1
|
Tran Ngoc Minh T, Verleng LJ, Schrama E, Busselaar J, Staal MD, de Vries E, Anholts JDH, Berkers CR, Borst J, Zaal EA, de Kivit S. CD27 costimulation supports metabolic fitness of CD4+ T cells by enhancing de novo nucleotide and protein synthesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf075. [PMID: 40324771 DOI: 10.1093/jimmun/vkaf075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/06/2025] [Indexed: 05/07/2025]
Abstract
T cells undergo many metabolic changes throughout the different phases of their response in lymphoid and nonlymphoid tissues. Cell metabolism meets demands for energy and biosynthesis, particularly during cell division and effector differentiation. As costimulatory receptors, CD28 and various TNF receptor (TNFR) family members shape T-cell clonal expansion, survival and effector functions and are important clinical targets. While CD28 is acknowledged as a metabolic regulator, little is known about how TNFRs shape T-cell metabolism. We here identify TNFR family member CD27 as a metabolic regulator in activated human CD4+ T cells. In the context of CD3 signaling and CD28 costimulation, CD27 proved to regulate specific metabolic functions, as determined by metabolomics and metabolic tracer experiments. CD27 costimulation supported upregulation of glycolysis, the pentose phosphate pathway and the TCA cycle, increasing the use of glucose-derived carbon and glutamine-derived nitrogen as building blocks for de novo nucleotide synthesis. It also promoted uptake of amino acids (AAs) and modulated pathways of AA metabolism. Accordingly, CD27 costimulation boosted protein translation in CD3- and CD3/CD28-activated CD4+ T cells, which proceeded via enhanced mTOR pathway activation. Remarkably, CD27, OX40 and 4-1BB all enhanced CD3-induced mTOR signaling, but only CD27 could overrule inhibitory PD-1 signaling. CD27 costimulation increased IL-2, IFNγ and TNFα production by CD3-activated CD4+ T cells, also in presence of PD-1 signaling. Next to previously defined beneficial effects of CD27 on activated T-cell survival and CTL differentiation and Th1 effector differentiation, these data support its essential contribution to T-cell metabolism and its relevance as a therapeutic target.
Collapse
Affiliation(s)
- Thi Tran Ngoc Minh
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Lotte J Verleng
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Ellen Schrama
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Julia Busselaar
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Mo D Staal
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Evert de Vries
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacqueline D H Anholts
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Celia R Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jannie Borst
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Esther A Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Sander de Kivit
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
2
|
Doherty CM, Patterson PR, Emeanuwa JA, Belmares Ortega J, Fox BA, Bzik DJ, Denkers EY. T lymphocyte-dependent IL-10 down-regulates a cytokine storm driven by Toxoplasma gondii GRA24. mBio 2024; 15:e0145524. [PMID: 39440975 PMCID: PMC11559025 DOI: 10.1128/mbio.01455-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
As a model organism in the study of immunity to infection, Toxoplasma gondii has been instrumental in establishing key principles of host anti-microbial defense and its regulation. Here, we employed an attenuated uracil auxotroph strain of Type I Toxoplasma designated OMP to further untangle the early immune response to this parasitic pathogen. Experiments using αβ T cell-deficient Tcrb-/- mice unexpectedly revealed that an intact αβ T lymphocyte compartment was essential to survive infection with OMP. Subsequent antibody depletion and knockout mouse experiments demonstrated contributions from CD4+ T cells and most predominantly CD8+ T cells in resistance. Using transgenic knockout mice, we found only a partial requirement for IFN-γ and a lack of requirement for Toll-like receptor (TLR) adaptor MyD88 in resistance. In contrast to other studies on Toxoplasma, the ability to survive OMP infection did not require IL-12p40. Surprisingly, T cell-dependent IL-10 was found to be critical for survival, and deficiency of this cytokine triggered an abnormally high systemic inflammatory response. We also found that parasite molecule GRA24, a dense granule protein that triggers TLR-independent IL-12 production, acts as a virulence factor contributing to death of OMP-infected Tcrb-/- and IL-10-/- mice. Furthermore, resistance against OMP was restored in Tcrb-/- mice via monoclonal depletion of IL-12p40, suggesting that GRA24-induced IL-12 underlies the fatal immunopathology observed. Collectively, our studies provide insight into a novel and rapidly arising T lymphocyte-dependent anti-inflammatory response to T. gondii which operates independently of MyD88 and IL-12 and that depends on the function of parasite-dense granule protein GRA24.IMPORTANCEAs a model infectious microbe and an important human pathogen, the apicomplexan Toxoplasma gondii has provided many important insights into innate and adaptive immunity to infection. We show here that a low virulence uracil auxotrophic Toxoplasma strain emerges as a virulent parasite in the absence of an intact T cell compartment. Both CD4+ and CD8+ T lymphocytes are required for optimal protection, in line with previous findings in other models of Toxoplasma infection. Nevertheless, several novel aspects of the response were identified in our study. Protection occurs independently of IL-12 and MyD88 and only partially requires IFN-γ. This is noteworthy particularly because the cytokines IL-12 and IFN-γ have previously been regarded as essential for protective immunity to T. gondii. Instead, we identified the anti-inflammatory effects of T cell-dependent IL-10 as the critical factor enabling host survival. The parasite dense granule protein GRA24, a host-directed mitogen-activated protein kinase activator, was identified as a major virulence factor in T cell-deficient hosts. Collectively, our results provide new and unexpected insights into host resistance to Toxoplasma.
Collapse
Affiliation(s)
- Claire M. Doherty
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Paige R. Patterson
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Julie A. Emeanuwa
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Jessica Belmares Ortega
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Eric Y. Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
3
|
Cheang NYZ, Tan KS, Tan PS, Purushotorma K, Yap WC, Tullett KM, Chua BYL, Yeoh AYY, Tan CQH, Qian X, Chen H, Tay DJW, Caminschi I, Tan YJ, Macary PA, Tan CW, Lahoud MH, Alonso S. Single-shot dendritic cell targeting SARS-CoV-2 vaccine candidate induces broad, durable and protective systemic and mucosal immunity in mice. Mol Ther 2024; 32:2299-2315. [PMID: 38715364 PMCID: PMC11286822 DOI: 10.1016/j.ymthe.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/06/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
Current coronavirus disease 2019 vaccines face limitations including waning immunity, immune escape by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, limited cellular response, and poor mucosal immunity. We engineered a Clec9A-receptor binding domain (RBD) antibody construct that delivers the SARS-CoV-2 RBD to conventional type 1 dendritic cells. Compared with non-targeting approaches, single dose immunization in mice with Clec9A-RBD induced far higher RBD-specific antibody titers that were sustained for up to 21 months after vaccination. Uniquely, increasing neutralizing and antibody-dependent cytotoxicity activities across the sarbecovirus family was observed, suggesting antibody affinity maturation over time. Consistently and remarkably, RBD-specific follicular T helper cells and germinal center B cells persisted up to 12 months after immunization. Furthermore, Clec9A-RBD immunization induced a durable mono- and poly-functional T-helper 1-biased cellular response that was strongly cross-reactive against SARS-CoV-2 variants of concern, including Omicron subvariants, and with a robust CD8+ T cell signature. Uniquely, Clec9A-RBD single-shot systemic immunization effectively primed RBD-specific cellular and humoral immunity in lung and resulted in significant protection against homologous SARS-CoV-2 challenge as evidenced by limited body weight loss and approximately 2 log10 decrease in lung viral loads compared with non-immunized controls. Therefore, Clec9A-RBD immunization has the potential to trigger robust and sustained, systemic and mucosal protective immunity against rapidly evolving SARS-CoV2 variants.
Collapse
Affiliation(s)
- Nicholas You Zhi Cheang
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peck Szee Tan
- Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Kiren Purushotorma
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Immunology Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wee Chee Yap
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Kirsteen McInnes Tullett
- Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Benson Yen Leong Chua
- Immunology Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aileen Ying-Yan Yeoh
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Caris Qi Hui Tan
- Histology Core Facility, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Xinlei Qian
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Immunology Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huixin Chen
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Douglas Jie Wen Tay
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Irina Caminschi
- Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Yee Joo Tan
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul Anthony Macary
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Immunology Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chee Wah Tan
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mireille Hanna Lahoud
- Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Sylvie Alonso
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Kawai A, Noda M, Hirata H, Munakata L, Matsuda T, Omata D, Takemura N, Onoe S, Hirose M, Kato T, Saitoh T, Hirai T, Suzuki R, Yoshioka Y. Lipid Nanoparticle with 1,2-Di-O-octadecenyl-3-trimethylammonium-propane as a Component Lipid Confers Potent Responses of Th1 Cells and Antibody against Vaccine Antigen. ACS NANO 2024; 18:16589-16609. [PMID: 38885198 PMCID: PMC11223497 DOI: 10.1021/acsnano.4c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Noda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruki Hirata
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lisa Munakata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Teppei Matsuda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daiki Omata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Naoki Takemura
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sakura Onoe
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mika Hirose
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsuya Saitoh
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshiro Hirai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Suzuki
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Yasuo Yoshioka
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of
Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Kirkpatrick C, Lu YCW. Deciphering CD4 + T cell-mediated responses against cancer. Mol Carcinog 2024; 63:1209-1220. [PMID: 38725218 PMCID: PMC11166516 DOI: 10.1002/mc.23730] [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: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 05/15/2024]
Abstract
It's been long thought that CD8+ cytotoxic T cells play a major role in T cell-mediated antitumor responses, whereas CD4+ T cells merely provide some assistance to CD8+ T cells as the "helpers." In recent years, numerous studies support the notion that CD4+ T cells play an indispensable role in antitumor responses. Here, we summarize and discuss the current knowledge regarding the roles of CD4+ T cells in antitumor responses and immunotherapy, with a focus on the molecular and cellular mechanisms behind these observations. These new insights on CD4+ T cells may pave the way to further optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Catherine Kirkpatrick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yong-Chen William Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
6
|
Pinto AT, Lukacs-Kornek V. The role of dendritic cells in MASH: friends or foes? Front Immunol 2024; 15:1379225. [PMID: 38650949 PMCID: PMC11033439 DOI: 10.3389/fimmu.2024.1379225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
Dendritic cells (DCs) are major antigen-presenting cells that connect innate and adaptive immunity. Hepatic DCs are less activated and contribute to maintain the tolerogenic environment of the liver under steady state. Several studies indicated DCs in metabolic dysfunction-associated steatohepatitis (MASH), representing a substantial burden on healthcare systems due to its association with liver-related morbidity and mortality. Studies highlighted the potential disease-promoting role of liver DCs in the development of MASH while other experimental systems suggested their protective role. This review discusses this controversy and the current understanding of how DCs affect the pathogenesis of MASH.
Collapse
Affiliation(s)
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| |
Collapse
|
7
|
Britsch S, Langer H, Duerschmied D, Becher T. The Evolving Role of Dendritic Cells in Atherosclerosis. Int J Mol Sci 2024; 25:2450. [PMID: 38397127 PMCID: PMC10888834 DOI: 10.3390/ijms25042450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis, a major contributor to cardiovascular morbidity and mortality, is characterized by chronic inflammation of the arterial wall. This inflammatory process is initiated and maintained by both innate and adaptive immunity. Dendritic cells (DCs), which are antigen-presenting cells, play a crucial role in the development of atherosclerosis and consist of various subtypes with distinct functional abilities. Following the recognition and binding of antigens, DCs become potent activators of cellular responses, bridging the innate and adaptive immune systems. The modulation of specific DC subpopulations can have either pro-atherogenic or atheroprotective effects, highlighting the dual pro-inflammatory or tolerogenic roles of DCs. In this work, we provide a comprehensive overview of the evolving roles of DCs and their subtypes in the promotion or limitation of atherosclerosis development. Additionally, we explore antigen pulsing and pharmacological approaches to modulate the function of DCs in the context of atherosclerosis.
Collapse
Affiliation(s)
- Simone Britsch
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Harald Langer
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Tobias Becher
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
| |
Collapse
|
8
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Fan Q, Yan R, Li Y, Lu L, Liu J, Li S, Fu T, Xue Y, Liu J, Li Z. Exploring Immune Cell Diversity in the Lacrimal Glands of Healthy Mice: A Single-Cell RNA-Sequencing Atlas. Int J Mol Sci 2024; 25:1208. [PMID: 38279208 PMCID: PMC10816500 DOI: 10.3390/ijms25021208] [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: 11/26/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
The lacrimal gland is responsible for maintaining the health of the ocular surface through the production of tears. However, our understanding of the immune system within the lacrimal gland is currently limited. Therefore, in this study, we utilized single-cell RNA sequencing and bioinformatic analysis to identify and analyze immune cells and molecules present in the lacrimal glands of normal mice. A total of 34,891 cells were obtained from the lacrimal glands of mice and classified into 18 distinct cell clusters using Seurat clustering. Within these cell populations, 26 different immune cell subpopulations were identified, including T cells, innate lymphocytes, macrophages, mast cells, dendritic cells, and B cells. Network analysis revealed complex cell-cell interactions between these immune cells, with particularly significant interactions observed among T cells, macrophages, plasma cells, and dendritic cells. Interestingly, T cells were found to be the main source of ligands for the Thy1 signaling pathway, while M2 macrophages were identified as the primary target of this pathway. Moreover, some of these immune cells were validated using immunohistological techniques. Collectively, these findings highlight the abundance and interactions of immune cells and provide valuable insights into the complexity of the lacrimal gland immune system and its relevance to associated diseases.
Collapse
Affiliation(s)
- Qiwei Fan
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Q.F.); (J.L.)
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
| | - Ruyu Yan
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Yan Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Liyuan Lu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Jiangman Liu
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Q.F.); (J.L.)
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
| | - Senmao Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Ting Fu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Jun Liu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Zhijie Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| |
Collapse
|
10
|
Shapir Itai Y, Barboy O, Salomon R, Bercovich A, Xie K, Winter E, Shami T, Porat Z, Erez N, Tanay A, Amit I, Dahan R. Bispecific dendritic-T cell engager potentiates anti-tumor immunity. Cell 2024; 187:375-389.e18. [PMID: 38242085 DOI: 10.1016/j.cell.2023.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 01/21/2024]
Abstract
Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.
Collapse
Affiliation(s)
- Yuval Shapir Itai
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Oren Barboy
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ran Salomon
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Akhiad Bercovich
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ken Xie
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eitan Winter
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tamar Shami
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ziv Porat
- Flow Cytometry Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
11
|
Wang X, Wei Y, He Z, Wang D, Zhang L, Du J, Zhang M, Jiang M, Chen N, Deng M, Li B, Song C, Chen D, Liu H, Xiao J, Liang H, Zhao H, Kong Y. CD70-induced differentiation of proinflammatory Th1/17/22/GM lymphocytes associated with disease progression and immune reconstitution during HIV infection. Emerg Microbes Infect 2023; 12:2271068. [PMID: 37824079 PMCID: PMC10606822 DOI: 10.1080/22221751.2023.2271068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
Immune overactivation is a hallmark of chronic HIV infection, which is critical to HIV pathogenesis and disease progression. The imbalance of helper T cell (Th) differentiation and subsequent cytokine dysregulation are generally considered to be the major drivers of excessive activation and inflammatory disorders in HIV infection. However, the accurate factors driving HIV-associated Th changes remained to be established. CD70, which was a costimulatory molecule, was found to increase on CD4+ T cells during HIV infection. Overexpression of CD70 on CD4+ T cells was recently reported to associate with highly pathogenic proinflammatory Th1/Th17 polarization in multiple sclerosis. Thus, the role of CD70 in the imbalance of Th polarization and immune overactivation during HIV infection needs to be investigated. Here, we found that the elevated frequency of CD70 + CD4+ T cells was negatively correlated with CD4 count and positively associated with immune activation in treatment-naïve people living with HIV (PLWH). More importantly, CD70 expression defined a population of proinflammatory Th1/17/22/GM subsets in PLWH. Blocking CD70 decreased the mRNA expression of subset-specific markers during Th1/17/22/GM polarization. Furthermore, we demonstrated that CD70 influenced the differentiation of these Th cells through STAT pathway. Finally, it was revealed that patients with a high baseline level of CD70 on CD4+ T cells exhibited a greater risk of poor immune reconstitution after antiretroviral therapy (ART) than those with low CD70. In general, our data highlighted the role of CD70 in Th1/17/22/GM differentiation during HIV infection and provided evidence for CD70 as a potential biomarker for predicting immune recovery.
Collapse
Affiliation(s)
- Xinyue Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Yuqing Wei
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Zhijiao He
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Di Wang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Leidan Zhang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Juan Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Mengyuan Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Meiqing Jiang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Na Chen
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Meiju Deng
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Bei Li
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Chuan Song
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Danying Chen
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Huan Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| | - Jiang Xiao
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hongyuan Liang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hongxin Zhao
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yaxian Kong
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Infectious Diseases, Beijing, People’s Republic of China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, People’s Republic of China
| |
Collapse
|
12
|
Clausen BE, Amon L, Backer RA, Berod L, Bopp T, Brand A, Burgdorf S, Chen L, Da M, Distler U, Dress RJ, Dudziak D, Dutertre CA, Eich C, Gabele A, Geiger M, Ginhoux F, Giusiano L, Godoy GJ, Hamouda AEI, Hatscher L, Heger L, Heidkamp GF, Hernandez LC, Jacobi L, Kaszubowski T, Kong WT, Lehmann CHK, López-López T, Mahnke K, Nitsche D, Renkawitz J, Reza RA, Sáez PJ, Schlautmann L, Schmitt MT, Seichter A, Sielaff M, Sparwasser T, Stoitzner P, Tchitashvili G, Tenzer S, Tochoedo NR, Vurnek D, Zink F, Hieronymus T. Guidelines for mouse and human DC functional assays. Eur J Immunol 2023; 53:e2249925. [PMID: 36563126 DOI: 10.1002/eji.202249925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Recent studies have provided evidence for an increasing number of phenotypically distinct conventional DC (cDC) subsets that on one hand exhibit a certain functional plasticity, but on the other hand are characterized by their tissue- and context-dependent functional specialization. Here, we describe a selection of assays for the functional characterization of mouse and human cDC. The first two protocols illustrate analysis of cDC endocytosis and metabolism, followed by guidelines for transcriptomic and proteomic characterization of cDC populations. Then, a larger group of assays describes the characterization of cDC migration in vitro, ex vivo, and in vivo. The final guidelines measure cDC inflammasome and antigen (cross)-presentation activity. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sven Burgdorf
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Luxia Chen
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Meihong Da
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Regine J Dress
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christina Eich
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Gabele
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Melanie Geiger
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Lucila Giusiano
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Gloria J Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Ahmed E I Hamouda
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lola C Hernandez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Tomasz Kaszubowski
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Wan Ting Kong
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Tamara López-López
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dominik Nitsche
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jörg Renkawitz
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Rifat A Reza
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Schlautmann
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Madeleine T Schmitt
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Malte Sielaff
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Tim Sparwasser
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venerology & Allergology, Medical University Innsbruck, Innsbruck, Austria
| | - Giorgi Tchitashvili
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Stefan Tenzer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz), Mainz, Germany
| | - Nounagnon R Tochoedo
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Damir Vurnek
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Fabian Zink
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Thomas Hieronymus
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Institute of Cell and Tumor Biology, RWTH Aachen University, Medical Faculty, Germany
| |
Collapse
|
13
|
Hu X, Jiang C, Gao Y, Xue X. Human dendritic cell subsets in the glioblastoma-associated microenvironment. J Neuroimmunol 2023; 383:578147. [PMID: 37643497 DOI: 10.1016/j.jneuroim.2023.578147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/24/2023] [Accepted: 07/05/2023] [Indexed: 08/31/2023]
Abstract
Glioblastoma (GBM) is the most aggressive type of glioma (Grade IV). The presence of cytotoxic T lymphocyte (CTLs) has been associated with improved outcomes in patients with GBM, and it is believed that the activation of CTLs by dendritic cells may play a critical role in controlling the growth of GBM. DCs are professional antigen-presenting cells (APC) that orchestrate innate and adaptive anti-GBM immunity. DCs can subsequently differentiate into plasmacytoid DCs (pDC), conventional DC1 (cDC1), conventional (cDC2), and monocyte-derived DCs (moDC) depending on environmental exposure. The different subsets of DCs exhibit varying functional capabilities in antigen presentation and T cell activation in producing an antitumor response. In this review, we focus on recent studies describing the phenotypic and functional characteristics of DC subsets in humans and their respective antitumor immunity and immunotolerance roles in the GBM-associated microenvironment. The critical components of crosstalk between DC subsets that contribute significantly to GBM-specific immune responses are also highlighted in this review with reference to the latest literature. Since DCs could be prime targets for therapeutic intervention, it is worth summarizing the relevance of DC subsets with respect to GBM-associated immunologic tolerance and their therapeutic potential.
Collapse
Affiliation(s)
- Xiaopeng Hu
- Medical Research Center, People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen 518000, China; Biosafety Level-3 Laboratory, Life Sciences Institute & Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China
| | - Chunmei Jiang
- Medical Research Center, People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen 518000, China
| | - Yang Gao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China.
| | - Xingkui Xue
- Medical Research Center, People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen 518000, China.
| |
Collapse
|
14
|
Zhao Y, Wu J, Xu H, Li Q, Zhang Y, Zhai Y, Tang M, Liu Y, Liu T, Ye Y, He M, He R, Xu Y, Zhou Z, Kan H, Zhang Y. Lead exposure suppresses the Wnt3a/β-catenin signaling to increase the quiescence of hematopoietic stem cells via reducing the expression of CD70 on bone marrow-resident macrophages. Toxicol Sci 2023; 195:123-142. [PMID: 37436718 DOI: 10.1093/toxsci/kfad067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023] Open
Abstract
Lead (Pb) is a heavy metal highly toxic to human health in the environment. The aim of this study was to investigate the mechanism of Pb impact on the quiescence of hematopoietic stem cells (HSC). WT C57BL/6 (B6) mice treated with 1250 ppm Pb via drinking water for 8 weeks had increased the quiescence of HSC in the bone marrow (BM), which was caused by the suppressed activation of the Wnt3a/β-catenin signaling. Mechanically, a synergistic action of Pb and IFNγ on BM-resident macrophages (BM-Mφ) reduced their surface expression of CD70, which thereby dampened the Wnt3a/β-catenin signaling to suppress the proliferation of HSC in mice. In addition, a joint action of Pb and IFNγ also suppressed the expression of CD70 on human Mφ to impair the Wnt3a/β-catenin signaling and reduce the proliferation of human HSC purified from umbilical cord blood of healthy donors. Moreover, correlation analyses showed that the blood Pb concentration was or tended to be positively associated with the quiescence of HSC, and was or tended to be negatively associated with the activation of the Wnt3a/β-catenin signaling in HSC in human subjects occupationally exposed to Pb. Collectively, these data indicate that an occupationally relevant level of Pb exposure suppresses the Wnt3a/β-catenin signaling to increase the quiescence of HSC via reducing the expression of CD70 on BM-Mφ in both mice and humans.
Collapse
Affiliation(s)
- Yifan Zhao
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jiaojiao Wu
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Hua Xu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Qian Li
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yufan Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yue Zhai
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Mengke Tang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yalin Liu
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Ting Liu
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yao Ye
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanyi Xu
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| | - Haidong Kan
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Krayem I, Sohrabi Y, Havelková H, Gusareva ES, Strnad H, Čepičková M, Volkova V, Kurey I, Vojtíšková J, Svobodová M, Demant P, Lipoldová M. Functionally distinct regions of the locus Leishmania major response 15 control IgE or IFNγ level in addition to skin lesions. Front Immunol 2023; 14:1145269. [PMID: 37600780 PMCID: PMC10437074 DOI: 10.3389/fimmu.2023.1145269] [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: 01/15/2023] [Accepted: 05/02/2023] [Indexed: 08/22/2023] Open
Abstract
Leishmaniasis, a disease caused by parasites of Leishmania spp., endangers more than 1 billion people living in endemic countries and has three clinical forms: cutaneous, mucocutaneous, and visceral. Understanding of individual differences in susceptibility to infection and heterogeneity of its pathology is largely lacking. Different mouse strains show a broad and heterogeneous range of disease manifestations such as skin lesions, splenomegaly, hepatomegaly, and increased serum levels of immunoglobulin E and several cytokines. Genome-wide mapping of these strain differences detected more than 30 quantitative trait loci (QTLs) that control the response to Leishmania major. Some control different combinations of disease manifestations, but the nature of this heterogeneity is not yet clear. In this study, we analyzed the L. major response locus Lmr15 originally mapped in the strain CcS-9 which carries 12.5% of the genome of the resistant strain STS on the genetic background of the susceptible strain BALB/c. For this analysis, we used the advanced intercross line K3FV between the strains BALB/c and STS. We confirmed the previously detected loci Lmr15, Lmr18, Lmr24, and Lmr27 and performed genetic dissection of the effects of Lmr15 on chromosome 11. We prepared the interval-specific recombinant strains 6232HS1 and 6229FUD, carrying two STS-derived segments comprising the peak linkage of Lmr15 whose lengths were 6.32 and 17.4 Mbp, respectively, and analyzed their response to L. major infection. These experiments revealed at least two linked but functionally distinct chromosomal regions controlling IFNγ response and IgE response, respectively, in addition to the control of skin lesions. Bioinformatics and expression analysis identified the potential candidate gene Top3a. This finding further clarifies the genetic organization of factors relevant to understanding the differences in the individual risk of disease.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Westfälische Wilhelms-Universität, Münster, Germany
| | - Helena Havelková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Elena S. Gusareva
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Hynek Strnad
- Department of Genomics and Bioinformatics, Institute of Molecular Genetics of The Czech Academy of Sciences, Prague, Czechia
| | - Marie Čepičková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Valeryia Volkova
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Iryna Kurey
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Jarmila Vojtíšková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Milena Svobodová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Peter Demant
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
16
|
Zhang S, Audiger C, Chopin M, Nutt SL. Transcriptional regulation of dendritic cell development and function. Front Immunol 2023; 14:1182553. [PMID: 37520521 PMCID: PMC10382230 DOI: 10.3389/fimmu.2023.1182553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Dendritic cells (DCs) are sentinel immune cells that form a critical bridge linking the innate and adaptive immune systems. Extensive research addressing the cellular origin and heterogeneity of the DC network has revealed the essential role played by the spatiotemporal activity of key transcription factors. In response to environmental signals DC mature but it is only following the sensing of environmental signals that DC can induce an antigen specific T cell response. Thus, whilst the coordinate action of transcription factors governs DC differentiation, sensing of environmental signals by DC is instrumental in shaping their functional properties. In this review, we provide an overview that focuses on recent advances in understanding the transcriptional networks that regulate the development of the reported DC subsets, shedding light on the function of different DC subsets. Specifically, we discuss the emerging knowledge on the heterogeneity of cDC2s, the ontogeny of pDCs, and the newly described DC subset, DC3. Additionally, we examine critical transcription factors such as IRF8, PU.1, and E2-2 and their regulatory mechanisms and downstream targets. We highlight the complex interplay between these transcription factors, which shape the DC transcriptome and influence their function in response to environmental stimuli. The information presented in this review provides essential insights into the regulation of DC development and function, which might have implications for developing novel therapeutic strategies for immune-related diseases.
Collapse
Affiliation(s)
- Shengbo Zhang
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Cindy Audiger
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michaël Chopin
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stephen L. Nutt
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
17
|
Chepngeno J, Amimo JO, Michael H, Raev SA, Jung K, Lee MV, Damtie D, Omwando A, Vlasova AN, Saif LJ. Vitamin A deficiency and vitamin A supplementation affect innate and T cell immune responses to rotavirus A infection in a conventional sow model. Front Immunol 2023; 14:1188757. [PMID: 37180172 PMCID: PMC10166828 DOI: 10.3389/fimmu.2023.1188757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/15/2023] Open
Abstract
Rotavirus A (RVA) causes ~200,000 diarrheal deaths annually in children <5yrs, mostly in low- and middle-income countries. Risk factors include nutritional status, social factors, breastfeeding status, and immunodeficiency. We evaluated the effects of vitamin A (VA) deficiency/VA supplementation and RVA exposure (anamnestic) on innate and T cell immune responses in RVA seropositive pregnant and lactating sows and passive protection of their piglets post-RVA challenge. Sows were fed VA deficient (VAD) or sufficient (VAS) diets starting at gestation day (GD)30. A subset of VAD sows received VA supplementation from GD|76 (30,000IU/day, VAD+VA). Sows (6 groups) were inoculated with porcine RVA G5P[7] (OSU strain) or Minimal Essential Medium (mock) at GD~90: VAD+RVA; VAS+RVA; VAD+VA+RVA; VAD-mock; VAS-mock; and VAD+VA-mock. Blood, milk, and gut-associated tissues were collected from sows at several time points to examine innate [natural killer (NK), dendritic (DC) cells], T cell responses and changes in genes involved in the gut-mammary gland (MG)-immunological axis trafficking. Clinical signs of RVA were evaluated post inoculation of sows and post-challenge of piglets. We observed decreased frequencies of NK cells, total and MHCII+ plasmacytoid DCs, conventional DCs, CD103+ DCs and CD4+/CD8+ and T regulatory cells (Tregs) and NK cell activity in VAD+RVA sows. Polymeric Ig receptor and retinoic acid receptor alpha (RARα) genes were downregulated in mesenteric lymph nodes and ileum of VAD+RVA sows. Interestingly, RVA-specific IFN-γ producing CD4+/CD8+ T cells were increased in VAD-Mock sows, coinciding with increased IL-22 suggesting inflammation in these sows. VA supplementation to VAD+RVA sows restored frequencies of NK cells and pDCs, and NK activity, but not tissue cDCs and blood Tregs. In conclusion, similar to our recent observations of decreased B cell responses in VAD sows that led to decreased passive immune protection of their piglets, VAD impaired innate and T cell responses in sows, while VA supplementation to VAD sows restored some, but not all responses. Our data reiterate the importance of maintaining adequate VA levels and RVA immunization in pregnant and lactating mothers to achieve optimal immune responses, efficient function of the gut-MG-immune cell-axis and to improve passive protection of their piglets.
Collapse
Affiliation(s)
- Juliet Chepngeno
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, The College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Joshua O. Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Husheem Michael
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Sergei A. Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Kwonil Jung
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Marcia V. Lee
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Debasu Damtie
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- The Ohio State University Global One Health LLC, Eastern Africa Regional Office, Addis Ababa, Ethiopia
| | - Alfred Omwando
- Department of Public Health, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, The College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, The College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
18
|
Bowakim-Anta N, Acolty V, Azouz A, Yagita H, Leo O, Goriely S, Oldenhove G, Moser M. Chronic CD27-CD70 costimulation promotes type 1-specific polarization of effector Tregs. Front Immunol 2023; 14:1023064. [PMID: 36993956 PMCID: PMC10041113 DOI: 10.3389/fimmu.2023.1023064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionMost T lymphocytes, including regulatory T cells, express the CD27 costimulatory receptor in steady state conditions. There is evidence that CD27 engagement on conventional T lymphocytes favors the development of Th1 and cytotoxic responses in mice and humans, but the impact on the regulatory lineage is unknown.MethodsIn this report, we examined the effect of constitutive CD27 engagement on both regulatory and conventional CD4+ T cells in vivo, in the absence of intentional antigenic stimulation.ResultsOur data show that both T cell subsets polarize into type 1 Tconvs or Tregs, characterized by cell activation, cytokine production, response to IFN-γ and CXCR3-dependent migration to inflammatory sites. Transfer experiments suggest that CD27 engagement triggers Treg activation in a cell autonomous fashion.ConclusionWe conclude that CD27 may regulate the development of Th1 immunity in peripheral tissues as well as the subsequent switch of the effector response into long-term memory.
Collapse
Affiliation(s)
- Natalia Bowakim-Anta
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Valérie Acolty
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Abdulkader Azouz
- Institute for Medical Immunology, Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Oberdan Leo
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Stanislas Goriely
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Institute for Medical Immunology, Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Guillaume Oldenhove
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- *Correspondence: Muriel Moser,
| |
Collapse
|
19
|
CD4 + T cells in cancer. NATURE CANCER 2023; 4:317-329. [PMID: 36894637 DOI: 10.1038/s43018-023-00521-2] [Citation(s) in RCA: 223] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 01/20/2023] [Indexed: 03/11/2023]
Abstract
Cancer immunology and immunotherapy are driving forces of research and development in oncology, mostly focusing on CD8+ T cells and the tumor microenvironment. Recent progress highlights the importance of CD4+ T cells, corresponding to the long-known fact that CD4+ T cells are central players and coordinators of innate and antigen-specific immune responses. Moreover, they have now been recognized as anti-tumor effector cells in their own right. Here we review the current status of CD4+ T cells in cancer, which hold great promise for improving knowledge and therapies in cancer.
Collapse
|
20
|
Backer RA, Probst HC, Clausen BE. Classical DC2 subsets and monocyte-derived DC: Delineating the developmental and functional relationship. Eur J Immunol 2023; 53:e2149548. [PMID: 36642930 DOI: 10.1002/eji.202149548] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/08/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023]
Abstract
To specifically tailor immune responses to a given pathogenic threat, dendritic cells (DC) are highly heterogeneous and comprise many specialized subtypes, including conventional DC (cDC) and monocyte-derived DC (MoDC), each with distinct developmental and functional characteristics. However, the functional relationship between cDC and MoDC is not fully understood, as the overlapping phenotypes of certain type 2 cDC (cDC2) subsets and MoDC do not allow satisfactory distinction of these cells in the tissue, particularly during inflammation. However, precise cDC2 and MoDC classification is required for studies addressing how these diverse cell types control immune responses and is therefore currently one of the major interests in the field of cDC research. This review will revise murine cDC2 and MoDC biology in the steady state and under inflammatory conditions and discusses the commonalities and differences between ESAMlo cDC2, inflammatory cDC2, and MoDC and their relative contribution to the initiation, propagation, and regulation of immune responses.
Collapse
Affiliation(s)
- Ronald A Backer
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hans Christian Probst
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
21
|
Stephan AS, Kosinska AD, Mück-Häusl M, Muschaweckh A, Jäger C, Röder N, Heikenwälder M, Dembek C, Protzer U. Evaluation of the Effect of CD70 Co-Expression on CD8 T Cell Response in Protein-Prime MVA-Boost Vaccination in Mice. Vaccines (Basel) 2023; 11:vaccines11020245. [PMID: 36851121 PMCID: PMC9966001 DOI: 10.3390/vaccines11020245] [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: 12/04/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Here, we investigate the potential of CD70 co-expression during viral vector boost vaccination to improve an antigen-specific T cell response. To determine the chance of activating antigen-specific T cells by CD70, we used the HBV core antigen as a model antigen in a heterologous protein-prime, Modified Vaccinia virus Ankara (MVA) boost vaccination scheme. Both the HBV core and a CD70 expression cassette were co-expressed upon delivery by an MVA vector under the same promoter linked by a P2A site. To compare immunogenicity with and without CD70 co-expression, HBV-naïve, C57BL/6 (wt) mice and HBV-transgenic mice were prime-vaccinated using recombinant HBV core antigen followed by the MVA vector boost. Co-expression of CD70 increased the number of vaccine-induced HBV core-specific CD8 T cells by >2-fold and improved their effector functions in HBV-naïve mice. In vaccinated HBV1.3tg mice, the number and functionality of HBV core-specific CD8 T cells was slightly increased upon CD70 co-expression in low-viremic, but not in high-viremic animals. CD70 co-expression did not impact liver damage as indicated by ALT levels in the serum, but increased the number of vaccine-induced, proliferative T cell clusters in the liver. Overall, this study indicates that orchestrated co-expression of CD70 and a vaccine antigen may be an interesting and safe means of enhancing antigen-specific CD8 T cell responses using vector-based vaccines, although in our study it was not sufficient to break immune tolerance.
Collapse
Affiliation(s)
- Ann-Sophie Stephan
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
| | - Anna D. Kosinska
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Martin Mück-Häusl
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
| | - Andreas Muschaweckh
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, 81675 Munich, Germany
| | - Clemens Jäger
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
| | - Natalie Röder
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
| | - Mathias Heikenwälder
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany
| | - Claudia Dembek
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
- Correspondence: (C.D.); (U.P.); Tel.: +49-89-4140-6821 (U.P.)
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, Helmholtz Zentrum München, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
- Correspondence: (C.D.); (U.P.); Tel.: +49-89-4140-6821 (U.P.)
| |
Collapse
|
22
|
Redondo-Urzainqui A, Hernández-García E, Cook ECL, Iborra S. Dendritic cells in energy balance regulation. Immunol Lett 2023; 253:19-27. [PMID: 36586424 DOI: 10.1016/j.imlet.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022]
Abstract
Besides their well-known role in initiating adaptive immune responses, several groups have studied the role of dendritic cells (DCs) in the context of chronic metabolic inflammation, such as in diet-induced obesity (DIO) or metabolic-associated fatty liver disease. DCs also have an important function in maintaining metabolic tissue homeostasis in steady-state conditions. In this review, we will briefly describe the different DC subsets, the murine models available to assess their function, and discuss the role of DCs in regulating energy balance and maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Ana Redondo-Urzainqui
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Emma Clare Laura Cook
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| |
Collapse
|
23
|
Cyran L, Serfling J, Kirschner L, Raifer H, Lohoff M, Hermanns HM, Kerstan A, Bodem J, Lutz MB. Flt3L, LIF, and IL-10 combination promotes the selective in vitro development of ESAM low cDC2B from murine bone marrow. Eur J Immunol 2022; 52:1946-1960. [PMID: 35357005 DOI: 10.1002/eji.202149663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/12/2022] [Accepted: 03/29/2022] [Indexed: 01/05/2023]
Abstract
The development of two conventional dendritic cells (DC) subsets (cDC1 and cDC2) and the plasmacytoid DC (pDC) in vivo and in cultures of bone marrow (BM) cells is mediated by the growth factor Flt3L. However, little is known about the factors that direct the development of the individual DC subsets. Here, we describe the selective in vitro generation of murine ESAMlow CD103- XCR1- CD172a+ CD11b+ cDC2 from BM by treatment with a combination of Flt3L, LIF, and IL-10 (collectively named as FL10). FL10 promotes common dendritic cell progenitors (CDP) proliferation in the cultures, similar to Flt3L and CDP sorted and cultured in FL10 generate exclusively cDC2. These cDC2 express the transcription factors Irf4, Klf4, and Notch2, and their growth is reduced using BM from Irf4-/- mice, but the expression of Batf3 and Tcf4 is low. Functionally they respond to TLR3, TLR4, and TLR9 signals by upregulation of the surface maturation markers MHC II, CD80, CD86, and CD40, while they poorly secrete proinflammatory cytokines. Peptide presentation to TCR transgenic OT-II cells induced proliferation and IFN-γ production that was similar to GM-CSF-generated BM-DC and higher than Flt3L-generated DC. Together, our data support that FL10 culture of BM cells selectively promotes CDP-derived ESAMlow cDC2 (cDC2B) development and survival in vitro.
Collapse
Affiliation(s)
- Laura Cyran
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Julia Serfling
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Luisa Kirschner
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Hartmann Raifer
- Institute of Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Michael Lohoff
- Institute of Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Heike M Hermanns
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Kerstan
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Sulczewski FB, Martino LA, Salles D, Yamamoto MM, Rosa DS, Boscardin SB. STAT3 signaling modulates the immune response induced after antigen targeting to conventional type 1 dendritic cells through the DEC205 receptor. Front Immunol 2022; 13:1006996. [PMID: 36330518 PMCID: PMC9624190 DOI: 10.3389/fimmu.2022.1006996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022] Open
Abstract
Conventional dendritic cells (cDC) are a group of antigen-presenting cells specialized in priming T cell responses. In mice, splenic cDC are divided into conventional type 1 DC (cDC1) and conventional type 2 (cDC2). cDC1 are specialized to prime the Th1 CD4+ T cell response, while cDC2 are mainly associated with the induction of follicular helper T cell responses to support germinal center formation. However, the mechanisms that control the functions of cDC1 and cDC2 are not fully understood, especially the signaling pathways that can modulate their ability to promote different CD4+ T cell responses. Here, we targeted a model antigen for cDC1 and cDC2, through DEC205 and DCIR2 receptors, respectively, to study the role of the STAT3 signaling pathway in the ability of these cells to prime CD4+ T cells. Our results show that, in the absence of the STAT3 signaling pathway, antigen targeting to cDC2 induced similar frequencies of Tfh cells between STAT3-deficient mice compared to fully competent mice. On the other hand, Th1 and Th1-like Tfh cell responses were significantly reduced in STAT3-deficient mice after antigen targeting to cDC1 via the DEC205 receptor. In summary, our results indicate that STAT3 signaling does not control the ability of cDC2 to promote Tfh cell responses after antigen targeting via the DCIR2 receptor, but modulates the function of cDC1 to promote Th1 and Th1-like Tfh T cell responses after antigen targeting via the DEC205 receptor.
Collapse
Affiliation(s)
| | - Larissa Alves Martino
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Davi Salles
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Márcio Massao Yamamoto
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Instituto de Investigação em Imunologia (iii), INCT, Sao Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
- Instituto de Investigação em Imunologia (iii), INCT, Sao Paulo, Brazil
- *Correspondence: Silvia Beatriz Boscardin,
| |
Collapse
|
25
|
Vogel I, Acolty V, Keler T, Goriely S, Leo O, Moser M. Agonistic anti-CD27 antibody ameliorates EAE by suppressing IL-17 production. Eur J Immunol 2022; 52:1620-1629. [PMID: 35856659 DOI: 10.1002/eji.202149698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 06/17/2021] [Accepted: 07/18/2022] [Indexed: 12/14/2022]
Abstract
CD27/CD70 costimulation enhances T-cell survival, memory formation and Th1-cell differentiation and effector function. In addition to promoting Th1 responses, CD27 signaling has been shown to exert a negative regulatory role on IL-17 production, resulting in increased sensitivity of CD27 KO mice to EAE. By inducing EAE in full CD27 KO mice, and in a novel, T-cell specific CD27 KO mouse strain (CD4-Cre x CD27flox/flox ), we demonstrate herein that CD27 engagement by its natural ligand (CD70) suppresses IL-17 production in a cell autonomous fashion. We further show that CD27 engagement by an agonistic antibody given after EAE induction or at symptom onset similarly suppresses IL-17 production by activated CD4+ T cells infiltrating the inflamed CNS while IFN-γ production was unaffected, leading to an amelioration of inflammatory-related symptoms. These findings propose CD27 costimulation as a potential candidate for therapeutic manipulation to treat autoimmune and autoinflammatory diseases characterized by excessive IL-17 production.
Collapse
Affiliation(s)
- Isabel Vogel
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB) - IBMM, Rue des Prof. Jeener et Brachet 12, Gosselies, 6041, Belgium.,NeuVasQ Biotechnologies, Rue Auguste Piccard 48, Gosselies, 6041, Belgium
| | - Valerie Acolty
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB) - IBMM, Rue des Prof. Jeener et Brachet 12, Gosselies, 6041, Belgium
| | - Tibor Keler
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Stanislas Goriely
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB) - IBMM, Rue des Prof. Jeener et Brachet 12, Gosselies, 6041, Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB) - IBMM, Rue des Prof. Jeener et Brachet 12, Gosselies, 6041, Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Université Libre de Bruxelles (ULB) - IBMM, Rue des Prof. Jeener et Brachet 12, Gosselies, 6041, Belgium
| |
Collapse
|
26
|
Guelen L, Fischmann TO, Wong J, Mauze S, Guadagnoli M, Bąbała N, Wagenaars J, Juan V, Rosen D, Prosise W, Habraken M, Lodewijks I, Gu D, Stammen-Vogelzangs J, Yu Y, Baker J, Lutje Hulsik D, Driessen-Engels L, Malashock D, Kreijtz J, Bertens A, de Vries E, Bovens A, Bramer A, Zhang Y, Wnek R, Troth S, Chartash E, Dobrenkov K, Sadekova S, van Elsas A, Cheung JK, Fayadat-Dilman L, Borst J, Beebe AM, Van Eenennaam H. Preclinical characterization and clinical translation of pharmacodynamic markers for MK-5890: a human CD27 activating antibody for cancer immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-005049. [PMID: 36100308 PMCID: PMC9472132 DOI: 10.1136/jitc-2022-005049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 11/06/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICI) have radically changed cancer therapy, but most patients with cancer are unresponsive or relapse after treatment. MK-5890 is a CD27 agonist antibody intended to complement ICI therapy. CD27 is a member of the tumor necrosis factor receptor superfamily that plays a critical role in promoting responses of T cells, B cells and NK cells. Methods Anti-CD27 antibodies were generated and selected for agonist activity using NF-кB luciferase reporter assays. Antibodies were humanized and characterized for agonism using in vitro T-cell proliferation assays. The epitope recognized on CD27 by MK-5890 was established by X-ray crystallography. Anti-tumor activity was evaluated in a human CD27 knock-in mouse. Preclinical safety was tested in rhesus monkeys. Pharmacodynamic properties were examined in mouse, rhesus monkeys and a phase 1 dose escalation clinical study in patients with cancer. Results Humanized anti-CD27 antibody MK-5890 (hIgG1) was shown to bind human CD27 on the cell surface with sub-nanomolar potency and to partially block binding to its ligand, CD70. Crystallization studies revealed that MK-5890 binds to a unique epitope in the cysteine-rich domain 1 (CRD1). MK-5890 activated CD27 expressed on 293T NF-κB luciferase reporter cells and, conditional on CD3 stimulation, in purified CD8+ T cells without the requirement of crosslinking. Functional Fc-receptor interaction was required to activate CD8+ T cells in an ex vivo tumor explant system and to induce antitumor efficacy in syngeneic murine subcutaneous tumor models. MK-5890 had monotherapy efficacy in these models and enhanced efficacy of PD-1 blockade. MK-5890 reduced in an isotype-dependent and dose-dependent manner circulating, but not tumor-infiltrating T-cell numbers in these mouse models. In rhesus monkey and human patients, reduction in circulating T cells was transient and less pronounced than in mouse. MK-5890 induced transient elevation of chemokines MCP-1, MIP-1α, and MIP-1β in the serum of mice, rhesus monkeys and patients with cancer. MK-5890 was well tolerated in rhesus monkeys and systemic exposure to MK-5890 was associated with CD27 occupancy at all doses. Conclusions MK-5890 is a novel CD27 agonistic antibody with the potential to complement the activity of PD-1 checkpoint inhibition in cancer immunotherapy and is currently undergoing clinical evaluation.
Collapse
Affiliation(s)
- Lars Guelen
- BioNovion/Aduro Biotech Europe, Oss, The Netherlands
| | - Thierry O Fischmann
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Jerelyn Wong
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - Smita Mauze
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | | | - Nikolina Bąbała
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Veronica Juan
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - David Rosen
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - Winnie Prosise
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, Kenilworth, New Jersey, USA
| | | | | | - Danling Gu
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | | | - Ying Yu
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - Jeanne Baker
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | | | | | - Dan Malashock
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - Joost Kreijtz
- BioNovion/Aduro Biotech Europe, Oss, The Netherlands
| | | | - Evert de Vries
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Astrid Bovens
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Arne Bramer
- BioNovion/Aduro Biotech Europe, Oss, The Netherlands
| | - Yiwei Zhang
- Clinical Development, Merck & Co Inc, Rahway, New Jersey, USA
| | - Richard Wnek
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Sean Troth
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, West Point, Pennsylvania, USA
| | - Elliot Chartash
- Clinical Development, Merck & Co Inc, Rahway, New Jersey, USA
| | | | - Svetlana Sadekova
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | | | - Jason K Cheung
- Process Research and Development, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Laurence Fayadat-Dilman
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | - Jannie Borst
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Amy M Beebe
- Discovery, Preclinical and Translational Medicine, Merck & Co Inc, South San Francisco, California, USA
| | | |
Collapse
|
27
|
Connors J, Taramangalam B, Cusimano G, Bell MR, Matt SM, Runner K, Gaskill PJ, DeFilippis V, Nikolich-Žugich J, Kutzler MA, Haddad EK. Aging alters antiviral signaling pathways resulting in functional impairment in innate immunity in response to pattern recognition receptor agonists. GeroScience 2022; 44:2555-2572. [PMID: 35849213 PMCID: PMC9289086 DOI: 10.1007/s11357-022-00612-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/16/2022] [Indexed: 01/06/2023] Open
Abstract
The progressive impairment of immunity to pathogens and vaccines with aging is a significant public health problem as the world population shifts to an increased percentage of older adults (> 65). We have previously demonstrated that cells obtained from older volunteers have delayed and defective induction of type I interferons and T cell and B cell helper cytokines in response to TLR ligands when compared to those from adult subjects. However, the underlying intracellular mechanisms are not well described. Herein, we studied two critical pathways important in the production of type I interferon (IFN), the interferon response factor 7 (pIRF7), and TANK-binding kinase (pTBK-1). We show a decrease in pIRF7 and pTBK-1 in cross-priming dendritic cells (cDC1s), CD4+ T cell priming DCs (cDC2s), and CD14dimCD16+ vascular patrolling monocytes from older adults (n = 11) following stimulation with pathway-specific agonists in comparison with young individuals (n = 11). The decrease in these key antiviral pathway proteins correlates with decreased phagocytosis, suggesting impaired function in Overall, our findings describe molecular mechanisms which explain the innate functional impairment in older adults and thus could inform us of novel approaches to restore these defects.
Collapse
Affiliation(s)
- Jennifer Connors
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Bhavani Taramangalam
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Gina Cusimano
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Matthew R. Bell
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Stephanie M. Matt
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Kaitlyn Runner
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Peter J. Gaskill
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Victor DeFilippis
- grid.5288.70000 0000 9758 5690Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR USA
| | - Janko Nikolich-Žugich
- grid.134563.60000 0001 2168 186XDepartment of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,grid.134563.60000 0001 2168 186XArizona Center On Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ USA
| | - Michele A. Kutzler
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Elias K. Haddad
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| |
Collapse
|
28
|
Nerb B, Dudziak D, Gessner A, Feuerer M, Ritter U. Have We Ignored Vector-Associated Microbiota While Characterizing the Function of Langerhans Cells in Experimental Cutaneous Leishmaniasis? FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.874081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
29
|
The Role of Type-2 Conventional Dendritic Cells in the Regulation of Tumor Immunity. Cancers (Basel) 2022; 14:cancers14081976. [PMID: 35454882 PMCID: PMC9028336 DOI: 10.3390/cancers14081976] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Recent studies revealed that type-2 conventional dendritic cells (cDC2s) play an important role in antitumor immunity by promoting cytotoxic T-cell responses and helper T-cell differentiation. This review outlines the role of cDC2s in tumor immunity and summarizes the latest progress regarding their potential in cancer vaccination and cDC2-targeted cancer immunotherapy. Abstract Conventional dendritic cells (cDCs) orchestrate immune responses to cancer and comprise two major subsets: type-1 cDCs (cDC1s) and type-2 cDCs (cDC2s). Compared with cDC1s, which are dedicated to the activation of CD8+ T cells, cDC2s are ontogenically and functionally heterogeneous, with their main function being the presentation of exogenous antigens to CD4+ T cells for the initiation of T helper cell differentiation. cDC1s play an important role in tumor-specific immune responses through cross-presentation of tumor-derived antigens for the priming of CD8+ T cells, whereas little is known of the role of cDC2s in tumor immunity. Recent studies have indicated that human cDC2s can be divided into at least two subsets and have implicated these cells in both anti- and pro-tumoral immune responses. Furthermore, the efficacy of cDC2-based vaccines as well as cDC2-targeted therapeutics has been demonstrated in both mouse models and human patients. Here we summarize current knowledge about the role of cDC2s in tumor immunity and address whether these cells are beneficial in the context of antitumor immune responses.
Collapse
|
30
|
STxB as an Antigen Delivery Tool for Mucosal Vaccination. Toxins (Basel) 2022; 14:toxins14030202. [PMID: 35324699 PMCID: PMC8948715 DOI: 10.3390/toxins14030202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy against cancer and infectious disease holds the promise of high efficacy with minor side effects. Mucosal vaccines to protect against tumors or infections disease agents that affect the upper airways or the lung are still lacking, however. One mucosal vaccine candidate is the B-subunit of Shiga toxin, STxB. In this review, we compare STxB to other immunotherapy vectors. STxB is a non-toxic protein that binds to a glycosylated lipid, termed globotriaosylceramide (Gb3), which is preferentially expressed by dendritic cells. We review the use of STxB for the cross-presentation of tumor or viral antigens in a MHC class I-restricted manner to induce humoral immunity against these antigens in addition to polyfunctional and persistent CD4+ and CD8+ T lymphocytes capable of protecting against viral infection or tumor growth. Other literature will be summarized that documents a powerful induction of mucosal IgA and resident memory CD8+ T cells against mucosal tumors specifically when STxB-antigen conjugates are administered via the nasal route. It will also be pointed out how STxB-based vaccines have been shown in preclinical cancer models to synergize with other therapeutic modalities (immune checkpoint inhibitors, anti-angiogenic therapy, radiotherapy). Finally, we will discuss how molecular aspects such as low immunogenicity, cross-species conservation of Gb3 expression, and lack of toxicity contribute to the competitive positioning of STxB among the different DC targeting approaches. STxB thereby appears as an original and innovative tool for the development of mucosal vaccines in infectious diseases and cancer.
Collapse
|
31
|
Gene Expression of CD70 and CD27 Is Increased in Alopecia Areata Lesions and Associated with Disease Severity and Activity. Dermatol Res Pract 2022; 2022:5004642. [PMID: 35300124 PMCID: PMC8923777 DOI: 10.1155/2022/5004642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background Alopecia areata (AA) is an acquired hair loss disorder induced by a cell-mediated autoimmune attack against anagen hair follicles. CD27-CD70 is a receptor-ligand complex which enhances T helper and cytotoxic T cell activation, survival, and proliferation. The overstimulation of this complex can lead to a lack of tolerance and the development of autoimmunity. Objectives This study aimed to assess the gene expression of CD27 and CD70 in patients with AA. Methods CD70 and CD27 mRNA expressions were evaluated by a quantitative real-time polymerase chain reaction in scalp biopsies from 40 AA patients (both AA lesions and non-lesional areas) and 40 healthy controls (HCs). The Severity of Alopecia Tool (SALT) score was used to assess AA severity. Patients were evaluated for signs of AA activity, including a positive hair pull test and dermoscopic features of black dots, broken hairs, and tapering hairs. Results The gene expression of CD70 and CD27 was significantly higher in AA lesions than in non-lesional areas (p < 0.001 for both) and HCs (p=0.004, p=0.014, respectively). There were significant positive correlations between AA severity and gene expression of CD70 (p < 0.001) and CD27 (p=0.030) in AA lesions. Significant associations were detected between signs of AA activity and lesional gene expression of CD70 and CD27. Additionally, CD70 and CD27 gene expression was significantly lower in non-lesional biopsies compared to HCs (p < 0.001). Conclusion Gene expression of CD70 and CD27 was increased in AA lesions and was associated with disease severity and activity. Thus, both molecules can be a predictor of AA severity and activity. Furthermore, the expression was reduced in non-lesional scalp areas. Thus, a lack of CD27 and CD70 expression may initially predispose to immunological dysregulation and the development of AA.
Collapse
|
32
|
Tesfaye DY, Bobic S, Lysén A, Huszthy PC, Gudjonsson A, Braathen R, Bogen B, Fossum E. Targeting Xcr1 on Dendritic Cells Rapidly Induce Th1-Associated Immune Responses That Contribute to Protection Against Influenza Infection. Front Immunol 2022; 13:752714. [PMID: 35296089 PMCID: PMC8918470 DOI: 10.3389/fimmu.2022.752714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Targeting antigen to conventional dendritic cells (cDCs) can improve antigen-specific immune responses and additionally be used to influence the polarization of the immune responses. However, the mechanisms by which this is achieved are less clear. To improve our understanding, we here evaluate molecular and cellular requirements for CD4+ T cell and antibody polarization after immunization with Xcl1-fusion vaccines that specifically target cDC1s. Xcl1-fusion vaccines induced an IgG2a/IgG2b-dominated antibody response and rapid polarization of Th1 cells both in vitro and in vivo. For comparison, we included fliC-fusion vaccines that almost exclusively induced IgG1, despite inducing a more mixed polarization of T cells. Th1 polarization and IgG2a induction with Xcl1-fusion vaccines required IL-12 secretion but were nevertheless maintained in BATF3-/- mice which lack IL-12-secreting migratory DCs. Interestingly, induction of IgG2a-dominated responses was highly dependent on the early kinetics of Th1 induction and was important for optimal protection in an influenza infection model. Early Th1 induction was dominant, since a combined Xcl1- and fliC-fusion vaccine induced IgG2a/IgG2b polarized antibody responses similar to Xcl1-fusion vaccines alone. In summary, our results demonstrate that targeting antigen to Xcr1+ cDC1s is an efficient strategy for enhancing IgG2a antibody responses through rapid Th1 induction, which can be utilized for improved vaccine design.
Collapse
Affiliation(s)
- Demo Yemane Tesfaye
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Sonja Bobic
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anna Lysén
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Peter Csaba Huszthy
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Center for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Arnar Gudjonsson
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ranveig Braathen
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Even Fossum
- Department of Immunology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
- Kristian Gerhard Jebsen Center for Research on Influenza Vaccines, University of Oslo and Oslo University Hospital, Oslo, Norway
- *Correspondence: Even Fossum,
| |
Collapse
|
33
|
Shen M, Du Y, Ye Y. Tumor-associated macrophages, dendritic cells, and neutrophils: biological roles, crosstalk, and therapeutic relevance. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:222-243. [PMID: 37724296 PMCID: PMC10388790 DOI: 10.1515/mr-2021-0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Tumor-associated myeloid cells constitute a series of plastic and heterogeneous cell populations within the tumor microenvironment (TME), and exhibit different phenotypes and functions in response to various microenvironmental signals. In light of promising preclinical data indicating that myeloid-based therapy can effectively suppress tumor growth, a series of novel immune-based therapies and approaches are currently undergoing clinical evaluation. A better understanding of the diversity and functional roles of different myeloid cell subtypes and of how they are associated with TME remodeling may help to improve cancer therapy. Herein, we focus on myeloid cells and discuss how tumor cells can simultaneously reprogram these cells through tumor-derived factors and metabolites. In addition, we discuss the interactions between myeloid cells and other cells in the TME that have the potential to directly or indirectly regulate tumor initiation, invasion, or angiogenesis. We further discuss the current and future potential applications of myeloid cells in the development of focused therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Mingyi Shen
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhua Du
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Sa-nguanmoo N, Namdee K, Khongkow M, Ruktanonchai U, Zhao Y, Liang XJ. Review: Development of SARS-CoV-2 immuno-enhanced COVID-19 vaccines with nano-platform. NANO RESEARCH 2021; 15:2196-2225. [PMID: 34659650 PMCID: PMC8501370 DOI: 10.1007/s12274-021-3832-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Vaccination is the most effective way to prevent coronavirus disease 2019 (COVID-19). Vaccine development approaches consist of viral vector vaccines, DNA vaccine, RNA vaccine, live attenuated virus, and recombinant proteins, which elicit a specific immune response. The use of nanoparticles displaying antigen is one of the alternative approaches to conventional vaccines. This is due to the fact that nano-based vaccines are stable, able to target, form images, and offer an opportunity to enhance the immune responses. The diameters of ultrafine nanoparticles are in the range of 1-100 nm. The application of nanotechnology on vaccine design provides precise fabrication of nanomaterials with desirable properties and ability to eliminate undesirable features. To be successful, nanomaterials must be uptaken into the cell, especially into the target and able to modulate cellular functions at the subcellular levels. The advantages of nano-based vaccines are the ability to protect a cargo such as RNA, DNA, protein, or synthesis substance and have enhanced stability in a broad range of pH, ambient temperatures, and humidity for long-term storage. Moreover, nano-based vaccines can be engineered to overcome biological barriers such as nonspecific distribution in order to elicit functions in antigen presenting cells. In this review, we will summarize on the developing COVID-19 vaccine strategies and how the nanotechnology can enhance antigen presentation and strong immunogenicity using advanced technology in nanocarrier to deliver antigens. The discussion about their safe, effective, and affordable vaccines to immunize against COVID-19 will be highlighted.
Collapse
Affiliation(s)
- Nawamin Sa-nguanmoo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - YongXiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning, 530021 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
35
|
Zhou L, Liu H, Liu K, Wei S. Gold Compounds and the Anticancer Immune Response. Front Pharmacol 2021; 12:739481. [PMID: 34588987 PMCID: PMC8473785 DOI: 10.3389/fphar.2021.739481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Gold compounds are not only well-explored for cytotoxic effects on tumors, but are also known to interact with the cancer immune system. The immune system deploys innate and adaptive mechanisms to protect against pathogens and prevent malignant transformation. The combined action of gold compounds with the activated immune system has shown promising results in cancer therapy through in vivo and in vitro experiments. Gold compounds are known to induce innate immune responses; however, these responses may contribute to adaptive immune responses. Gold compounds play the role of a major hapten that acts synergistically in innate immunity. Gold compounds support cancer cell antigenicity and promote anti-tumor immune response by inducing the release of CRT, ATP, HMGB1, HSP, and NKG2D to enhance immunogenicity. Gold compounds affect various immune cells (including suppressor regulatory T cells), inhibit myeloid derived suppressor cells, and enhance the function and number of dendritic cells. Gold nanoparticles (AuNPs) have potential for improving the effect of immunotherapy and reducing the toxicity and side effects of the treatment process. Thus, AuNPs provide an ideal opportunity for exploring the combination of anticancer gold compounds and immunotherapeutic interventions.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Amaral-Silva D, Gonçalves R, Torrão RC, Torres R, Falcão S, Gonçalves MJ, Araújo MP, Martins MJ, Lopes C, Neto A, Marona J, Costa T, Castelão W, Silva AB, Silva I, Lourenço MH, Mateus M, Gonçalves NP, Manica S, Costa M, Pimentel-Santos FM, Mourão AF, Branco JC, Soares H. Direct tissue-sensing reprograms TLR4 + Tfh-like cells inflammatory profile in the joints of rheumatoid arthritis patients. Commun Biol 2021; 4:1135. [PMID: 34580414 PMCID: PMC8476501 DOI: 10.1038/s42003-021-02659-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
CD4+ T cells mediate rheumatoid arthritis (RA) pathogenesis through both antibody-dependent and independent mechanisms. It remains unclear how synovial microenvironment impinges on CD4+ T cells pathogenic functions. Here, we identified a TLR4+ follicular helper T (Tfh) cell-like population present in the blood and expanded in synovial fluid. TLR4+ T cells possess a two-pronged pathogenic activity whereby direct TLR4+ engagement by endogenous ligands in the arthritic joint reprograms them from an IL-21 response, known to sponsor antibody production towards an IL-17 inflammatory program recognized to fuel tissue damage. Ex vivo, synovial fluid TLR4+ T cells produced IL-17, but not IL-21. Blocking TLR4 signaling with a specific inhibitor impaired IL-17 production in response to synovial fluid recognition. Mechanistically, we unveiled that T-cell HLA-DR regulates their TLR4 expression. TLR4+ T cells appear to uniquely reconcile an ability to promote systemic antibody production with a local synovial driven tissue damage program. In order to identify how the synovial microenvironment impinges on CD4+ T cells pathogenic functions in Rheumatoid Arthritis (RA), Amaral-Silva examined RA patient blood and synovial fluif and identified the presence of a TLR4+ follicular helper T (Tfh) cell-like population. They provided mechanistic insight into how TLR4+ T cells uniquely reconcile an ability to promote systemic antibody production with a local synovial driven-tissue damage program.
Collapse
Affiliation(s)
- Daniela Amaral-Silva
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rute Gonçalves
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rita C. Torrão
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rita Torres
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Sandra Falcão
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Maria João Gonçalves
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria Paula Araújo
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria José Martins
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Carina Lopes
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Agna Neto
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - José Marona
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Tiago Costa
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Walter Castelão
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Ana Bento Silva
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Inês Silva
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria Helena Lourenço
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Margarida Mateus
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Nuno Pina Gonçalves
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Santiago Manica
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Manuela Costa
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Fernando M. Pimentel-Santos
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Ana Filipa Mourão
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Jaime C. Branco
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
- grid.10772.330000000121511713CHRC|CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Rua do Instituto Bacteriológico 5, Lisboa, Portugal
| |
Collapse
|
37
|
Issabekova A, Zhumabekova M, Zhunussova M, Ogay V. The Crosstalk Between Dendritic Cells, Cytokine-Induced Killer Cells And Cancer Cells From The Perspective Of Combination Therapy. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Dendritic cells (DCs) are considered the most potent professional antigen-presenting cells (APCs) that elicit adaptive antitumour immunity. DCs integrate multiple environmental signals by efficiently processing tumour-associated antigens (TAAs) and migrating to draining lymph nodes (dLNs), where they present foreign antigens to T cells for priming. DCs thus serve as a major link between innate and adaptive immunity. Although DCs (mostly monocyte-derived DCs [mo-DCs]) have already been used in cancer therapies, such approaches have shown limited efficacy. Mo-DCs have the unique ability to present antigens to T cells in peripheral tissues. CD3+CD56+ cytokine-induced killer (CIK) cells are characterized by both MHC-restricted and MHC-unrestricted antitumour cytotoxicity against a broad range of cancer cells. This review presents an overview of the mechanisms by which mo-DCs and CIK cells’ interact with each other and with tumours. The maturation of DCs was identified as a crucial step in the development of effective DC-based vaccines against cancer. A further improved adoptive immunotherapy strategy involves a combination of mature mo-DCs and CIK cells. Combination therapy presents many opportunities for cancer treatment, as reported by a number of clinical trials. However, there is a lack of fundamental studies on the interaction of in vitro-generated mo-DCs with CIK cells. We discuss several methods of boosting DC-based vaccines and review the current knowledge of contact-dependent and cytokine-induced interactions of mo-DCs with CIK cells. We highlight that the combination of mo-DCs with CIK cells activates MHC-restricted and MHC-unrestricted immune responses.
Collapse
|
38
|
Gou S, Wang S, Liu W, Chen G, Zhang D, Du J, Yan Z, Wang H, Zhai W, Sui X, Wu Y, Qi Y, Gao Y. Adjuvant-free peptide vaccine targeting Clec9a on dendritic cells can induce robust antitumor immune response through Syk/IL-21 axis. Theranostics 2021; 11:7308-7321. [PMID: 34158852 PMCID: PMC8210616 DOI: 10.7150/thno.56406] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/02/2021] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) can process the antigens of cancer vaccine and thus stimulate the CD8+ T cells to recognize and kill the tumor cells that express these antigens. However, lack of promising carriers for presenting the antigens to DCs is one of the main barriers to the development of clinically effective cancer vaccines. Another limitation is the risk of inflammatory side effects induced by the adjuvants. It is still unclear how we can develop ideal adjuvant-free DC vaccine carriers without adjuvants. Methods: A 12-mer peptide carrier (CBP-12) with high affinity for Clec9a expressed on DCs was developed using an in silico rational optimization method. The therapeutic effects of the adjuvant-free vaccine comprising CBP-12 and exogenous or endogenous antigenic peptides were investigated in terms of antigen cross-presentation efficacy, specific cytotoxic T lymphocyte response, and antitumor activity. We also explored the mechanism involved in the antitumor effects of the adjuvant-free CBP-12 vaccine. Finally, we assessed the effects of the CBP-12 conjugated peptide vaccine combined with radiotherapy. Results: Here, we developed CBP-12 as a vaccine carrier that enhanced the uptake and cross-presentation of the antigens, thus inducing strong CD8+ T cell responses and antitumor effects in both anti-PD-1-responsive (B16-OVA) and -resistant (B16) models, even in adjuvant-free conditions. CBP-12 bound to and activated Clec9a, thereby stimulating Clec9a+ DC to product IL-21, but not IL-12 by activating of Syk. The antitumor effects of the CBP-12 conjugated peptide vaccines could be blocked by an IL-21 neutralizing antibody. We also observed the synergistic antitumor effects of the CBP-12 conjugated peptide vaccine combined with radiotherapy. Conclusions: CBP-12 could serve as an adjuvant-free peptide vaccine carrier for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Dendritic Cells/immunology
- Drug Delivery Systems
- Female
- Interleukins/genetics
- Interleukins/immunology
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Knockout
- Peptides/immunology
- Peptides/pharmacology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/immunology
- Syk Kinase/genetics
- Syk Kinase/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/pharmacology
Collapse
Affiliation(s)
- Shanshan Gou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shuai Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwen Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Dongyang Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhongyi Yan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongfei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
39
|
Sohn M, Na HY, Shin HS, Ryu SH, Park S, In H, Choi W, Park JS, Hwang S, Chu MK, Park CG. Global Gene Expression of T Cells Is Differentially Regulated by Peritoneal Dendritic Cell Subsets in an IL-2 Dependent Manner. Front Immunol 2021; 12:648348. [PMID: 34079542 PMCID: PMC8165281 DOI: 10.3389/fimmu.2021.648348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) in peripheral tissues may have a unique role to regulate innate and adaptive immune responses to antigens that enter the tissues. Peritoneal cavity is the body compartment surrounding various tissues and organs and housing diverse immune cells. Here, we investigated the specialized features of classical DC (cDC) subsets following the intraperitoneal injection of a model antigen ovalbumin (OVA). Peritoneal cDC1s were superior to cDC2s in activating OVA-specific CD8 T cells, while both cDCs were similar in stimulating OVA-specific CD4 T cells. Each peritoneal cDC subset differentially regulated the homing properties of CD8 T cells. CD8 T cells stimulated by cDC1s displayed a higher level of lung-homing receptor CCR4, whereas those stimulated by cDC2s prominently expressed various homing receptors including gut-homing molecules CCR9 and α4β7. Also, we found that cDC1s played a dominating role over cDC2s in controlling the overall gene expression of CD8 T cells. Soluble factor(s) emanating from CD8 T cells stimulated by peritoneal cDC1s were responsible for mediating this dominance of cDC1s, and we identified IL-2 as a soluble factor regulating the global gene expression of T cells. Collectively, our study indicates that different peritoneal cDC subsets effectively diversify T cell responses by altering the level of cytokines, such as IL-2, in the milieu.
Collapse
Affiliation(s)
- Moah Sohn
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Young Na
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Soo Shin
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Seul Hye Ryu
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sejung Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyunju In
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Wanho Choi
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Soo Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soomin Hwang
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Kyung Chu
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chae Gyu Park
- Laboratory of Immunology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Therapeutic Antibody Research Center, GENUV Inc., Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
40
|
Deng Y, Münz C. Roles of Lytic Viral Replication and Co-Infections in the Oncogenesis and Immune Control of the Epstein-Barr Virus. Cancers (Basel) 2021; 13:2275. [PMID: 34068598 PMCID: PMC8126045 DOI: 10.3390/cancers13092275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Epstein-Barr virus (EBV) is the prototypic human tumor virus whose continuous lifelong immune control is required to prevent lymphomagenesis in the more than 90% of the human adult population that are healthy carriers of the virus. Here, we review recent evidence that this immune control has not only to target latent oncogenes, but also lytic replication of EBV. Furthermore, genetic variations identify the molecular machinery of cytotoxic lymphocytes as essential for this immune control and recent studies in mice with reconstituted human immune system components (humanized mice) have begun to provide insights into the mechanistic role of these molecules during EBV infection. Finally, EBV often does not act in isolation to cause disease. Some of EBV infection-modulating co-infections, including human immunodeficiency virus (HIV) and Kaposi sarcoma-associated herpesvirus (KSHV), have been modeled in humanized mice. These preclinical in vivo models for EBV infection, lymphomagenesis, and cell-mediated immune control do not only promise a better understanding of the biology of this human tumor virus, but also the possibility to explore vaccine candidates against it.
Collapse
Affiliation(s)
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland;
| |
Collapse
|
41
|
Natural Killer-Dendritic Cell Interactions in Liver Cancer: Implications for Immunotherapy. Cancers (Basel) 2021; 13:cancers13092184. [PMID: 34062821 PMCID: PMC8124166 DOI: 10.3390/cancers13092184] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The reciprocal crosstalk between dendritic cells (DCs) and natural killer (NK) cells plays a pivotal role in regulating immune defense against viruses and tumors. The Th-cell polarizing ability, cytokine-producing capacity, chemokine expression, and migration of DCs are regulated by activated NK cells. Conversely, the effector functions including lysis and cytokine production, proliferation, and migration of NK cells are influenced by close interactions with activated DCs. In this review, we explore the impact of DC–NK cell crosstalk and its therapeutic potential in immune control of liver malignances. Abstract Natural killer (NK) and dendritic cells (DCs) are innate immune cells that play a crucial role in anti-tumor immunity. NK cells kill tumor cells through direct cytotoxicity and cytokine secretion. DCs are needed for the activation of adaptive immune responses against tumor cells. Both NK cells and DCs are subdivided in several subsets endowed with specialized effector functions. Crosstalk between NK cells and DCs leads to the reciprocal control of their activation and polarization of immune responses. In this review, we describe the role of NK cells and DCs in liver cancer, focusing on the mechanisms involved in their reciprocal control and activation. In this context, intrahepatic NK cells and DCs present unique immunological features, due to the constant exposure to non-self-circulating antigens. These interactions might play a fundamental role in the pathology of primary liver cancer, namely hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Additionally, the implications of these immune changes are relevant from the perspective of improving the cancer immunotherapy strategies in HCC and ICC patients.
Collapse
|
42
|
Hatscher L, Amon L, Heger L, Dudziak D. Inflammasomes in dendritic cells: Friend or foe? Immunol Lett 2021; 234:16-32. [PMID: 33848562 DOI: 10.1016/j.imlet.2021.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/14/2022]
Abstract
Inflammasomes are cytosolic multiprotein complexes that crucially contribute to host defense against pathogens but are also involved in the pathogenesis of autoinflammatory diseases. Inflammasome formation leads to activation of effector caspases (caspase-1, 4, 5, or 11), the proteolytic maturation of IL-1β and IL-18 as well as cleavage of the pore-forming protein Gasdermin D. Dendritic cells are major regulators of immune responses as they bridge innate and adaptive immunity. We here summarize the current knowledge on inflammasome expression and formation in murine bone marrow-, human monocyte-derived as well as murine and human primary dendritic cells. Further, we discuss both, the beneficial and detrimental, involvement of inflammasome activation in dendritic cells in cancer, infections, and autoimmune diseases. As inflammasome activation is typically accompanied by Gasdermin d-mediated pyroptosis, which is an inflammatory form of programmed cell death, inflammasome formation in dendritic cells seems ill-advised. Therefore, we propose that hyperactivation, which is inflammasome activation without the induction of pyroptosis, may be a general model of inflammasome activation in dendritic cells to enhance Th1, Th17 as well as cytotoxic T cell responses.
Collapse
Affiliation(s)
- Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany.
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany; Medical Immunology Campus Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Germany.
| |
Collapse
|
43
|
Ortutay Z, Grönholm A, Laitinen M, Keresztes-Andrei M, Hermelo I, Pesu M. Identification of Novel Genetic Regulatory Region for Proprotein Convertase FURIN and Interferon Gamma in T Cells. Front Immunol 2021; 12:630389. [PMID: 33679774 PMCID: PMC7930619 DOI: 10.3389/fimmu.2021.630389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
The proprotein convertase enzyme FURIN promotes the proteolytic maturation of pro-proteins and thereby it serves as an important factor for maintaining cellular homeostasis. In T cells, FURIN is critical for maintaining the T regulatory cell dependent peripheral immune tolerance and intact T helper cell polarization. The enzymatic activity of FURIN is directly associated with its expression levels, but genetic determinants for cell-type specific Furin gene regulation have remained elusive. By exploring the histone acetyltransferase p300 binding patterns in T helper cells, a putative regulatory region at ca. 20kB upstream of Furin gene was identified. When this region was deleted with CRISPR/Cas9 the production of Furin mRNA was significantly reduced in activated mouse T cells. Genome-wide RNA profiling by sequencing revealed that the novel Furin regulator region also impacted the expression of several genes that have previously been associated with the Th1 type hall mark cytokine IFNγ regulation or function. Finally, Furin genetic regulatory region was found to specifically promote the secretion of IFNγ by activated T cells. In sum, our data unravels the presence of Furin expression regulatory region in T cells that has characteristics of a super-enhancer for Th1 cell fate.
Collapse
Affiliation(s)
- Zsuzsanna Ortutay
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anna Grönholm
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Melina Laitinen
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Melinda Keresztes-Andrei
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Ismail Hermelo
- Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marko Pesu
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
44
|
Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021; 39:131-166. [PMID: 33481643 DOI: 10.1146/annurev-immunol-061020-053707] [Citation(s) in RCA: 434] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.
Collapse
Affiliation(s)
- Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | | | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| |
Collapse
|
45
|
Suen JL, Wu TT, Li YH, Lee CL, Kuo FC, Yan PS, Wu CF, Tran M, Wang CJ, Hung CH, Wu MT, Chan MWY, Huang SK. Environmental Factor-Mediated Transgenerational Inheritance of Igf2r Hypomethylation and Pulmonary Allergic Response via Targeting Dendritic Cells. Front Immunol 2020; 11:603831. [PMID: 33424850 PMCID: PMC7786300 DOI: 10.3389/fimmu.2020.603831] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/23/2020] [Indexed: 11/13/2022] Open
Abstract
The developmental origin of allergic diseases has been suggested, but the molecular basis remains enigmatic. Exposure to environmental factors, such as di-(2-ethylhexyl) phthalate (DEHP; a common plasticizer), is suggested to be associated with increased childhood allergic asthma, but the causal relationship and its underlying mechanism remain unknown. This study explored the transgenerational mechanism of DEHP on allergic asthma and dendritic cell (DC) homeostasis through epigenetic modification. In a murine model, ancestral exposure of C57BL/6 mice to low-dose DEHP led to trans-generational promoter hypomethylation of the insulin-like growth factor 2 receptor (Igf2r), concomitant with enhanced Igf2r expression and increased apoptosis prominently in CD8α+ DCs upon ligand stimulation, with consequent reduction in their IL-12 secretion and subsequent T cell-derived IFN-γ, thereby promoting a default Th2-associated pulmonary allergic response. Increased apoptosis was also noted in circulating IGF2Rhigh human DCs. Further, in human placenta, the methylation level at the orthologous IGF2R promoter region was shown to be inversely correlated with the level of maternal DEHP intake. These results support the importance of ancestral phthalate exposure in conferring the trans-generational risk of allergic phenotypes, featuring hypo-methylation of the IGF2R gene and dysregulated DC homeostasis.
Collapse
Affiliation(s)
- Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tai-Ting Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yue-Hyuan Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Lai Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu-Chen Kuo
- Department of Gynecology and Obstetrics, E-Da Hospital, Kaohsiung, Taiwan
- Graduate Institute of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Pearlly S. Yan
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Chia-Fang Wu
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mita Tran
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chien-Jen Wang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Hsing Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Ming-Tsang Wu
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Community Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
46
|
Abstract
Dendritic cells are a specialized subset of hematopoietic cells essential for mounting immunity against tumors and infectious disease as well as inducing tolerance for maintenance of homeostasis. DCs are equipped with number of immunoregulatory or stimulatory molecules that interact with other leukocytes to modulate their functions. Recent advances in DC biology identified a specific role for the conventional dendritic cell type 1 (cDC1) in eliciting cytotoxic CD8+ T cells essential for clearance of tumors and infected cells. The critical role of this subset in eliciting immune responses or inducing tolerance has largely been defined in mice whereas the biology of human cDC1 is poorly characterized owing to their extremely low frequency in tissues. A detailed characterization of the functions of many immunoregulatory and stimulatory molecules expressed by human cDC1 is critical for understanding their biology to exploit this subset for designing novel therapeutic modalities against cancer, infectious disease and autoimmune disorders.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Kristen J Radford
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Extramural member Parker Institute of Cancer Immunotherapy, CA, United States.
| |
Collapse
|
47
|
Oba T, Long MD, Keler T, Marsh HC, Minderman H, Abrams SI, Liu S, Ito F. Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s. Nat Commun 2020; 11:5415. [PMID: 33110069 PMCID: PMC7592056 DOI: 10.1038/s41467-020-19192-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/02/2020] [Indexed: 01/01/2023] Open
Abstract
The ability of cancer cells to ensure T-cell exclusion from the tumor microenvironment is a significant mechanism of resistance to anti-PD-1/PD-L1 therapy. Evidence indicates crucial roles of Batf3-dependent conventional type-1 dendritic cells (cDC1s) for inducing antitumor T-cell immunity; however, strategies to maximize cDC1 engagement remain elusive. Here, using multiple orthotopic tumor mouse models resistant to anti-PD-L1-therapy, we are testing the hypothesis that in situ induction and activation of tumor-residing cDC1s overcomes poor T-cell infiltration. In situ immunomodulation with Flt3L, radiotherapy, and TLR3/CD40 stimulation induces an influx of stem-like Tcf1+ Slamf6+ CD8+ T cells, triggers regression not only of primary, but also untreated distant tumors, and renders tumors responsive to anti-PD-L1 therapy. Furthermore, serial in situ immunomodulation (ISIM) reshapes repertoires of intratumoral T cells, overcomes acquired resistance to anti-PD-L1 therapy, and establishes tumor-specific immunological memory. These findings provide new insights into cDC1 biology as a critical determinant to overcome mechanisms of intratumoral T-cell exclusion.
Collapse
Affiliation(s)
- Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, NJ, USA
| | | | - Hans Minderman
- Flow & Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
48
|
Intradermal Delivery of Dendritic Cell-Targeting Chimeric mAbs Genetically Fused to Type 2 Dengue Virus Nonstructural Protein 1. Vaccines (Basel) 2020; 8:vaccines8040565. [PMID: 33019498 PMCID: PMC7712967 DOI: 10.3390/vaccines8040565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 11/21/2022] Open
Abstract
Targeting dendritic cells (DCs) by means of monoclonal antibodies (mAbs) capable of binding their surface receptors (DEC205 and DCIR2) has previously been shown to enhance the immunogenicity of genetically fused antigens. This approach has been repeatedly demonstrated to enhance the induced immune responses to passenger antigens and thus represents a promising therapeutic and/or prophylactic strategy against different infectious diseases. Additionally, under experimental conditions, chimeric αDEC205 or αDCIR2 mAbs are usually administered via an intraperitoneal (i.p.) route, which is not reproducible in clinical settings. In this study, we characterized the delivery of chimeric αDEC205 or αDCIR2 mAbs via an intradermal (i.d.) route, compared the elicited humoral immune responses, and evaluated the safety of this potential immunization strategy under preclinical conditions. As a model antigen, we used type 2 dengue virus (DENV2) nonstructural protein 1 (NS1). The results show that the administration of chimeric DC-targeting mAbs via the i.d. route induced humoral immune responses to the passenger antigen equivalent or superior to those elicited by i.p. immunization with no toxic effects to the animals. Collectively, these results clearly indicate that i.d. administration of DC-targeting chimeric mAbs presents promising approaches for the development of subunit vaccines, particularly against DENV and other flaviviruses.
Collapse
|
49
|
Ramos MI, Garcia S, Helder B, Aarrass S, Reedquist KA, Jacobsen SE, Tak PP, Lebre MC. cDC1 are required for the initiation of collagen-induced arthritis. J Transl Autoimmun 2020; 3:100066. [PMID: 33015599 PMCID: PMC7522802 DOI: 10.1016/j.jtauto.2020.100066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is chronic autoimmune disease which etiology remains unknown. Several cell types have been described to potentiate/aggravate the arthritic process however the initiating event in synovial inflammation is still elusive. Dendritic cells (DCs) are essential for the initiation of primary immune responses and thus we hypothesized that these cells might be crucial for RA induction. DCs are a heterogeneous population of cells comprising different subsets with distinct phenotype and function. Here we investigated which DC subset(s) is/are crucial for the initiation of the arthritic process. We have previously demonstrated that Flt3−/− mice, with reduced DCs, were protected from collagen induced arthritis (CIA). Here we have shown that GM-CSF derived DCs in Flt3L−/− mice are functional but not sufficient to induce arthritis. Batf3−/− mice lacking both CD103+ and CD8α+ cDC1 were resistant to collagen induced arthritis (CIA), demonstrating that this DC subset is crucial for arthritis development. CEP-701 (a Flt3L inhibitor) treatment prevented CIA induction, and reduced dramatically the numbers CD103+ cDC1s present in the lymph nodes and synovium. Hence this study identified cDC1 as the main subset orchestrating the initiation of cell-mediated immunity in arthritis. Flt3L independent DCs present in Flt3L−/− mice are functional but are not sufficient to induce arthritis. BATF3−/− mice lacking cDC1 are protected from arthritis development indicating that cDC1 are necessary for disease induction. Treatment with a Flt3L inhibitor, CEP701, reduced cDC1 populations and prevented arthritis induction.
Collapse
Affiliation(s)
- Maria Ines Ramos
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Samuel Garcia
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Boy Helder
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Saida Aarrass
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Kris A Reedquist
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Sten E Jacobsen
- Haematopoietic Stem Cell Laboratory and MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Maria Cristina Lebre
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
50
|
Guignabert C. Dendritic Cells in Pulmonary Hypertension: Foot Soldiers or Hidden Enemies? Am J Respir Cell Mol Biol 2020; 63:551-552. [PMID: 32804536 PMCID: PMC7605161 DOI: 10.1165/rcmb.2020-0330ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Christophe Guignabert
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France, and.,Institut National de la Santé et de la Recherche Médicale (INSERM) - Unité Mixte de Recherche en Santé (UMR_S) 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|