1
|
Yuan B, Wang Y, Chu Y, Yuan X. Do the monocyte-derived dendritic cells exert a pivotal role in the early onset of experimental autoimmune uveitis? BMC Ophthalmol 2025; 25:165. [PMID: 40175949 PMCID: PMC11963667 DOI: 10.1186/s12886-025-04014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Eyes are recognized as immunological privileged site. However, the onset of autoimmune uveitis (AU) prompts an influx of dendritic cells (DCs) into the retinas, tasked with presenting auto-antigens, thereby exacerbating the inflammatory response. Monocyte-derived DCs (moDCs) implicated in various autoimmune disorders, but their specific involvement in AU remains unclear. This study aims to investigate the constitution and dynamics of retinal DCs subsequent to the induction of experimental autoimmune uveitis (EAU). METHODS In our study, an EAU model was established in C57BL/6J mice, and prednisolone acetate (PA) eye drops were administrated unilaterally to the right eye from 5 days post-immunization (dpi). The infiltration of Gr-1+CD115+CD11c-MHC-II- cells (monocytes), Gr-1+CD115+CD11c+MHC-II+ cells (moDCs) and Gr-1-CD115-CD11c+MHC-II+ cells (conventional dendritic cells, cDCs) within retina were detected by flow cytometry and immunofluorescence stain at 7, 10, 13, and 16 dpi. Additionally, the protein expression and mRNA expression of pivotal cytokines associated with moDCs and inflammation were analysed by western blotting and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. RESULTS Our findings unveiled a notable rise in moDCs infiltration and differentiation from 7 to 13 dpi. The administration of PA eye drops did not yield a significant variance in either the quantity or the differentiation rate of moDCs. Throughout the initial stages of EAU, the expression of GM-CSF remained consistent, while TGF-β1 exhibited a sustained increase until 13 dpi in the control group and until 10 dpi following PA treatment. Anti-inflammatory cytokines Il-10 and Il-4 displayed no significant increase until 16 dpi after PA administration. CONCLUSIONS Our results indicate that moDCs exhibited an earlier and more substantial infiltration into the inflamed retina compared to cDCs. This heightened presence of moDCs appeared to play a dominant role in the presentation of auto-antigens during the initial stages of EAU, consequently contributing to the exaggerated autoimmune response within the ocular milieu. The administration of PA exhibited no discernible impact on either the differentiation or the infiltration of moDCs.
Collapse
Affiliation(s)
- Bo Yuan
- School of Medicine, Nankai University, Tianjin, China
- Aier Eye Hospital, Tianjin University, Tianjin, China
| | - Yajie Wang
- Tianjin Eye Hospital, Tianjin, China
- Tianjin key Lab of Ophthalmology and Visual Science, Tianjin, China
| | - Yanhua Chu
- Tianjin Eye Hospital, Tianjin, China
- Tianjin key Lab of Ophthalmology and Visual Science, Tianjin, China
| | - Xiaoyong Yuan
- School of Medicine, Nankai University, Tianjin, China.
- Tianjin Eye Hospital, Tianjin, China.
- Tianjin key Lab of Ophthalmology and Visual Science, Tianjin, China.
| |
Collapse
|
2
|
Shen M, Li Z, Wang J, Xiang H, Xie Q. Traditional Chinese herbal medicine: harnessing dendritic cells for anti-tumor benefits. Front Immunol 2024; 15:1408474. [PMID: 39364399 PMCID: PMC11446781 DOI: 10.3389/fimmu.2024.1408474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Chinese Herbal Medicine (CHM) is being more and more used in cancer treatment because of its ability to regulate the immune system. Chinese Herbal Medicine has several advantages over other treatment options, including being multi-component, multi-target, and having fewer side effects. Dendritic cells (DCs) are specialized antigen presenting cells that play a vital part in connecting the innate and adaptive immune systems. They are also important in immunotherapy. Recent evidence suggests that Chinese Herbal Medicine and its components can positively impact the immune response by targeting key functions of dendritic cells. In this review, we have summarized the influences of Chinese Herbal Medicine on the immunobiological feature of dendritic cells, emphasized an anti-tumor effect of CHM-treated DCs, and also pointed out deficiencies in the regulation of DC function by Chinese Herbal Medicine and outlined future research directions.
Collapse
Affiliation(s)
- Mengyi Shen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhen Li
- School of Preventive Medicine Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jing Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Hongjie Xiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
3
|
Mildner A, Kim KW, Yona S. Unravelling monocyte functions: from the guardians of health to the regulators of disease. DISCOVERY IMMUNOLOGY 2024; 3:kyae014. [PMID: 39430099 PMCID: PMC11486918 DOI: 10.1093/discim/kyae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 10/22/2024]
Abstract
Monocytes are a key component of the innate immune system. They undergo intricate developmental processes within the bone marrow, leading to diverse monocyte subsets in the circulation. In a state of healthy homeostasis, monocytes are continuously released into the bloodstream, destined to repopulate specific tissue-resident macrophage pools where they fulfil tissue-specific functions. However, under pathological conditions monocytes adopt various phenotypes to resolve inflammation and return to a healthy physiological state. This review explores the nuanced developmental pathways and functional roles that monocytes perform, shedding light on their significance in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Alexander Mildner
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, 20014 Turku, Finland
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Simon Yona
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
4
|
Dotta E, Maciola AK, Baccega T, Pasqual G. Dendritic cells steering antigen and leukocyte traffic in lymph nodes. FEBS Lett 2024. [PMID: 38997244 DOI: 10.1002/1873-3468.14982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Dendritic cells (DCs) play a central role in initiating and shaping the adaptive immune response, thanks to their ability to uptake antigens and present them to T cells. Once in the lymph node (LN), DCs can spread the antigen to other DCs, expanding the pool of cells capable of activating specific T-cell clones. Additionally, DCs can modulate the dynamics of other immune cells, by increasing naïve T-cell dwell time, thereby facilitating the scanning for cognate antigens, and by selectively recruiting other leukocytes. Here we discuss the role of DCs in orchestrating antigen and leukocyte trafficking within the LN, together with the implications of this trafficking on T-cell activation and commitment to effector function.
Collapse
Affiliation(s)
- Enrico Dotta
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Agnieszka Katarzyna Maciola
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Tania Baccega
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
5
|
Salvadé V, Manuel O, Golshayan D, Obregon C. Monocyte-derived dendritic cells can be detected in urine of kidney transplant recipients with pathogenic asymptomatic bacteriuria. FRONTIERS IN TRANSPLANTATION 2024; 3:1366104. [PMID: 38993772 PMCID: PMC11235355 DOI: 10.3389/frtra.2024.1366104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/27/2024] [Indexed: 07/13/2024]
Abstract
Urinary tract infections (UTI) are an important clinical problem in kidney transplant recipients (KTR). Asymptomatic bacteriuria (ASB) is frequent in these patients and often resolved by the immune system, but a significant proportion may progress to complicated UTI, which may compromise allograft function and survival. It is essential to determine the involvement of the immune system in the infectious process. Dendritic cells (DCs) are recognised as playing a pivotal role in initiating inflammatory responses capable of priming antigen-specific T cells, a crucial step in determining the fate of local inflammation. Little is known about their role in the control of UTI. In this brief communication, we report an incidental finding in a group of 16 stable KTR in which monocyte-derived dendritic cells (ModDCs), analysed by flow cytometry, were found in urine of patients with ASB and high bacterial counts >107 cfu/ml. Within this group, one patient developed pyelonephritis in the following days. These findings suggest that the immune system, in particular DCs, may be recruited during the course of a UTI and, to our knowledge, present for the first time evidence that inflammatory ModDCs can be detected in urine. Their frequency may reflect the degree of infection. This finding suggests the potential for exploring whether these cells may be useful in distinguishing between pathogenic ASB and those that can be resolved by the immune system.
Collapse
Affiliation(s)
- Vanja Salvadé
- Transplantation Centre, Departments of Medicine and Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Oriol Manuel
- Transplantation Centre, Departments of Medicine and Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Centre, Departments of Medicine and Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Carolina Obregon
- Transplantation Centre, Departments of Medicine and Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Woman-Mother-Child Department, Pediatric Service, Pediatric Nephrology Laboratory, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
6
|
Jiménez-Cortegana C, Palomares F, Alba G, Santa-María C, de la Cruz-Merino L, Sánchez-Margalet V, López-Enríquez S. Dendritic cells: the yin and yang in disease progression. Front Immunol 2024; 14:1321051. [PMID: 38239364 PMCID: PMC10794555 DOI: 10.3389/fimmu.2023.1321051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Dendritic cells (DCs) are antigen presenting cells that link innate and adaptive immunity. DCs have been historically considered as the most effective and potent cell population to capture, process and present antigens to activate naïve T cells and originate favorable immune responses in many diseases, such as cancer. However, in the last decades, it has been observed that DCs not only promote beneficial responses, but also drive the initiation and progression of some pathologies, including inflammatory bowel disease (IBD). In line with those notions, different therapeutic approaches have been tested to enhance or impair the concentration and role of the different DC subsets. The blockade of inhibitory pathways to promote DCs or DC-based vaccines have been successfully assessed in cancer, whereas the targeting of DCs to inhibit their functionality has proved to be favorable in IBD. In this review, we (a) described the general role of DCs, (b) explained the DC subsets and their role in immunogenicity, (c) analyzed the role of DCs in cancer and therapeutic approaches to promote immunogenic DCs and (d) analyzed the role of DCs in IBD and therapeutic approaches to reduced DC-induced inflammation. Therefore, we aimed to highlight the "yin-yang" role of DCs to improve the understand of this type of cells in disease progression.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Francisca Palomares
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Consuelo Santa-María
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, Seville, Spain
| | - Luis de la Cruz-Merino
- Clinical Oncology Dept. Medicine Department, University of Seville, Virgen Macarena University Hospital, Seville, Spain
| | - Victor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Soledad López-Enríquez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
7
|
Mahajan D, Kumar T, Rath PK, Sahoo AK, Mishra BP, Kumar S, Nayak NR, Jena MK. Dendritic Cells and the Establishment of Fetomaternal Tolerance for Successful Human Pregnancy. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0010. [PMID: 38782369 DOI: 10.2478/aite-2024-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 05/25/2024]
Abstract
Pregnancy is a remarkable event where the semi-allogeneic fetus develops in the mother's uterus, despite genetic and immunological differences. The antigen handling and processing at the maternal-fetal interface during pregnancy appear to be crucial for the adaptation of the maternal immune system and for tolerance to the developing fetus and placenta. Maternal antigen-presenting cells (APCs), such as macrophages (Mφs) and dendritic cells (DCs), are present at the maternal-fetal interface throughout pregnancy and are believed to play a crucial role in this process. Despite numerous studies focusing on the significance of Mφs, there is limited knowledge regarding the contribution of DCs in fetomaternal tolerance during pregnancy, making it a relatively new and growing field of research. This review focuses on how the behavior of DCs at the maternal-fetal interface adapts to pregnancy's unique demands. Moreover, it discusses how DCs interact with other cells in the decidual leukocyte network to regulate uterine and placental homeostasis and the local maternal immune responses to the fetus. The review particularly examines the different cell lineages of DCs with specific surface markers, which have not been critically reviewed in previous publications. Additionally, it emphasizes the impact that even minor disruptions in DC functions can have on pregnancy-related complications and proposes further research into the potential therapeutic benefits of targeting DCs to manage these complications.
Collapse
Affiliation(s)
- Deviyani Mahajan
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Tarun Kumar
- Department of Veterinary Clinical Complex, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125001, India
| | - Prasana Kumar Rath
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Anjan Kumar Sahoo
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
- Department of Veterinary Surgery and Radiology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Bidyut Prava Mishra
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
- Department of Livestock Products Technology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Sudarshan Kumar
- Proteomics and Structural Biology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Nihar Ranjan Nayak
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| |
Collapse
|
8
|
Feng F, Li Z, Xie Q, Song W. Phenotypic and functional differences of dendritic cells in tumor. J Cancer Res Ther 2023; 19:1509-1516. [PMID: 38156916 DOI: 10.4103/jcrt.jcrt_2383_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024]
Abstract
Dendritic cells (DCs) are a unique class of immune cells vital to the immune system, functioning as antigen-presenting cells that play a key role in launching both cellular and humoral immune responses. They are crucial in preventing infectious diseases and regulating tumor growth. DCs can be categorized based on various criteria such as phenotype, function, and tissue location, resulting in several subgroups. Generally, DCs are divided into two primary groups: plasmacytoid DCs (pDCs) and conventional DCs (cDCs), which are further classified into Type I classical DCs (cDC1) and Type II classical DCs (cDC2). cDC1 cells are distinguishable by specific gene programs and associated markers, while cDC2 cells display more diversity. Moreover, there is an ongoing debate surrounding a recently identified subgroup called DC3, and whether it can be considered a distinct cell type in the maturation process of DCs remains uncertain. Most of these DC subgroups rely on the growth factor Fms-like tyrosine kinase 3 ligand (FLT3L) for differentiation from a common DC precursor (CDP), guided by various cytokines. Although the general classification of DC subgroups is similar in both humans and mice, numerous phenotypic and functional variations exist within each subgroup. Therefore, comprehending these differences between DC subgroups in humans and mice holds the potential to significantly advance relevant research.
Collapse
Affiliation(s)
- Fengtian Feng
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhen Li
- School of Preventive Medicine Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qi Xie
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wengang Song
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
9
|
Du X, Li M, Huan C, Lv G. Dendritic cells in liver transplantation immune response. Front Cell Dev Biol 2023; 11:1277743. [PMID: 37900282 PMCID: PMC10606587 DOI: 10.3389/fcell.2023.1277743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Dendritic cells (DCs) are the most powerful antigen presenting cells (APCs), they are considered one of the key regulatory factors in the liver immune system. There is currently much interest in modulating DC function to improve transplant immune response. In liver transplantation, DCs participate in both the promotion and inhibition of the alloreponse by adopting different phenotypes and function. Thus, in this review, we discussed the origin, maturation, migration and pathological effects of several DC subsets, including the conventional DC (cDC), plasmacytoid DC (pDC) and monocyte-derived DC (Mo-DC) in liver transplantation, and we summarized the roles of these DC subsets in liver transplant rejection and tolerance. In addition, we also outlined the latest progress in DC-based related treatment regimens. Overall, our discussion provides a beneficial resource for better understanding the biology of DCs and their manipulation to improve the immune adaptability of patients in transplant status.
Collapse
Affiliation(s)
- Xiaodong Du
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Chen Huan
- Center of Infectious Diseases and Pathogen Biology, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Wang Z, Zhang J, An F, Zhang J, Meng X, Liu S, Xia R, Wang G, Yan C. The mechanism of dendritic cell-T cell crosstalk in rheumatoid arthritis. Arthritis Res Ther 2023; 25:193. [PMID: 37798668 PMCID: PMC10552435 DOI: 10.1186/s13075-023-03159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterised by joint pain and swelling, synovial hyperplasia, cartilage damage, and bone destruction. The mechanisms of dendritic cell (DC) and T cell-mediated crosstalk have gradually become a focus of attention. DCs regulate the proliferation and differentiation of CD4+ T cell subtypes through different cytokines, surface molecules, and antigen presentation. DC-T cell crosstalk also blocks antigen presentation by DCs, ultimately maintaining immune tolerance. DC-T cell crosstalk mainly involves chemokines, surface molecules (TonEBP, NFATc1), the PD-L1/PD-1 signalling axis, and the TGF-β signalling axis. In addition, DC-T cell crosstalk in RA is affected by glycolysis, reactive oxygen species, vitamin D, and other factors. These factors lead to the formation of an extremely complex regulatory network involving various mechanisms. This article reviews the key immune targets of DC-T cell crosstalk and elucidates the mechanism of DC-T cell crosstalk in RA to provide a basis for the treatment of patients with RA.
Collapse
Affiliation(s)
- Zhandong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Jinlong Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Xiangrui Meng
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Shiqing Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Ruoliu Xia
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Gang Wang
- Rheumatism and Orthopaedics Department, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
11
|
Abascal J, Oh MS, Liclican EL, Dubinett SM, Salehi-Rad R, Liu B. Dendritic Cell Vaccination in Non-Small Cell Lung Cancer: Remodeling the Tumor Immune Microenvironment. Cells 2023; 12:2404. [PMID: 37830618 PMCID: PMC10571973 DOI: 10.3390/cells12192404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains one of the leading causes of death worldwide. While NSCLCs possess antigens that can potentially elicit T cell responses, defective tumor antigen presentation and T cell activation hinder host anti-tumor immune responses. The NSCLC tumor microenvironment (TME) is composed of cellular and soluble mediators that can promote or combat tumor growth. The composition of the TME plays a critical role in promoting tumorigenesis and dictating anti-tumor immune responses to immunotherapy. Dendritic cells (DCs) are critical immune cells that activate anti-tumor T cell responses and sustain effector responses. DC vaccination is a promising cellular immunotherapy that has the potential to facilitate anti-tumor immune responses and transform the composition of the NSCLC TME via tumor antigen presentation and cell-cell communication. Here, we will review the features of the NSCLC TME with an emphasis on the immune cell phenotypes that directly interact with DCs. Additionally, we will summarize the major preclinical and clinical approaches for DC vaccine generation and examine how effective DC vaccination can transform the NSCLC TME toward a state of sustained anti-tumor immune signaling.
Collapse
Affiliation(s)
- Jensen Abascal
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Michael S. Oh
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Elvira L. Liclican
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Steven M. Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095-1690, USA
| | - Ramin Salehi-Rad
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Bin Liu
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| |
Collapse
|
12
|
Liu JC, Wang P, Zeng QX, Yang C, Lyu M, Li Y, Yeung WSB, Chiu PCN, Haidl G, Allam JP, Duan YG. Myd88 Signaling Is Involved in the Inflammatory Response in LPS-Induced Mouse Epididymitis and Bone-Marrow-Derived Dendritic Cells. Int J Mol Sci 2023; 24:ijms24097838. [PMID: 37175545 PMCID: PMC10178089 DOI: 10.3390/ijms24097838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/30/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Epididymitis is an epididymal inflammation that may lead to male infertility. Dendritic cells (DCs) and myeloid differentiation primary response gene 88 (Myd88) were associated with epididymitis in rodents. However, the functions of Myd88 on epididymal DCs remain unclear. This study investigated the role of Myd88 in DCs for epididymitis. The Myd88 signaling pathway, phenotypes of DC subsets, and cytokines were investigated in lipopolysaccharide (LPS)-induced epididymitis in mice. CRISPR-Cas9 was used to knockout Myd88 in bone-marrow-derived dendritic cells (BMDCs) and immortalized mouse epididymal (DC2) cell line. In the vivo experiments, levels of the proinflammatory cytokines IL-1α, IL-6, IL-17A, TNF-α, IL-1β, MCP-1, and GM-CSF, mRNA for MyD88 related genes, and the percentages of monocyte-derived DCs (Mo-DCs) were significantly elevated in mice with epididymitis. In the vitro experiments, LPS significantly promoted the apoptosis of BMDCs. In addition, the concentration of inflammatory cytokines in BMDCs and DC2s were increased in the LPS group, while decreasing after the knockout of Myd88. These findings indicate that Myd88 on DCs is involved in the inflammation of epididymitis in mice, which may be a potential target for better strategies regarding the treatment of immunological male infertility.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Peng Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qun-Xiong Zeng
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chen Yang
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Minmin Lyu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yanfeng Li
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - William Shu-Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Philip Chi-Ngong Chiu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Gerhard Haidl
- Department of Andrology, Bonn University Hospital, Campus-Venusberg 1, 53127 Bonn, Germany
| | - Jean-Pierre Allam
- Department of Andrology, Bonn University Hospital, Campus-Venusberg 1, 53127 Bonn, Germany
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
13
|
Yao CL, Tseng TY. The synergistic and enhancive effects of IL-6 and M-CSF to expand and differentiate functional dendritic cells from human monocytes under serum-free condition. J Biol Eng 2023; 17:6. [PMID: 36703209 PMCID: PMC9881386 DOI: 10.1186/s13036-023-00325-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Dendritic cells (DCs) are differentiated from monocytes, and have a strong ability to perform phagocytosis, present antigens and activate T cell immune response. Therefore, DCs are one of the key factors in fighting cancer in immunotherapy, and it is an important issue to develop a serum-free system for DC differentiation and expansion in vitro for clinical application. RESULTS In this study, IL-6 and M-CSF were determined and a concentration combination of cytokines was optimized to develop an optimal DC serum-free differentiation medium (SF-DC Optimal) that can effectively differentiate CD14+ monocytes into CD40+CD209+ DCs. After differentiation, the morphology, growth kinetics, surface antigen expression, phagocytosis ability, cytokine secretion, mixed lymphocyte reaction and stimulation for maturation of the differentiated DCs were checked and confirmed. Importantly, this research is the first report finding that the addition an extra low concentration of IL-6 and M-CSF exhibited a synergistic effect with GM-CSF and IL-4 to generate higher numbers and more fully functional DCs than the addition of GM-CSF and IL-4 only under serum-free condition. CONCLUSION A large number of functional DCs can be generated by using SF-DC Optimal medium and provide an alternative source of DCs for related basic research and clinical applications.
Collapse
Affiliation(s)
- Chao-Ling Yao
- grid.64523.360000 0004 0532 3255Department of Chemical Engineering, National Cheng Kung University, No. 1, University Road, Tainan, 70101 Taiwan
| | - Tsung-Yu Tseng
- grid.64523.360000 0004 0532 3255Department of Chemical Engineering, National Cheng Kung University, No. 1, University Road, Tainan, 70101 Taiwan
| |
Collapse
|
14
|
Katopodi T, Petanidis S, Charalampidis C, Chatziprodromidou I, Eskitzis P, Tsavlis D, Zarogoulidis P, Kosmidis C, Matthaios D, Porpodis K. Tumor-Infiltrating Dendritic Cells: Decisive Roles in Cancer Immunosurveillance, Immunoediting, and Tumor T Cell Tolerance. Cells 2022; 11:cells11203183. [PMID: 36291050 PMCID: PMC9600942 DOI: 10.3390/cells11203183] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
The tumor microenvironment plays a key role in progression of tumorigenesis, tumor progression, and metastasis. Accumulating data reveal that dendritic cells (DCs) appear to play a key role in the development and progression of metastatic neoplasia by driving immune system dysfunction and establishing immunosuppression, which is vital for tumor evasion of host immune response. Consequently, in this review, we will discuss the function of tumor-infiltrating DCs in immune cell signaling pathways that lead to treatment resistance, tumor recurrence, and immunosuppression. We will also review DC metabolism, differentiation, and plasticity, which are essential for metastasis and the development of lung tumors. Furthermore, we will take into account the interaction between myeloid cells and DCs in tumor-related immunosuppression. We will specifically look into the molecular immune-related mechanisms in the tumor microenvironment that result in reduced drug sensitivity and tumor relapse, as well as methods for combating drug resistance and focusing on immunosuppressive tumor networks. DCs play a crucial role in modulating the immune response. Especially, as cancer progresses, DCs may switch from playing an immunostimulatory to an inhibitory role. This article’s main emphasis is on tumor-infiltrating DCs. We address how they affect tumor growth and expansion, and we highlight innovative approaches for therapeutic modulation of these immunosuppressive DCs which is necessary for future personalized therapeutic approaches.
Collapse
Affiliation(s)
- Theodora Katopodi
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Savvas Petanidis
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-2310-999-205; Fax: +30-2310-999-208
| | | | | | - Panagiotis Eskitzis
- Department of Obstetrics, University of Western Macedonia, 50100 Kozani, Greece
| | - Drosos Tsavlis
- Laboratory of Experimental Physiology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paul Zarogoulidis
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece
| | - Christoforos Kosmidis
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece
| | | | - Konstantinos Porpodis
- Pulmonary Department-Oncology Unit, “G.Papanikolaou” General Hospital, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| |
Collapse
|
15
|
Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells 2022; 11:cells11193028. [PMID: 36230990 PMCID: PMC9563837 DOI: 10.3390/cells11193028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) are a unique myeloid cell lineage that play a central role in the priming of the adaptive immune response. As such, they are an attractive target for immune oncology based therapeutic approaches. However, targeting these cells has proven challenging with many studies proving inconclusive or of no benefit in a clinical trial setting. In this review, we highlight the known and unknown about this rare but powerful immune cell. As technologies have expanded our understanding of the complexity of DC development, subsets and response features, we are now left to apply this knowledge to the design of new therapeutic strategies in cancer. We propose that utilization of these technologies through a multiomics approach will allow for an improved directed targeting of DCs in a clinical trial setting. In addition, the DC research community should consider a consensus on subset nomenclature to distinguish new subsets from functional or phenotypic changes in response to their environment.
Collapse
|
16
|
Wang D, Cui Q, Yang YJ, Liu AQ, Zhang G, Yu JC. Application of dendritic cells in tumor immunotherapy and progress in the mechanism of anti-tumor effect of Astragalus polysaccharide (APS) modulating dendritic cells: a review. Biomed Pharmacother 2022; 155:113541. [PMID: 36127221 DOI: 10.1016/j.biopha.2022.113541] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells (APCs) that are essential in mediating the body's natural and adaptive immune responses. The body can regulate the function of DCs in various ways to enhance their antitumor effects. In the tumour microenvironment (TME), antigen-specific T cell responses are initiated through DC processing and delivery of tumour-associated antigens (TAAs); conversely, tumour cells inhibit DC recruitment by releasing metabolites, cytokines and other regulatory TME and function. Different subpopulations of DCs exist in tumour tissues, and their functions vary. Insight into DC subgroups in TME allows assessment of the effectiveness of tumour immunotherapy. Astragalus polysaccharide (APS) is the main component of the Chinese herb Astragalus membranaceus. The study found that the antitumor effects of APS are closely related to DCs. APS can promote the expression of surface molecules CD80 and CD86, promote the maturation of DCs, and activate CTL to exert antitumor effects. We reviewed the application of DCs in tumor immunotherapy and the mechanism of modulation of DCs by Astragalus polysaccharide to provide new directions and strategies for tumor therapy and new drug development.
Collapse
Affiliation(s)
- Dong Wang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Qian Cui
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Yan Jie Yang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - A Qing Liu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Guan Zhang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Jian Chun Yu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China.
| |
Collapse
|
17
|
Zhou C, Liang T, Jiang J, Zhang Z, Chen J, Chen T, Chen L, Sun X, Huang S, Zhu J, Wu S, Zhan X, Liu C. Immune cell infiltration-related clinical diagnostic model for Ankylosing Spondylitis. Front Genet 2022; 13:949882. [PMID: 36263434 PMCID: PMC9575679 DOI: 10.3389/fgene.2022.949882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background: The pathogenesis and diagnosis of Ankylosing Spondylitis (AS) has remained uncertain due to several reasons, including the lack of studies on the local and systemic immune response in AS. To construct a clinical diagnostic model, this study identified the micro RNA-messenger RNA (miRNA-mRNA) interaction network and immune cell infiltration-related hub genes associated with AS. Materials and Methods: Total RNA was extracted and purified from the interspinous ligament tissue samples of three patients with AS and three patients without AS; miRNA and mRNA microarrays were constructed using the extracted RNA. Bioinformatic tools were used to construct an miRNA-mRNA network, identify hub genes, and analyze immune infiltration associated with AS. Next, we collected the blood samples and clinical characteristics of 359 patients (197 with AS and 162 without AS). On the basis of the clinical characteristics and results of the routine blood tests, we selected immune-related cells whose numbers were significantly different in patients with AS and patients without AS. Univariate and multivariate logistic regression analysis was performed to construct a nomogram. Immunohistochemistry staining analysis was utilized to verify the differentially expression of LYN in AS and controls. Results: A total of 225 differentially expressed miRNAs (DE miRNAs) and 406 differentially expressed mRNAs (DE mRNAs) were identified from the microarray. We selected 15 DE miRNAs and 38 DE mRNAs to construct a miRNA-mRNA network. The expression of LYN, an immune-related gene, correlated with the counts of monocytes, neutrophils, and dendritic cells. Based on the independent predictive factors of sex, age, and counts of monocytes, neutrophils, and white blood cells, a nomogram was established. Receiver operating characteristic (ROC) analysis was performed to evaluate the nomogram, with a C-index of 0.835 and AUC of 0.855. Conclusion:LYN, an immune-related hub gene, correlated with immune cell infiltration in patients with AS. In addition, the counts of monocytes and neutrophils were the independent diagnostic factors for AS. If verified in future studies, a diagnostic model based on these findings may be used to predict AS effectively.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xinli Zhan
- *Correspondence: Xinli Zhan, ; Chong Liu,
| | - Chong Liu
- *Correspondence: Xinli Zhan, ; Chong Liu,
| |
Collapse
|
18
|
Kamio N, Yokota A, Tokuda Y, Ogasawara C, Nakano M, Nagao M, Tashiro K, Maekawa T, Onai N, Hirai H. A Novel CD135+ Subset of Mouse Monocytes with a Distinct Differentiation Pathway and Antigen-Presenting Properties. THE JOURNAL OF IMMUNOLOGY 2022; 209:498-509. [DOI: 10.4049/jimmunol.2100024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/24/2022] [Indexed: 01/04/2023]
Abstract
Abstract
The mononuclear phagocyte system (MPS), composed of monocytes/macrophages and dendritic cells (DCs), plays a critical role at the interface of the innate and adaptive immune systems. However, the simplicity of MPS has been challenged recently by discoveries of novel cellular components. In the current study, we identified the CD135+ subset of monocytes as a novel class of APCs in mice. CD135+ monocytes were readily found in the bone marrow, spleen, and peripheral blood at steady state, and they expressed markers specific to DCs, including MHC class II and CD209a, along with markers for monocytes/macrophages. In addition, this subset phagocytosed bacteria and activated naive T lymphocytes, fulfilling the criteria for APCs. CD135+ monocytes were derived directly from macrophage DC progenitors, not from common monocyte progenitors or other monocytes, suggesting that these are distinct from conventional monocytes. These findings facilitate our understanding of the MPS network that regulates immune responses for host defense.
Collapse
Affiliation(s)
- Naoka Kamio
- *Department of Clinical Laboratory Medicine, Kyoto University Hospital, Kyoto, Japan
- †Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- ‡Laboratory of Stem Cell Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Asumi Yokota
- ‡Laboratory of Stem Cell Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- §Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, OH
| | - Yuichi Tokuda
- ¶Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Ogasawara
- ‖Department of Immunology, Kanazawa Medical University, Japan; and
| | - Masakazu Nakano
- ¶Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Miki Nagao
- *Department of Clinical Laboratory Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Kei Tashiro
- ¶Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taira Maekawa
- †Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- #Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Nobuyuki Onai
- ‖Department of Immunology, Kanazawa Medical University, Japan; and
| | - Hideyo Hirai
- *Department of Clinical Laboratory Medicine, Kyoto University Hospital, Kyoto, Japan
- †Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- ‡Laboratory of Stem Cell Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
19
|
Lasaviciute G, Barz M, van der Heiden M, Arasa C, Tariq K, Quin J, Östlund Farrants AK, Sverremark-Ekström E. Gut commensal Limosilactobacillus reuteri induces atypical memory-like phenotype in human dendritic cells in vitro. Gut Microbes 2022; 14:2045046. [PMID: 35258405 PMCID: PMC8920211 DOI: 10.1080/19490976.2022.2045046] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Memory-like responses in innate immune cells confer nonspecific protection against secondary exposures. A number of microbial agents have been found to induce enhanced or diminished recall responses in innate cells, however, studies investigating the ability of probiotic bacteria to trigger such effects are lacking. Here, we show that priming of human monocytes with a secretome from the gut probiotic bacterium Limosilactobacillus (L.) reuteri induces a mixed secondary response phenotype in monocyte-derived dendritic cells (mo-DCs), with a strong IL-6 and IL-1β response but low TNFα, IL-23 and IL-27 secretion. Instead, blood DC priming with L. reuteri-secretome resembles a tolerant state upon secondary exposure. A similar pattern was found in conventional and gut-like (retinoic acid exposed) DCs, although retinoic acid hampered TNFα and IL-6 production and enrichment of histone modifications in L. reuteri-secretome primed mo-DC cultures. Further, we show that the memory-like phenotype of mo-DCs, induced by priming stimuli, is important for subsequent T helper (Th) cell differentiation pathways and might determine the inflammatory nature of Th cells. We also show enhanced recall responses characterized by robust inflammatory cytokines and lactate production in the gut-like mo-DCs derived from β-glucan primed monocytes. Such responses were accompanied with enriched histone modifications at the promoter of genes associated with a trained phenotype in myeloid cells. Altogether, we demonstrate that a gut commensal-derived secretome prompts recall responses in human DCs which differ from that induced by classical training agents such as β-glucan. Our results could be beneficial for future therapeutic interventions where T cell responses are needed to be modulated.
Collapse
Affiliation(s)
- Gintare Lasaviciute
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Myriam Barz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marieke van der Heiden
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Claudia Arasa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Kanwal Tariq
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jaclyn Quin
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | | | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden,CONTACT Eva Sverremark-Ekström Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, SE-106 91, Sweden
| |
Collapse
|
20
|
Nauman G, Danzl NM, Lee J, Borsotti C, Madley R, Fu J, Hölzl MA, Dahmani A, Dorronsoro Gonzalez A, Chavez É, Campbell SR, Yang S, Satwani P, Liu K, Sykes M. Defects in Long-Term APC Repopulation Ability of Adult Human Bone Marrow Hematopoietic Stem Cells (HSCs) Compared with Fetal Liver HSCs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1652-1663. [PMID: 35315788 PMCID: PMC8976823 DOI: 10.4049/jimmunol.2100966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/25/2022] [Indexed: 04/28/2023]
Abstract
Immunodeficient mice reconstituted with immune systems from patients, or personalized immune (PI) mice, are powerful tools for understanding human disease. Compared with immunodeficient mice transplanted with human fetal thymus tissue and fetal liver-derived CD34+ cells administered i.v. (Hu/Hu mice), PI mice, which are transplanted with human fetal thymus and adult bone marrow (aBM) CD34+ cells, demonstrate reduced levels of human reconstitution. We characterized APC and APC progenitor repopulation in human immune system mice and detected significant reductions in blood, bone marrow (BM), and splenic APC populations in PI compared with Hu/Hu mice. APC progenitors and hematopoietic stem cells (HSCs) were less abundant in aBM CD34+ cells compared with fetal liver-derived CD34+ cell preparations, and this reduction in APC progenitors was reflected in the BM of PI compared with Hu/Hu mice 14-20 wk posttransplant. The number of HSCs increased in PI mice compared with the originally infused BM cells and maintained functional repopulation potential, because BM from some PI mice 28 wk posttransplant generated human myeloid and lymphoid cells in secondary recipients. Moreover, long-term PI mouse BM contained functional T cell progenitors, evidenced by thymopoiesis in thymic organ cultures. Injection of aBM cells directly into the BM cavity, transgenic expression of hematopoietic cytokines, and coinfusion of human BM-derived mesenchymal stem cells synergized to enhance long-term B cell and monocyte levels in PI mice. These improvements allow a sustained time frame of 18-22 wk where APCs and T cells are present and greater flexibility for modeling immune disease pathogenesis and immunotherapies in PI mice.
Collapse
Affiliation(s)
- Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Nichole M Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jaeyop Lee
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Chiara Borsotti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Markus A Hölzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Alexander Dahmani
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Akaitz Dorronsoro Gonzalez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Éstefania Chavez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Sean R Campbell
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Suxiao Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Prakash Satwani
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Pediatrics, Columbia University Medical Center, Columbia University, New York, NY
| | - Kang Liu
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT; and
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY;
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY
| |
Collapse
|
21
|
HIV transmitting mononuclear phagocytes; integrating the old and new. Mucosal Immunol 2022; 15:542-550. [PMID: 35173293 PMCID: PMC9259493 DOI: 10.1038/s41385-022-00492-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023]
Abstract
In tissue, mononuclear phagocytes (MNP) are comprised of Langerhans cells, dendritic cells, macrophages and monocyte-derived cells. They are the first immune cells to encounter HIV during transmission and transmit the virus to CD4 T cells as a consequence of their antigen presenting cell function. To understand the role these cells play in transmission, their phenotypic and functional characterisation is important. With advancements in high parameter single cell technologies, new MNPs subsets are continuously being discovered and their definition and classification is in a state of flux. This has important implications for our knowledge of HIV transmission, which requires a deeper understanding to design effective vaccines and better blocking strategies. Here we review the historical research of the role MNPs play in HIV transmission up to the present day and revaluate these studies in the context of our most recent understandings of the MNP system.
Collapse
|
22
|
Mysore V, Cullere X, Settles ML, Ji X, Kattan MW, Desjardins M, Durbin-Johnson B, Gilboa T, Baden LR, Walt DR, Lichtman AH, Jehi L, Mayadas TN. Protective heterologous T cell immunity in COVID-19 induced by the trivalent MMR and Tdap vaccine antigens. MED 2021; 2:1050-1071.e7. [PMID: 34414383 PMCID: PMC8363466 DOI: 10.1016/j.medj.2021.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/25/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND T cells control viral infection, promote vaccine durability, and in coronavirus disease 2019 (COVID-19) associate with mild disease. We investigated whether prior measles-mumps-rubella (MMR) or tetanus-diphtheria-pertussis (Tdap) vaccination elicits cross-reactive T cells that mitigate COVID-19. METHODS Antigen-presenting cells (APC) loaded ex vivo with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), MMR, or Tdap antigens and autologous T cells from COVID-19-convalescent participants, uninfected individuals, and COVID-19 mRNA-vaccinated donors were co-cultured. T cell activation and phenotype were detected by interferon-γ (IFN-γ) enzyme-linked immunospot (ELISpot) assays and flow cytometry. ELISAs (enzyme-linked immunosorbant assays) and validation studies identified the APC-derived cytokine(s) driving T cell activation. TCR clonotyping and single-cell RNA sequencing (scRNA-seq) identified cross-reactive T cells and their transcriptional profile. A propensity-weighted analysis of COVID-19 patients estimated the effects of MMR and Tdap vaccination on COVID-19 outcomes. FINDINGS High correlation was observed between T cell responses to SARS-CoV-2 (spike-S1 and nucleocapsid) and MMR and Tdap proteins in COVID-19-convalescent and -vaccinated individuals. The overlapping T cell population contained an effector memory T cell subset (effector memory re-expressing CD45RA on T cells [TEMRA]) implicated in protective, anti-viral immunity, and their detection required APC-derived IL-15, known to sensitize T cells to activation. Cross-reactive TCR repertoires detected in antigen-experienced T cells recognizing SARS-CoV-2, MMR, and Tdap epitopes had TEMRA features. Indices of disease severity were reduced in MMR- or Tdap-vaccinated individuals by 32%-38% and 20%-23%, respectively, among COVID-19 patients. CONCLUSIONS Tdap and MMR memory T cells reactivated by SARS-CoV-2 may provide protection against severe COVID-19. FUNDING This study was supported by a National Institutes of Health (R01HL065095, R01AI152522, R01NS097719) donation from Barbara and Amos Hostetter and the Chleck Foundation.
Collapse
Affiliation(s)
- Vijayashree Mysore
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Xavier Cullere
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Matthew L Settles
- Bioinformatics Core Facility in the Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Xinge Ji
- Quantitative Health Science Department, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael W Kattan
- Quantitative Health Science Department, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michaël Desjardins
- Department of Medicine, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | | | - Tal Gilboa
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Lara Jehi
- Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Merah-Mourah F, Cohen SO, Haziot A. A Two-Stage Flow Cytometry Strategy to Distinguish Single Cells from Doublets in Heterogeneous Cell Mixtures and Improve Cell Cluster Identification: Application to Human Monocyte Subpopulations. Curr Protoc 2021; 1:e229. [PMID: 34416100 DOI: 10.1002/cpz1.229] [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: 11/11/2022]
Abstract
Flow cytometry is a powerful method, widely used to identify cell types present in tissues, to describe their phenotypes, and to purify cells for functional analyses. As a single cell technique, flow cytometry relies on identifying and excluding cell doublets and aggregates present in samples in the initial gating steps. This identification is based on detection of events generating electrical pulses falling outside of linear variations of pulse height, width, and area in a singlet population with increasing cell sizes. In heterogeneous cell mixtures, however, with cell types varying extensively in size and granularity, exclusion of doublets has the risk of removing single cells that co-localize with doublets of another cell type. This is particularly the case when doublets of a smaller cell type overlap with large cells of a distinct, larger cell type. Here, we describe a gating method to reduce this risk. In this protocol, initial gating steps aim to segregate cells according to physical characteristics (such as size and granularity) and gene expression properties in order to obtain more homogeneous cell clusters. Doublet exclusion is then performed separately in each cluster, minimizing the risk of confusion between single cells and doublets. To illustrate this protocol, human blood monocytes are separated and analyzed. By implementing this protocol, we were able to reveal the existence of a population of large monocytes previously unrecognized using conventional gating strategies. In subsequent functional assays, we have shown that this novel population exhibits unique inflammatory responses, highlighting the need and pertinence of this approach to identify and characterize infrequent-yet functionally relevant-cell populations present in complex cell mixtures. © 2021 Wiley Periodicals LLC. Basic Protocol: Distinguishing single cells from doublets in heterogeneous cell mixtures by flow cytometry.
Collapse
Affiliation(s)
- Fadila Merah-Mourah
- INSERM U976, Institut de Recherche Saint Louis, Paris, France.,Université de Paris, Paris, France.,Current address: Department of Endocrinology, Diabetology, Nutrition, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Shannon O Cohen
- INSERM U976, Institut de Recherche Saint Louis, Paris, France.,Université de Paris, Paris, France
| | - Alain Haziot
- INSERM U976, Institut de Recherche Saint Louis, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
24
|
Nuñez-Reza KJ, Naldi A, Sánchez-Jiménez A, Leon-Apodaca AV, Santana MA, Thomas-Chollier M, Thieffry D, Medina-Rivera A. Logical modelling of in vitro differentiation of human monocytes into dendritic cells unravels novel transcriptional regulatory interactions. Interface Focus 2021; 11:20200061. [PMID: 34123352 PMCID: PMC8193469 DOI: 10.1098/rsfs.2020.0061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells, thereby connecting innate and adaptive immunity. Because of their role in establishing adaptive immunity, they constitute promising targets for immunotherapy. Monocytes can differentiate into DCs in vitro in the presence of colony-stimulating factor 2 (CSF2) and interleukin 4 (IL4), activating four signalling pathways (MAPK, JAK/STAT, NFKB and PI3K). However, the downstream transcriptional programme responsible for DC differentiation from monocytes (moDCs) remains unknown. By analysing the scientific literature on moDC differentiation, we established a preliminary logical model that helped us identify missing information regarding the activation of genes responsible for this differentiation, including missing targets for key transcription factors (TFs). Using ChIP-seq and RNA-seq data from the Blueprint consortium, we defined active and inactive promoters, together with differentially expressed genes in monocytes, moDCs and macrophages, which correspond to an alternative cell fate. We then used this functional genomic information to predict novel targets for previously identified TFs. By integrating this information, we refined our model and recapitulated the main established facts regarding moDC differentiation. Prospectively, the resulting model should be useful to develop novel immunotherapies targeting moDCs.
Collapse
Affiliation(s)
- Karen J Nuñez-Reza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Aurélien Naldi
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Arantza Sánchez-Jiménez
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Ana V Leon-Apodaca
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Morgane Thomas-Chollier
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Denis Thieffry
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| |
Collapse
|
25
|
Mysore V, Cullere X, Settles ML, Ji X, Kattan MW, Desjardins M, Durbin-Johnson B, Gilboa T, Baden LR, Walt DR, Lichtman A, Jehi L, Mayadas TN. Protective heterologous T cell immunity in COVID-19 induced by MMR and Tdap vaccine antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.03.441323. [PMID: 33972940 PMCID: PMC8109200 DOI: 10.1101/2021.05.03.441323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
T cells are critical for control of viral infection and effective vaccination. We investigated whether prior Measles-Mumps-Rubella (MMR) or Tetanus-Diphtheria-pertussis (Tdap) vaccination elicit cross-reactive T cells that mitigate COVID-19. Using co-cultures of antigen presenting cells (APC) loaded with antigens and autologous T cells, we found a high correlation between responses to SARS-CoV-2 (Spike-S1 and Nucleocapsid) and MMR and Tdap vaccine proteins in both SARS-CoV-2 infected individuals and individuals immunized with mRNA-based SARS-CoV-2 vaccines. The overlapping T cell population contained effector memory T cells (TEMRA) previously implicated in anti-viral immunity and their activation required APC-derived IL-15. TCR- and scRNA-sequencing detected cross-reactive clones with TEMRA features among the cells recognizing SARS-CoV-2, MMR and Tdap epitopes. A propensity-weighted analysis of 73,582 COVID-19 patients revealed that severe disease outcomes (hospitalization and transfer to intensive care unit or death) were reduced in MMR or Tdap vaccinated individuals by 38-32% and 23-20% respectively. In summary, SARS-CoV-2 re-activates memory T cells generated by Tdap and MMR vaccines, which may reduce disease severity.
Collapse
|
26
|
Bertram KM, Truong NR, Smith JB, Kim M, Sandgren KJ, Feng KL, Herbert JJ, Rana H, Danastas K, Miranda-Saksena M, Rhodes JW, Patrick E, Cohen RC, Lim J, Merten SL, Harman AN, Cunningham AL. Herpes Simplex Virus type 1 infects Langerhans cells and the novel epidermal dendritic cell, Epi-cDC2s, via different entry pathways. PLoS Pathog 2021; 17:e1009536. [PMID: 33905459 PMCID: PMC8104422 DOI: 10.1371/journal.ppat.1009536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 05/07/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Skin mononuclear phagocytes (MNPs) provide the first interactions of invading viruses with the immune system. In addition to Langerhans cells (LCs), we recently described a second epidermal MNP population, Epi-cDC2s, in human anogenital epidermis that is closely related to dermal conventional dendritic cells type 2 (cDC2) and can be preferentially infected by HIV. Here we show that in epidermal explants topically infected with herpes simplex virus (HSV-1), both LCs and Epi-cDC2s interact with HSV-1 particles and infected keratinocytes. Isolated Epi-cDC2s support higher levels of infection than LCs in vitro, inhibited by acyclovir, but both MNP subtypes express similar levels of the HSV entry receptors nectin-1 and HVEM, and show similar levels of initial uptake. Using inhibitors of endosomal acidification, actin and cholesterol, we found that HSV-1 utilises different entry pathways in each cell type. HSV-1 predominantly infects LCs, and monocyte-derived MNPs, via a pH-dependent pathway. In contrast, Epi-cDC2s are mainly infected via a pH-independent pathway which may contribute to the enhanced infection of Epi-cDC2s. Both cells underwent apoptosis suggesting that Epi-cDC2s may follow the same dermal migration and uptake by dermal MNPs that we have previously shown for LCs. Thus, we hypothesize that the uptake of HSV and infection of Epi-cDC2s will stimulate immune responses via a different pathway to LCs, which in future may help guide HSV vaccine development and adjuvant targeting.
Collapse
Affiliation(s)
- Kirstie M. Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jacinta B. Smith
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Min Kim
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Kerrie J. Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Konrad L. Feng
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jason J. Herbert
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Kevin Danastas
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Monica Miranda-Saksena
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jake W. Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Ralph C. Cohen
- Department of Surgery, University of Sydney and The Children’s Hospital at Westmead, Westmead, Australia
| | - Jake Lim
- Department of Surgery, Westmead Private Hospital, Westmead, Australia
| | - Steven L. Merten
- Department of Surgery, Macquarie University Hospital, Macquarie Park, Australia
| | - Andrew N. Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
- * E-mail:
| |
Collapse
|
27
|
Leite Dantas R, Freff J, Ambrée O, Beins EC, Forstner AJ, Dannlowski U, Baune BT, Scheu S, Alferink J. Dendritic Cells: Neglected Modulators of Peripheral Immune Responses and Neuroinflammation in Mood Disorders? Cells 2021; 10:941. [PMID: 33921690 PMCID: PMC8072712 DOI: 10.3390/cells10040941] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Affective disorders (AD) including major depressive disorder (MDD) and bipolar disorder (BD) are common mood disorders associated with increased disability and poor health outcomes. Altered immune responses characterized by increased serum levels of pro-inflammatory cytokines and neuroinflammation are common findings in patients with AD and in corresponding animal models. Dendritic cells (DCs) represent a heterogeneous population of myeloid cells that orchestrate innate and adaptive immune responses and self-tolerance. Upon sensing exogenous and endogenous danger signals, mature DCs secrete proinflammatory factors, acquire migratory and antigen presenting capacities and thus contribute to neuroinflammation in trauma, autoimmunity, and neurodegenerative diseases. However, little is known about the involvement of DCs in the pathogenesis of AD. In this review, we summarize the current knowledge on DCs in peripheral immune responses and neuroinflammation in MDD and BD. In addition, we consider the impact of DCs on neuroinflammation and behavior in animal models of AD. Finally, we will discuss therapeutic perspectives targeting DCs and their effector molecules in mood disorders.
Collapse
Affiliation(s)
- Rafael Leite Dantas
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Jana Freff
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Oliver Ambrée
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany;
- Center of Cellular Nanoanalytics, University of Osnabrück, 49076 Osnabrück, Germany
| | - Eva C. Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
| | - Andreas J. Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, 52428 Jülich, Germany
| | - Udo Dannlowski
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
| | - Bernhard T. Baune
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany;
| | - Judith Alferink
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| |
Collapse
|
28
|
Rhodes JW, Botting RA, Bertram KM, Vine EE, Rana H, Baharlou H, Vegh P, O'Neil TR, Ashhurst AS, Fletcher J, Parnell GP, Graham JD, Nasr N, Lim JJK, Barnouti L, Haertsch P, Gosselink MP, Di Re A, Reza F, Ctercteko G, Jenkins GJ, Brooks AJ, Patrick E, Byrne SN, Hunter E, Haniffa MA, Cunningham AL, Harman AN. Human anogenital monocyte-derived dendritic cells and langerin+cDC2 are major HIV target cells. Nat Commun 2021; 12:2147. [PMID: 33846309 PMCID: PMC8042121 DOI: 10.1038/s41467-021-22375-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue mononuclear phagocytes (MNP) are specialised in pathogen detection and antigen presentation. As such they deliver HIV to its primary target cells; CD4 T cells. Most MNP HIV transmission studies have focused on epithelial MNPs. However, as mucosal trauma and inflammation are now known to be strongly associated with HIV transmission, here we examine the role of sub-epithelial MNPs which are present in a diverse array of subsets. We show that HIV can penetrate the epithelial surface to interact with sub-epithelial resident MNPs in anogenital explants and define the full array of subsets that are present in the human anogenital and colorectal tissues that HIV may encounter during sexual transmission. In doing so we identify two subsets that preferentially take up HIV, become infected and transmit the virus to CD4 T cells; CD14+CD1c+ monocyte-derived dendritic cells and langerin-expressing conventional dendritic cells 2 (cDC2).
Collapse
Affiliation(s)
- Jake W Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Rachel A Botting
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia.,Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Erica E Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Peter Vegh
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Thomas R O'Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Anneliese S Ashhurst
- School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - James Fletcher
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Grant P Parnell
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - J Dinny Graham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | | | | | - Peter Haertsch
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia
| | - Martijn P Gosselink
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Angelina Di Re
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Faizur Reza
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Grahame Ctercteko
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Gregory J Jenkins
- Department of Obstetrics and Gynaecology, Westmead Hospital, Westmead, NSW, Australia
| | - Andrew J Brooks
- Department of Urology, Westmead Hospital, Westmead, NSW, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Maths and Statistics, Faculty of Science, The University of Sydney, Camperdown, NSW, Australia
| | - Scott N Byrne
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | | | - Muzlifah A Haniffa
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK.,Wellcome Sanger Institute, Hinxton, UK.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia. .,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
29
|
Zyulina V, Yan KK, Ju B, Schwarzenberger E, Passegger C, Tam-Amersdorfer C, Pan Q, Sconocchia T, Pollack C, Shaner B, Zebisch A, Easton J, Yu J, Silva JM, Strobl H. The miR-424(322)/503 gene cluster regulates pro- versus anti-inflammatory skin DC subset differentiation by modulating TGF-β signaling. Cell Rep 2021; 35:109049. [PMID: 33910004 DOI: 10.1016/j.celrep.2021.109049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/26/2020] [Accepted: 04/06/2021] [Indexed: 11/15/2022] Open
Abstract
Transforming growth factor β (TGF-β) family ligands are key regulators of dendritic cell (DC) differentiation and activation. Epidermal Langerhans cells (LCs) require TGF-β family signaling for their differentiation, and canonical TGF-β1 signaling secures a non-activated LC state. LCs reportedly control skin inflammation and are replenished from peripheral blood monocytes, which also give rise to pro-inflammatory monocyte-derived DCs (moDCs). By studying mechanisms in inflammation, we previously screened LCs versus moDCs for differentially expressed microRNAs (miRNAs). This revealed that miR-424/503 is the most strongly inversely regulated (moDCs > LCs). We here demonstrate that miR-424/503 is induced during moDC differentiation and promotes moDC differentiation in human and mouse. Inversely, forced repression of miR-424 during moDC differentiation facilitates TGF-β1-dependent LC differentiation. Mechanistically, miR-424/503 deficiency in monocyte/DC precursors leads to the induction of TGF-β1 response genes critical for LC differentiation. Therefore, the miR-424/503 gene cluster plays a decisive role in anti-inflammatory LC versus pro-inflammatory moDC differentiation from monocytes.
Collapse
Affiliation(s)
- Victoria Zyulina
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Koon-Kiu Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Elke Schwarzenberger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Christina Passegger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Qingfei Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Christian Pollack
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Bridget Shaner
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, 8010 Graz, Austria; Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Jose M Silva
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA.
| | - Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria.
| |
Collapse
|
30
|
Abstract
As the professional antigen-presenting cells of the immune system, dendritic cells (DCs) sense the microenvironment and shape the ensuing adaptive immune response. DCs can induce both immune activation and immune tolerance according to the peripheral cues. Recent work has established that DCs comprise several phenotypically and functionally heterogeneous subsets that differentially regulate T lymphocyte differentiation. This review summarizes both mouse and human DC subset phenotypes, development, diversification, and function. We focus on advances in our understanding of how different DC subsets regulate distinct CD4+ T helper (Th) cell differentiation outcomes, including Th1, Th2, Th17, T follicular helper, and T regulatory cells. We review DC subset intrinsic properties, local tissue microenvironments, and other immune cells that together determine Th cell differentiation during homeostasis and inflammation.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Shuting Chen
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
31
|
Ackaert C, Smiejkowska N, Xavier C, Sterckx YGJ, Denies S, Stijlemans B, Elkrim Y, Devoogdt N, Caveliers V, Lahoutte T, Muyldermans S, Breckpot K, Keyaerts M. Immunogenicity Risk Profile of Nanobodies. Front Immunol 2021; 12:632687. [PMID: 33767701 PMCID: PMC7985456 DOI: 10.3389/fimmu.2021.632687] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/15/2021] [Indexed: 12/31/2022] Open
Abstract
Nanobodies (Nbs), the variable domains of camelid heavy chain-only antibodies, are a promising class of therapeutics or in vivo imaging reagents entering the clinic. They possess unique characteristics, including a minimal size, providing fast pharmacokinetics, high-target specificity, and an affinity in the (sub-)nanomolar range in conjunction with an easy selection and production, which allow them to outperform conventional antibodies for imaging and radiotherapeutic purposes. As for all protein theranostics, extended safety assessment and investigation of their possible immunogenicity in particular are required. In this study, we assessed the immunogenicity risk profile of two Nbs that are in phase II clinical trials: a first Nb against Human Epidermal growth factor Receptor 2 (HER2) for PET imaging of breast cancer and a second Nb with specificity to the Macrophage Mannose Receptor (MMR) for PET imaging of tumor-associated macrophages. For the anti-HER2 Nb, we show that only one out of 20 patients had a low amount of pre-existing anti-drug antibodies (ADAs), which only marginally increased 3 months after administering the Nb, and without negative effects of safety and pharmacokinetics. Further in vitro immunogenicity assessment assays showed that both non-humanized Nbs were taken up by human dendritic cells but exhibited no or only a marginal capacity to activate dendritic cells or to induce T cell proliferation. From our data, we conclude that monomeric Nbs present a low immunogenicity risk profile, which is encouraging for their future development toward potential clinical applications.
Collapse
Affiliation(s)
- Chloé Ackaert
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Natalia Smiejkowska
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catarina Xavier
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Yann G J Sterckx
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Laboratory of Medical Biochemistry, University of Antwerp (UA), Wilrijk, Belgium
| | | | - Benoit Stijlemans
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yvon Elkrim
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vicky Caveliers
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Tony Lahoutte
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Serge Muyldermans
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| |
Collapse
|
32
|
Wooster AL, Girgis LH, Brazeale H, Anderson TS, Wood LM, Lowe DB. Dendritic cell vaccine therapy for colorectal cancer. Pharmacol Res 2021; 164:105374. [PMID: 33348026 PMCID: PMC7867624 DOI: 10.1016/j.phrs.2020.105374] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths in the United States despite an array of available treatment options. Current standard-of-care interventions for this malignancy include surgical resection, chemotherapy, and targeted therapies depending on the disease stage. Specifically, infusion of anti-vascular endothelial growth factor agents in combination with chemotherapy was an important development in improving the survival of patients with advanced colorectal cancer, while also helping give rise to other forms of anti-angiogenic therapies. Yet, one approach by which tumor angiogenesis may be further disrupted is through the administration of a dendritic cell (DC) vaccine targeting tumor-derived blood vessels, leading to cytotoxic immune responses that decrease tumor growth and synergize with other systemic therapies. Early generations of such vaccines exhibited protection against various forms of cancer in pre-clinical models, but clinical results have historically been disappointing. Sipuleucel-T (Provenge®) was the first, and to-date, only dendritic cell-based therapy to receive FDA approval after significantly increasing overall survival in prostate cancer patients. The unparalleled success of Sipuleucel-T has helped revitalize the clinical development of dendritic cell vaccines, which will be examined in this review. We also highlight the promise of these vaccines to instill anti-angiogenic immunity for individuals with advanced colorectal cancer.
Collapse
Affiliation(s)
- Amanda L Wooster
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Lydia H Girgis
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Hayley Brazeale
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Trevor S Anderson
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Laurence M Wood
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Devin B Lowe
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
33
|
Alamri A, Fisk D, Upreti D, Kung SKP. A Missing Link: Engagements of Dendritic Cells in the Pathogenesis of SARS-CoV-2 Infections. Int J Mol Sci 2021; 22:1118. [PMID: 33498725 PMCID: PMC7865603 DOI: 10.3390/ijms22031118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) connect the innate and adaptive arms of the immune system and carry out numerous roles that are significant in the context of viral disease. Their functions include the control of inflammatory responses, the promotion of tolerance, cross-presentation, immune cell recruitment and the production of antiviral cytokines. Based primarily on the available literature that characterizes the behaviour of many DC subsets during Severe acute respiratory syndrome (SARS) and coronavirus disease 2019 (COVID-19), we speculated possible mechanisms through which DC could contribute to COVID-19 immune responses, such as dissemination of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to lymph nodes, mounting dysfunctional inteferon responses and T cell immunity in patients. We highlighted gaps of knowledge in our understanding of DC in COVID-19 pathogenesis and discussed current pre-clinical development of therapies for COVID-19.
Collapse
Affiliation(s)
- Abdulaziz Alamri
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| | - Derek Fisk
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| | - Deepak Upreti
- Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada;
| | - Sam K. P. Kung
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| |
Collapse
|
34
|
Wei R, Lai N, Zhao L, Zhang Z, Zhu X, Guo Q, Chu C, Fu X, Li X. Dendritic cells in pregnancy and pregnancy-associated diseases. Biomed Pharmacother 2021; 133:110921. [PMID: 33378991 DOI: 10.1016/j.biopha.2020.110921] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) play a critical immuno-modulating role in pregnancy, which requires the maternal immune system to tolerate semiallogeneic fetus and at the same time to maintain adequate defense against pathogens. DCs interact closely with other immune components such as T cells, natural killer cells and macrophages, as well as the endocrine system to keep a pregnancy-friendly environment. Aberrant DC activities have been related to various pregnancy-associated diseases such as recurrent spontaneous abortion, preterm birth, pre-eclampsia, peripartum cardiomyopathy and infectious pregnancy complications. These findings make DCs an attractive candidate for prevention or therapy on the pregnancy-associated diseases. Here, we review recent findings that provide new insights into the roles of DCs in pregnancy and the related diseases. We also discuss the medical potentials to manipulate DCs in clinics. Whereas this is an emerging area with much work remaining, we anticipate that a better understanding of the role of DCs in maternal-fetal immunotolerance and a therapeutic manipulation of DCs will help women suffering from the pregnancy-associated diseases.
Collapse
Affiliation(s)
- Ran Wei
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Nannan Lai
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, PR China
| | - Lin Zhao
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Zhen Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Xiaoxiao Zhu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Qiang Guo
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Chu Chu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Xiaoxiao Fu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Xia Li
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| |
Collapse
|
35
|
Trained immunity and tolerance in innate lymphoid cells, monocytes, and dendritic cells during allergen-specific immunotherapy. J Allergy Clin Immunol 2020; 147:1865-1877. [PMID: 33039478 DOI: 10.1016/j.jaci.2020.08.042] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Despite the efficacy of allergen-specific immunotherapy (AIT), the role of trained immunity and tolerance in this process has not been elucidated. OBJECTIVE Here, we have performed a comprehensive longitudinal analysis of the systemic innate immune cell repertoire during the course of AIT. METHODS Patients with allergy received standard preseasonal subcutaneous AIT with allergoids to birch and/or grass. Healthy controls were monitored without any intervention. Flow cytometry of innate lymphoid cell (ILC), natural killer cell, monocyte cell, and dendritic cell (DC) subsets was performed at baseline, 3 months (birch season), 6 months (grass seasons), and 12 months after the therapy in patients or at similar seasonal time points in controls. Additional analyses were performed in the third-year birch and grass season. RESULTS We observed a durable decrease in group 2 ILCs and an increase of group 1 ILCs after AIT, with dynamic changes in their composition. We found that an expansion of CD127+CD25++ clusters caused observed shifts in the heterogeneity of group 1 ILCs. In addition, we observed development of CD127+CD25++c-Kit+ group 3 ILC clusters. Moreover, we found an increase in the number of intermediate monocytes in parallel with a reduction in nonclassical monocytes during the first year after AIT. Classical and intermediate monocytes presented significant heterogeneity in patients with allergy, but AIT reduced the HLA-DR++ clusters. Finally, an increase in plasmacytoid DCs and CD141+ myeloid DCs was observed in individuals with allergy, whereas the number of CD1c+ myeloid DCs was reduced during the first year of AIT. CONCLUSION AIT induces changes in the composition and heterogeneity of circulating innate immune cells and brings them to the level observed in healthy individuals. Monitoring of ILCs, monocytes, and DCs during AIT might serve as a novel biomarker strategy.
Collapse
|
36
|
Xue J, Xu L, Zhu H, Bai M, Li X, Zhao Z, Zhong H, Cheng G, Li X, Hu F, Su Y. CD14 +CD16 - monocytes are the main precursors of osteoclasts in rheumatoid arthritis via expressing Tyro3TK. Arthritis Res Ther 2020; 22:221. [PMID: 32958023 PMCID: PMC7507256 DOI: 10.1186/s13075-020-02308-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Background Monocytes as precursors of osteoclasts in rheumatoid arthritis (RA) are well demonstrated, while monocyte subsets in osteoclast formation are still controversial. Tyro3 tyrosine kinase (Tyro3TK) is a member of the receptor tyrosine kinase family involved in immune homeostasis, the role of which in osteoclast differentiation was reported recently. This study aimed to compare the osteoclastic capacity of CD14+CD16+ and CD14+CD16− monocytes in RA and determine the potential involvement of Tyro3TK in their osteoclastogenesis. Methods Osteoclasts were induced from CD14+CD16+ and CD14+CD16− monocyte subsets isolated from healthy control (HC) and RA patients in vitro and evaluated by tartrate-resistant acid phosphatase (TRAP) staining. Then, the expression of Tyro3TK on CD14+CD16+ and CD14+CD16− monocyte subsets in the peripheral blood of RA, osteoarthritis (OA) patients, and HC were evaluated by flow cytometry and qPCR, and their correlation with RA patient clinical and immunological features was analyzed. The role of Tyro3TK in CD14+CD16− monocyte-mediated osteoclastogenesis was further investigated by osteoclast differentiation assay with Tyro3TK blockade. Results The results revealed that CD14+CD16− monocytes were the primary source of osteoclasts. Compared with HC and OA patients, the expression of Tyro3TK on CD14+CD16− monocytes in RA patients was significantly upregulated and positively correlated with the disease manifestations, such as IgM level, tender joint count, and the disease activity score. Moreover, anti-Tyro3TK antibody could inhibit Gas6-mediated osteoclast differentiation from CD14+CD16− monocytes in a dose-dependent manner. Conclusions These findings indicate that elevated Tyro3TK on CD14+CD16− monocytes serves as a critical signal for osteoclast differentiation in RA.
Collapse
Affiliation(s)
- Jimeng Xue
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Huaqun Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Mingxin Bai
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xin Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhen Zhao
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Hua Zhong
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Gong Cheng
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xue Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China. .,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China. .,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China. .,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| |
Collapse
|
37
|
Eliasse Y, Redoules D, Espinosa E. Impact of Avène Thermal Spring Water on immune cells. J Eur Acad Dermatol Venereol 2020; 34 Suppl 5:21-26. [DOI: 10.1111/jdv.16335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Y. Eliasse
- Inserm U1037 Centre de Recherche en Cancérologie de Toulouse (CRCT) Toulouse France
- Université de Toulouse Université Paul Sabatier Toulouse France
| | - D. Redoules
- Pierre Fabre Dermo‐Cosmétique Toulouse France
| | - E. Espinosa
- Inserm U1037 Centre de Recherche en Cancérologie de Toulouse (CRCT) Toulouse France
- Université de Toulouse Université Paul Sabatier Toulouse France
| |
Collapse
|
38
|
ASGR1 and Its Enigmatic Relative, CLEC10A. Int J Mol Sci 2020; 21:ijms21144818. [PMID: 32650396 PMCID: PMC7404283 DOI: 10.3390/ijms21144818] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
The large family of C-type lectin (CLEC) receptors comprises carbohydrate-binding proteins that require Ca2+ to bind a ligand. The prototypic receptor is the asialoglycoprotein receptor-1 (ASGR1, CLEC4H1) that is expressed primarily by hepatocytes. The early work on ASGR1, which is highly specific for N-acetylgalactosamine (GalNAc), established the foundation for understanding the overall function of CLEC receptors. Cells of the immune system generally express more than one CLEC receptor that serve diverse functions such as pathogen-recognition, initiation of cellular signaling, cellular adhesion, glycoprotein turnover, inflammation and immune responses. The receptor CLEC10A (C-type lectin domain family 10 member A, CD301; also called the macrophage galactose-type lectin, MGL) contains a carbohydrate-recognition domain (CRD) that is homologous to the CRD of ASGR1, and thus, is also specific for GalNAc. CLEC10A is most highly expressed on immature DCs, monocyte-derived DCs, and alternatively activated macrophages (subtype M2a) as well as oocytes and progenitor cells at several stages of embryonic development. This receptor is involved in initiation of TH1, TH2, and TH17 immune responses and induction of tolerance in naïve T cells. Ligand-mediated endocytosis of CLEC receptors initiates a Ca2+ signal that interestingly has different outcomes depending on ligand properties, concentration, and frequency of administration. This review summarizes studies that have been carried out on these receptors.
Collapse
|
39
|
Caër C, Wick MJ. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front Immunol 2020; 11:410. [PMID: 32256490 PMCID: PMC7093381 DOI: 10.3389/fimmu.2020.00410] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex immune-mediated disease of the gastrointestinal tract that increases morbidity and negatively influences the quality of life. Intestinal mononuclear phagocytes (MNPs) have a crucial role in maintaining epithelial barrier integrity while controlling pathogen invasion by activating an appropriate immune response. However, in genetically predisposed individuals, uncontrolled immune activation to intestinal flora is thought to underlie the chronic mucosal inflammation that can ultimately result in IBD. Thus, MNPs are involved in fine-tuning mucosal immune system responsiveness and have a critical role in maintaining homeostasis or, potentially, the emergence of IBD. MNPs include monocytes, macrophages and dendritic cells, which are functionally diverse but highly complementary. Despite their crucial role in maintaining intestinal homeostasis, specific functions of human MNP subsets are poorly understood, especially during diseases such as IBD. Here we review the current understanding of MNP ontogeny, as well as the recently identified human intestinal MNP subsets, and discuss their role in health and IBD.
Collapse
Affiliation(s)
- Charles Caër
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Hoober JK, Eggink LL, Cote R. Stories From the Dendritic Cell Guardhouse. Front Immunol 2019; 10:2880. [PMID: 31921144 PMCID: PMC6919295 DOI: 10.3389/fimmu.2019.02880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells [dendritic cells (DCs), macrophages, monocytes, neutrophils, and mast cells] utilize C-type (Ca2+-dependent) lectin-like (CLEC) receptors to identify and internalize pathogens or danger signals. As monitors of environmental imbalances, CLEC receptors are particularly important in the function of DCs. Activation of the immune system requires, in sequence, presentation of antigen to the T cell receptor (TCR) by DCs, interaction of co-stimulatory factors such as CD40/80/86 on DCs with CD40L and CD28 on T cells, and production of IL-12 and/or IFN-α/β to amplify T cell differentiation and expansion. Without this sequence of events within an inflammatory environment, or in a different order, antigen-specific T cells become unresponsive, are deleted or become regulatory T cells. Thus, the mode by which CLEC receptors on DCs are engaged can either elicit activation of T cells to achieve an immune response or induce tolerance. This minireview illustrates these aspects with Dectin-1, DEC205, the mannose receptor and CLEC10A as examples.
Collapse
Affiliation(s)
| | | | - Robert Cote
- Susavion Biosciences, Inc., Tempe, AZ, United States
| |
Collapse
|
41
|
Schönrich G, Raftery MJ. Dendritic Cells (DCs) as "Fire Accelerants" of Hantaviral Pathogenesis. Viruses 2019; 11:v11090849. [PMID: 31540199 PMCID: PMC6783833 DOI: 10.3390/v11090849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 01/20/2023] Open
Abstract
Hantaviruses are widespread zoonotic pathogens found around the globe. Depending on their geographical location, hantaviruses can cause two human syndromes, haemorrhagic fever with renal syndrome (HFRS) or hantavirus pulmonary syndrome (HPS). HPS and HFRS have many commonalities amongst which excessive activation of immune cells is a prominent feature. Hantaviruses replicate in endothelial cells (ECs), the major battlefield of hantavirus-induced pathogenesis, without causing cytopathic effects. This indicates that a misdirected response of human immune cells to hantaviruses is causing damage. As dendritic cells (DCs) orchestrate antiviral immune responses, they are in the focus of research analysing hantavirus-induced immunopathogenesis. In this review, we discuss the interplay between hantaviruses and DCs and the immunological consequences thereof.
Collapse
Affiliation(s)
- Günther Schönrich
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Martin J Raftery
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
42
|
Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. Immunity 2019; 51:573-589.e8. [PMID: 31474513 DOI: 10.1016/j.immuni.2019.08.008] [Citation(s) in RCA: 344] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/24/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
Human mononuclear phagocytes comprise phenotypically and functionally overlapping subsets of dendritic cells (DCs) and monocytes, but the extent of their heterogeneity and distinct markers for subset identification remains elusive. By integrating high-dimensional single-cell protein and RNA expression data, we identified distinct markers to delineate monocytes from conventional DC2 (cDC2s). Using CD88 and CD89 for monocytes and HLA-DQ and FcεRIα for cDC2s allowed for their specific identification in blood and tissues. We also showed that cDC2s could be subdivided into phenotypically and functionally distinct subsets based on CD5, CD163, and CD14 expression, including a distinct subset of circulating inflammatory CD5-CD163+CD14+ cells related to previously defined DC3s. These inflammatory DC3s were expanded in systemic lupus erythematosus patients and correlated with disease activity. These findings further unravel the heterogeneity of DC subpopulations in health and disease and may pave the way for the identification of specific DC subset-targeting therapies.
Collapse
|
43
|
Lee YS, Radford KJ. The role of dendritic cells in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:123-178. [PMID: 31810552 DOI: 10.1016/bs.ircmb.2019.07.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy harnesses the ability of the immune system to recognize and eliminate cancer. The potent ability of dendritic cells (DCs) to initiate and regulate adaptive immune responses underpins the successful generation of anti-tumor immune responses. DCs are a heterogeneous leukocyte population comprised of distinct subsets that drive specific types of immune responses. Understanding how DCs induce tumor immune responses and the mechanisms adopted by tumors to evade DC surveillance is essential to render immunotherapies more effective. This review discusses current knowledge of the roles played by different DC subsets in human cancer and how these might be manipulated as new immunotherapeutics to improve CD8+ T cell-mediated immune responses, with a particular focus on the conventional type 1 DCs (cDC1).
Collapse
Affiliation(s)
- Yoke Seng Lee
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Kristen J Radford
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia.
| |
Collapse
|
44
|
Bros M, Haas K, Moll L, Grabbe S. RhoA as a Key Regulator of Innate and Adaptive Immunity. Cells 2019; 8:cells8070733. [PMID: 31319592 PMCID: PMC6678964 DOI: 10.3390/cells8070733] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
RhoA is a ubiquitously expressed cytoplasmic protein that belongs to the family of small GTPases. RhoA acts as a molecular switch that is activated in response to binding of chemokines, cytokines, and growth factors, and via mDia and the ROCK signaling cascade regulates the activation of cytoskeletal proteins, and other factors. This review aims to summarize our current knowledge on the role of RhoA as a general key regulator of immune cell differentiation and function. The contribution of RhoA for the primary functions of innate immune cell types, namely neutrophils, macrophages, and conventional dendritic cells (DC) to (i) get activated by pathogen-derived and endogenous danger signals, (ii) migrate to sites of infection and inflammation, and (iii) internalize pathogens has been fairly established. In activated DC, which constitute the most potent antigen-presenting cells of the immune system, RhoA is also important for the presentation of pathogen-derived antigen and the formation of an immunological synapse between DC and antigen-specific T cells as a prerequisite to induce adaptive T cell responses. In T cells and B cells as the effector cells of the adaptive immune system Rho signaling is pivotal for activation and migration. More recently, mutations of Rho and Rho-modulating factors have been identified to predispose for autoimmune diseases and as causative for hematopoietic malignancies.
Collapse
Affiliation(s)
- Matthias Bros
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Katharina Haas
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Lorna Moll
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
45
|
Durand M, Walter T, Pirnay T, Naessens T, Gueguen P, Goudot C, Lameiras S, Chang Q, Talaei N, Ornatsky O, Vassilevskaia T, Baulande S, Amigorena S, Segura E. Human lymphoid organ cDC2 and macrophages play complementary roles in T follicular helper responses. J Exp Med 2019; 216:1561-1581. [PMID: 31072818 PMCID: PMC6605753 DOI: 10.1084/jem.20181994] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/05/2019] [Accepted: 04/16/2019] [Indexed: 01/05/2023] Open
Abstract
CD4+ T follicular helper (Tfh) cells are essential for inducing efficient humoral responses. T helper polarization is classically orientated by dendritic cells (DCs), which are composed of several subpopulations with distinct functions. Whether human DC subsets display functional specialization for Tfh polarization remains unclear. Here we find that tonsil cDC2 and CD14+ macrophages are the best inducers of Tfh polarization. This ability is intrinsic to the cDC2 lineage but tissue dependent for macrophages. We further show that human Tfh cells comprise two effector states producing either IL-21 or CXCL13. Distinct mechanisms drive the production of Tfh effector molecules, involving IL-12p70 for IL-21 and activin A and TGFβ for CXCL13. Finally, using imaging mass cytometry, we find that tonsil CD14+ macrophages localize in situ in the B cell follicles, where they can interact with Tfh cells. Our results indicate that human lymphoid organ cDC2 and macrophages play complementary roles in the induction of Tfh responses.
Collapse
Affiliation(s)
- Mélanie Durand
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
- Université Paris Descartes, Paris, France
| | - Thomas Walter
- Mines ParisTech, Paris-Sciences-et-Lettres Research University, Center for Computational Biology, Paris, France
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U900, Paris, France
| | - Tiphène Pirnay
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
| | - Thomas Naessens
- Target and Translational Science, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Paul Gueguen
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
- Université Paris Descartes, Paris, France
| | - Christel Goudot
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
| | - Sonia Lameiras
- Institut Curie, Paris-Sciences-et-Lettres Research University, Next Generation Sequencing Platform, Paris, France
| | | | | | | | | | - Sylvain Baulande
- Institut Curie, Paris-Sciences-et-Lettres Research University, Next Generation Sequencing Platform, Paris, France
| | - Sebastian Amigorena
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
| | - Elodie Segura
- Institut Curie, Paris-Sciences-et-Lettres Research University, Institut National de la Santé et de la Recherche Médicale, U932, Paris, France
| |
Collapse
|
46
|
Bernelin-Cottet C, Urien C, McCaffrey J, Collins D, Donadei A, McDaid D, Jakob V, Barnier-Quer C, Collin N, Bouguyon E, Bordet E, Barc C, Boulesteix O, Leplat JJ, Blanc F, Contreras V, Bertho N, Moore AC, Schwartz-Cornil I. Electroporation of a nanoparticle-associated DNA vaccine induces higher inflammation and immunity compared to its delivery with microneedle patches in pigs. J Control Release 2019; 308:14-28. [PMID: 31265882 DOI: 10.1016/j.jconrel.2019.06.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022]
Abstract
DNA vaccination is an attractive technology, based on its well-established manufacturing process, safety profile, adaptability to rapidly combat pandemic pathogens, and stability at ambient temperature; however an optimal delivery method of DNA remains to be determined. As pigs are a relevant model for humans, we comparatively evaluated the efficiency of vaccine DNA delivery in vivo to pigs using dissolvable microneedle patches, intradermal inoculation with needle (ID), surface electroporation (EP), with DNA associated or not to cationic poly-lactic-co-glycolic acid nanoparticles (NPs). We used a luciferase encoding plasmid (pLuc) as a reporter and vaccine plasmids encoding antigens from the Porcine Reproductive and Respiratory Syndrome Virus (PRRSV), a clinically-significant swine arterivirus. Patches were successful at inducing luciferase expression in skin although at lower level than EP. EP induced the cutaneaous recruitment of granulocytes, of MHC2posCD172Apos myeloid cells and type 1 conventional dendritic cells, in association with local production of IL-1β, IL-8 and IL-17; these local responses were more limited with ID and undetectable with patches. The addition of NP to EP especially promoted the recruitment of the MHC2posCD172Apos CD163int and CD163neg myeloid subsets. Notably we obtained the strongest and broadest IFNγ T-cell response against a panel of PRRSV antigens with DNA + NPs delivered by EP, whereas patches and ID were ineffective. The anti-PRRSV IgG responses were the highest with EP administration independently of NPs, mild with ID, and undetectable with patches. These results contrast with the immunogenicity and efficacy previously induced in mice with patches. This study concludes that successful DNA vaccine administration in skin can be achieved in pigs with electroporation and patches, but only the former induces local inflammation, humoral and cellular immunity, with the highest potency when NPs were used. This finding shows the importance of evaluating the delivery and immunogenicity of DNA vaccines beyond the mouse model in a preclinical model relevant to human such as pig and reveals that EP with DNA combined to NP induces strong immunogenicity.
Collapse
Affiliation(s)
| | - Céline Urien
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Joanne McCaffrey
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | - Damien Collins
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | - Agnese Donadei
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | | | - Virginie Jakob
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Christophe Barnier-Quer
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Nicolas Collin
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Edwige Bouguyon
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Elise Bordet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | | | | | - Jean-Jacques Leplat
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Fany Blanc
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Vanessa Contreras
- Immunology of viral infections and autoimmune diseases, IDMIT Department, IBFJ, INSERM U1184-CEA - Université Paris Sud 11, Fontenay-Aux-Roses et Le Kremlin-Bicêtre, France
| | - Nicolas Bertho
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France; BIOEPAR, Oniris, INRA, 44307 Nantes, France
| | - Anne C Moore
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | | |
Collapse
|
47
|
O'Brien LJ, Guillerey C, Radford KJ. Can Dendritic Cell Vaccination Prevent Leukemia Relapse? Cancers (Basel) 2019; 11:cancers11060875. [PMID: 31234526 PMCID: PMC6627518 DOI: 10.3390/cancers11060875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/20/2019] [Indexed: 01/02/2023] Open
Abstract
Leukemias are clonal proliferative disorders arising from immature leukocytes in the bone marrow. While the advent of targeted therapies has improved survival in certain subtypes, relapse after initial therapy is a major problem. Dendritic cell (DC) vaccination has the potential to induce tumor-specific T cells providing long-lasting, anti-tumor immunity. This approach has demonstrated safety but limited clinical success until recently, as DC vaccination faces several barriers in both solid and hematological malignancies. Importantly, vaccine-mediated stimulation of protective immune responses is hindered by the aberrant production of immunosuppressive factors by cancer cells which impede both DC and T cell function. Leukemias present the additional challenge of severely disrupted hematopoiesis owing to both cytogenic defects in hematopoietic progenitors and an abnormal hematopoietic stem cell niche in the bone marrow; these factors accentuate systemic immunosuppression and DC malfunction. Despite these obstacles, several recent clinical trials have caused great excitement by extending survival in Acute Myeloid Leukemia (AML) patients through DC vaccination. Here, we review the phenotype and functional capacity of DCs in leukemia and approaches to harness DCs in leukemia patients. We describe the recent clinical successes in AML and detail the multiple new strategies that might enhance prognosis in AML and other leukemias.
Collapse
Affiliation(s)
- Liam J O'Brien
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Camille Guillerey
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
48
|
Wylie B, Macri C, Mintern JD, Waithman J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers (Basel) 2019; 11:E521. [PMID: 30979057 PMCID: PMC6521027 DOI: 10.3390/cancers11040521] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 12/16/2022] Open
Abstract
Inducing effective anti-tumor immunity has become a major therapeutic strategy against cancer. Dendritic cells (DC) are a heterogenous population of antigen presenting cells that infiltrate tumors. While DC play a critical role in the priming and maintenance of local immunity, their functions are often diminished, or suppressed, by factors encountered in the tumor microenvironment. Furthermore, DC populations with immunosuppressive activities are also recruited to tumors, limiting T cell infiltration and promoting tumor growth. Anti-cancer therapies can impact the function of tumor-associated DC and/or alter their phenotype. Therefore, the design of effective anti-cancer therapies for clinical translation should consider how best to boost tumor-associated DC function to drive anti-tumor immunity. In this review, we discuss the different subsets of tumor-infiltrating DC and their role in anti-tumor immunity. Moreover, we describe strategies to enhance DC function within tumors and harness these cells for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Ben Wylie
- Phylogica, Harry Perkins Institute, QEII Medical Centre, Nedlands, WA 6009, Australia.
| | - Christophe Macri
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21, Molecular Science and Biotechnology Institute, Parkville, VIC 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21, Molecular Science and Biotechnology Institute, Parkville, VIC 3010, Australia.
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Northern Entrance, Perth Children's Hospital, Nedlands, WA 6009, Australia.
| |
Collapse
|