features effectively inhibit the growth of melanoma. Intriguingly, the bifunctional therapeutic effect that reverses the tumor immunosuppressive microenvironment is dependent on the activation of plasmacytoid dendritic cells (pDCs) and the secretion of type I interferon (IFN). Our study suggests that ssRNA-Pim-3-shRNA dual-function therapy is expected to become a promising therapeutic strategy for melanoma and other solid tumors with immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuan Hu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Qie Guo
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Xin Yu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Liwei Shao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Cai Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
61
|
Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:1-68. [PMID: 31810551 DOI: 10.1016/bs.ircmb.2019.07.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are a unique class of immune cells that act as a bridge between innate and adaptive immunity. The discovery of DCs by Cohen and Steinman in 1973 laid the foundation for DC biology, and the advances in the field identified different versions of DCs with unique properties and functions. DCs originate from hematopoietic stem cells, and their differentiation is modulated by Flt3L. They are professional antigen-presenting cells that patrol the environmental interphase, sites of infection, or infiltrate pathological tissues looking for antigens that can be used to activate effector cells. DCs are critical for the initiation of the cellular and humoral immune response and protection from infectious diseases or tumors. DCs can take up antigens using specialized surface receptors such as endocytosis receptors, phagocytosis receptors, and C type lectin receptors. Moreover, DCs are equipped with an array of extracellular and intracellular pattern recognition receptors for sensing different danger signals. Upon sensing the danger signals, DCs get activated, upregulate costimulatory molecules, produce various cytokines and chemokines, take up antigen and process it and migrate to lymph nodes where they present antigens to both CD8 and CD4 T cells. DCs are classified into different subsets based on an integrated approach considering their surface phenotype, expression of unique and conserved molecules, ontogeny, and functions. They can be broadly classified as conventional DCs consisting of two subsets (DC1 and DC2), plasmacytoid DCs, inflammatory DCs, and Langerhans cells.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Mansi Saxena
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
62
|
Lee AY, Körner H. The CCR6-CCL20 axis in humoral immunity and T-B cell immunobiology. Immunobiology 2019; 224:449-454. [DOI: 10.1016/j.imbio.2019.01.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023]
|
63
|
pDC Activation by TLR7/8 Ligand CL097 Compared to TLR7 Ligand IMQ or TLR9 Ligand CpG. J Immunol Res 2019; 2019:1749803. [PMID: 31093508 PMCID: PMC6481147 DOI: 10.1155/2019/1749803] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/19/2019] [Accepted: 02/24/2019] [Indexed: 02/07/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) express high levels of the toll-like receptors (TLRs) TLR7 and TLR9. In response to TLR7 and TLR9 ligands, pDCs are primary producers of type I interferons. Our previous study demonstrated that pDCs activated by the TLR7 ligand imiquimod (IMQ) and the TLR9 ligand CpG A can kill breast cancer cells in vitro and inhibit tumor growth in vivo. Moreover, we observed a distinctive morphological, phenotypic change in pDCs after activation by IMQ and CpG A. However, the effect of other TLR7 and TLR9 ligands on pDCs remains less understood. In this study, we treat pDCs with the TLR7 ligand IMQ, TLR7/8 ligands (CL097 and CL075), and three TLR9 ligands (different types of CpGs). The size of pDCs increased significantly after activation by TLR7, or TLR7/8 ligands. TLR7, TLR7/8, and TLR9 ligands similarly modulated cytokine release, as well as protein expression of pDC markers, costimulatory molecules, and cytotoxic molecules. Interestingly, TLR7/8 ligands, especially CL097, induced stronger responses. These results are relevant to the further study of the role and mechanism of pDC-induced antitumor effects and may aid in the development of a new strategy for future tumor immunotherapy.
Collapse
|
64
|
Fu C, Jiang A. Dendritic Cells and CD8 T Cell Immunity in Tumor Microenvironment. Front Immunol 2018; 9:3059. [PMID: 30619378 PMCID: PMC6306491 DOI: 10.3389/fimmu.2018.03059] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) play a central role in the regulation of the balance between CD8 T cell immunity vs. tolerance to tumor antigens. Cross-priming, a process which DCs activate CD8 T cells by cross-presenting exogenous antigens, plays a critical role in generating anti-tumor CD8 T cell immunity. However, there are compelling evidences now that the tumor microenvironment (TME)-mediated suppression and modulation of tumor-infiltrated DCs (TIDCs) impair their function in initiating potent anti-tumor immunity and even promote tumor progression. Thus, DC-mediated cross-presentation of tumor antigens in tumor-bearing hosts often induces T cell tolerance instead of immunity. As tumor-induced immunosuppression remains one of the major hurdles for cancer immunotherapy, understanding how DCs regulate anti-tumor CD8 T cell immunity in particular within TME has been under intensive investigation. Recent reports on the Batf3-dependent type 1 conventional DCs (cDC1s) in anti-tumor immunity have greatly advanced our understanding on the interplay of DCs and CD8 T cells in the TME, highlighted by the critical role of CD103+ cDC1s in the cross-priming of tumor antigen-specific CD8 T cells. In this review, we will discuss recent advances in anti-tumor CD8 T cell cross-priming by CD103+ cDC1s in TME, and share perspective on future directions including therapeutic applications and memory CD8 T cell responses.
Collapse
Affiliation(s)
- Chunmei Fu
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Aimin Jiang
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
65
|
Klymenko Y, Nephew KP. Epigenetic Crosstalk between the Tumor Microenvironment and Ovarian Cancer Cells: A Therapeutic Road Less Traveled. Cancers (Basel) 2018; 10:E295. [PMID: 30200265 PMCID: PMC6162502 DOI: 10.3390/cancers10090295] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022] Open
Abstract
Metastatic dissemination of epithelial ovarian cancer (EOC) predominantly occurs through direct cell shedding from the primary tumor into the intra-abdominal cavity that is filled with malignant ascitic effusions. Facilitated by the fluid flow, cells distribute throughout the cavity, broadly seed and invade through peritoneal lining, and resume secondary tumor growth in abdominal and pelvic organs. At all steps of this unique metastatic process, cancer cells exist within a multidimensional tumor microenvironment consisting of intraperitoneally residing cancer-reprogramed fibroblasts, adipose, immune, mesenchymal stem, mesothelial, and vascular cells that exert miscellaneous bioactive molecules into malignant ascites and contribute to EOC progression and metastasis via distinct molecular mechanisms and epigenetic dysregulation. This review outlines basic epigenetic mechanisms, including DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA regulators, and summarizes current knowledge on reciprocal interactions between each participant of the EOC cellular milieu and tumor cells in the context of aberrant epigenetic crosstalk. Promising research directions and potential therapeutic strategies that may encompass epigenetic tailoring as a component of complex EOC treatment are discussed.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Cell, Molecular and Cancer Biology Program, Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA.
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46617, USA.
| | - Kenneth P Nephew
- Cell, Molecular and Cancer Biology Program, Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA.
- Department of Cellular and Integrative Physiology and Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
66
|
Toll-like receptor 9 ligands increase type I interferon induced B-cell activating factor expression in chronic rhinosinusitis with nasal polyposis. Clin Immunol 2018; 197:19-26. [PMID: 30056130 DOI: 10.1016/j.clim.2018.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 07/10/2018] [Accepted: 07/25/2018] [Indexed: 11/23/2022]
Abstract
B-cell activating factor (BAFF) has been proposed to play a crucial role in the pathogenesis of chronic rhinosinusitis with nasal polyp (CRSwNP). The aim of this study was to evaluate the role of toll-like receptor (TLR) 9-mediated BAFF activation on the pathogenesis of CRSwNP. NP and uncinate tissue (UT) were obtained from patients with CRSwNP or CRS without NP, and control subjects. The expression of TLR9, high mobility group box-1 protein (HMGB1), type I interferon (IFN), BAFF, and anti-double stranded DNA (dsDNA) antibody were examined in the tissues and the cultured dispersed NP cells (DNPCs). The expression of TLR9, HMGB1, type I IFN, BAFF, and anti-dsDNA antibody were elevated in NP tissue compared to the UTs. Exposure to TLR9 agonist increased the type I IFN expression in vitro, which further increased BAFF production. In conclusion, we provided a novel therapeutic potential of TLR9 agonist in CRSwNP.
Collapse
|