1
|
Ise W, Koike T, Shimada N, Yamamoto H, Tai Y, Shirai T, Kawakami R, Kuwabara M, Kawai C, Shida K, Inoue T, Hojo N, Ichiyama K, Sakaguchi S, Shiroguchi K, Suzuki K, Kurosaki T. KLF2 expression in IgG plasma cells at their induction site regulates the migration program. J Exp Med 2025; 222:e20241019. [PMID: 39976598 PMCID: PMC11841683 DOI: 10.1084/jem.20241019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/27/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
Newly generated plasma cells in secondary lymphoid organs migrate to niches in the bone marrow, wherein they survive as long-lived plasma cells (LLPCs). Although LLPCs have been extensively characterized, it is still unclear what the key determinant(s) are for plasma cell longevity. One model postulates that plasma cell heterogeneity is established at the induction site, thereby instructing their longevity. Here, we found that, among newly generated IgG plasma cells, integrin β7hi marks plasma cells predisposed to home to the bone marrow, whereas integrin β7lo cells remain in secondary lymphoid organs. Mechanistically, this egress-prone fraction had a higher expression of the KLF2 transcription factor, the loss of which resulted in defective egress by downregulating S1PR1 and CD11b. Disruption of plasma cell egress results in defective antibody durability, thereby making mice more susceptible to influenza reinfection. Thus, the migration program of plasma cells established at the induction site plays a critical role in determining antibody durability.
Collapse
Affiliation(s)
- Wataru Ise
- Regulation of Host Defense Team, Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Takuya Koike
- Regulation of Host Defense Team, Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Department of Molecular Systems Immunology, University of Tokyo Pandemic Preparedness, Infection, and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Nozomi Shimada
- Regulation of Host Defense Team, Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiromi Yamamoto
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yuki Tai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Taiichiro Shirai
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ryoji Kawakami
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mana Kuwabara
- Regulation of Host Defense Team, Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Chie Kawai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kyoko Shida
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Systems Immunology, University of Tokyo Pandemic Preparedness, Infection, and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Nozomi Hojo
- Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Kenji Ichiyama
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Experimental Pathology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Katsuyuki Shiroguchi
- Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Kazuhiro Suzuki
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| |
Collapse
|
2
|
Wang H, Li Y, Qiu D, Pan Q, Xu Y, Liu Y, Wu Y. Personalized Nanomedicine-Mediated immune regulation for Anti-Rejection in organ transplantation. Int J Pharm 2025; 674:125450. [PMID: 40122222 DOI: 10.1016/j.ijpharm.2025.125450] [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: 11/21/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
The advent of personalized medicine and nanomedicine has led to significant advancements in organ transplantation. Personalized medicine leverages individual patient profiles, including genetic, epigenetic, and immune characteristics, to tailor treatment regimens. Nanomedicine, involving the use of nanoparticles and nanotechnology, offers precise drug delivery and innovative diagnostic tools. The integration of personalized nanomedicine into these fields has the potential to revolutionize transplantation by enhancing graft survival, minimizing adverse effects, and achieving immune tolerance. This review explores the current landscape of personalized nanomedicine for organ transplantation, focusing on immune modulation and therapeutic strategies tailored to individual patient profiles. We also discuss future research directions, including large-scale clinical trials, and regulatory considerations. This review concludes by examining the potential of personalized nanomedicine in improving long-term transplant outcomes and enhancing patient quality of life.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yutong Li
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Qiu
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinyu Pan
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yong Xu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
3
|
Huang Y, Du Z, Lai Z, Wen D, Huang L, He M, Wu Z, Li H, OuYang H, Wu W, Kan A, Shi M. Single-Nucleus and Spatial Transcriptome Profiling Delineates the Multicellular Ecosystem in Hepatocellular Carcinoma After Hepatic Arterial Infusion Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405749. [PMID: 39686623 PMCID: PMC11791974 DOI: 10.1002/advs.202405749] [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: 05/25/2024] [Revised: 11/08/2024] [Indexed: 12/18/2024]
Abstract
Hepatic arterial infusion chemotherapy (HAIC) has emerged as a promising treatment strategy for hepatocellular carcinoma (HCC), but a detailed understanding of the multicellular ecosystem after HAIC treatment is lacking. Here, we collected tumor samples from treatment-naïve primary and post-HAIC HCC, and integrated single-nucleus RNA sequencing with spatial transcriptomics to characterize the tumor ecosystem in the post-HAIC HCC. Increased fractions and enhanced cellular communication of CD4+ T, CD20+ B, and dendritic cell subtypes were identified in post-HAIC tumors. Moreover, it is substantiated that HAIC promoted tertiary lymphoid structures (TLS) formation, and addressed the roles of TLSs as spatial niches of cellular communication. Specifically, intermediate exhausted CD8+ T cells expressing Granzyme-K and PD-1 (PD-1+CD8+ Tex-int) expanded following HAIC and exhibited a functionally antitumor phenotype. PD-1+CD8+ Tex-int accumulated in the TLS vicinity and disseminated throughout the tumor microenvironment, demonstrating potential as an effective biomarker for HAIC-based treatment in HCC. This study provides valuable resources and biological insights in the cellular underpinnings of HAIC treatment.
Collapse
Affiliation(s)
- YeXing Huang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZeFeng Du
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZhiCheng Lai
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - DongSheng Wen
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - LiChang Huang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - MinKe He
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZiChao Wu
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - HuiFang Li
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - HanYue OuYang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - WenChao Wu
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - Anna Kan
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - Ming Shi
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| |
Collapse
|
4
|
Schett G, Nagy G, Krönke G, Mielenz D. B-cell depletion in autoimmune diseases. Ann Rheum Dis 2024; 83:1409-1420. [PMID: 38777374 DOI: 10.1136/ard-2024-225727] [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: 02/26/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
B cells have a pivotal function in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis and systemic lupus erythematosus. In autoimmune disease, B cells orchestrate antigen presentation, cytokine production and autoantibody production, the latter via their differentiation into antibody-secreting plasmablasts and plasma cells. This article addresses the current therapeutic strategies to deplete B cells in order to ameliorate or potentially even cure autoimmune disease. It addresses the main target antigens in the B-cell lineage that are used for therapeutic approaches. Furthermore, it summarises the current evidence for successful treatment of autoimmune disease with monoclonal antibodies targeting B cells and the limitations and challenges of these approaches. Finally, the concept of deep B-cell depletion and immunological reset by chimeric antigen receptor T cells is discussed, as well as the lessons from this approach for better understanding the role of B cells in autoimmune disease.
Collapse
Affiliation(s)
- Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - György Nagy
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Charite, Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| |
Collapse
|
5
|
Pereira AL, Galli S, Nombela‐Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere 2024; 8:e133. [PMID: 39086665 PMCID: PMC11289431 DOI: 10.1002/hem3.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 08/02/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are the cornerstone of the hematopoietic system. HSCs sustain the continuous generation of mature blood derivatives while self-renewing to preserve a relatively constant pool of progenitors throughout life. Yet, long-term maintenance of functional HSCs exclusively takes place in association with their native tissue microenvironment of the bone marrow (BM). HSCs have been long proposed to reside in fixed and identifiable anatomical units found in the complex BM tissue landscape, which control their identity and fate in a deterministic manner. In the last decades, tremendous progress has been made in the dissection of the cellular and molecular fabric of the BM, the structural organization governing tissue function, and the plethora of interactions established by HSCs. Nonetheless, a holistic model of the mechanisms controlling HSC regulation in their niche is lacking to date. Here, we provide an overview of our current understanding of BM anatomy, HSC localization, and crosstalk within local cellular neighborhoods in murine and human tissues, and highlight fundamental open questions on how HSCs functionally integrate in the BM microenvironment.
Collapse
Affiliation(s)
- Ana Luísa Pereira
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - Serena Galli
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - César Nombela‐Arrieta
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| |
Collapse
|
6
|
Hlavackova E, Krenova Z, Kerekes A, Slanina P, Vlkova M. B cell subsets reconstitution and immunoglobulin levels in children and adolescents with B non-Hodgkin lymphoma after treatment with single anti CD20 agent dose included in chemotherapeutic protocols: single center experience and review of the literature. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024; 168:167-176. [PMID: 37227099 DOI: 10.5507/bp.2023.021] [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: 09/30/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND RTX, an anti-CD20 monoclonal antibody, added to chemotherapy has proven to be effective in children and adolescents with high-grade, high-risk and matured non-Hodgkin lymphoma. RTX leads to prompt CD19+ B lymphocyte depletion. However, despite preserved immunoglobulin production by long-lived plasmablasts after treatment, patients remain at risk of prolonged hypogammaglobulinemia. Further, there are few general guidelines for immunology laboratories and clinical feature monitoring after B cell-targeted therapies. The aim of this paper is to describe B cell reconstitution and immunoglobulin levels after pediatric B-NHL protocols, that included a single RTX dose and to review the literature. METHODS A retrospective single-center study on the impact of a single RTX dose included in a chemotherapeutic pediatric B Non-Hodgkin Lymphoma (B-NHL) treatment protocols. Immunology laboratory and clinical features were evaluated over an eight hundred days follow-up (FU) period, after completing B-NHL treatment. RESULTS Nineteen patients (fifteen Burkitt lymphoma, three Diffuse large B cell lymphoma, and one Marginal zone B cell lymphoma) fulfilled the inclusion criteria. Initiation of B cell subset reconstitution occurred a median of three months after B-NHL treatment. Naïve and transitional B cells declined over the FU in contrast to the marginal zone and the switched memory B cell increase. The percentage of patients with IgG, IgA, and IgM hypogammaglobulinemia declined consistently over the FU. Prolonged IgG hypogammaglobulinemia was detectable in 9%, IgM in 13%, and IgA in 25%. All revaccinated patients responded to protein-based vaccines by specific IgG antibody production increase. Following antibiotic prophylaxes, none of the patients with hypogammaglobulinemia manifested with either a severe or opportunistic infection course. CONCLUSION The addition of a single RTX dose to the chemotherapeutic treatment protocols was not shown to increase the risk of developing secondary antibody deficiency in B-NHL pediatric patients. Observed prolonged hypogammaglobulinemia remained clinically silent. However interdisciplinary agreement on regular long-term immunology FU after anti-CD20 agent treatment is required.
Collapse
Affiliation(s)
- Eva Hlavackova
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Zdenka Krenova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Arpad Kerekes
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Peter Slanina
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcela Vlkova
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
7
|
Lu F, Xu J, Liu Y, Ren Z, Chen J, Gong W, Yin Y, Li Y, Qian L, He X, Han X, Lin Z, Lu J, Zhang W, Liu J, Menard D, Han ET, Cao J. Plasmodium vivax serological exposure markers: PvMSP1-42-induced humoral and memory B-cell response generates long-lived antibodies. PLoS Pathog 2024; 20:e1012334. [PMID: 38941356 PMCID: PMC11239109 DOI: 10.1371/journal.ppat.1012334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/11/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024] Open
Abstract
Plasmodium vivax serological exposure markers (SEMs) have emerged as promising tools for the actionable surveillance and implementation of targeted interventions to accelerate malaria elimination. To determine the dynamic profiles of SEMs in current and past P. vivax infections, we screened and selected 11 P. vivax proteins from 210 putative proteins using protein arrays, with a set of serum samples obtained from patients with acute P. vivax and documented past P. vivax infections. Then we used a murine protein immune model to initially investigate the humoral and memory B cell response involved in the generation of long-lived antibodies. We show that of the 11 proteins, especially C-terminal 42-kDa region of P. vivax merozoite surface protein 1 (PvMSP1-42) induced longer-lasting long-lived antibodies, as these antibodies were detected in individuals infected with P. vivax in the 1960-1970s who were not re-infected until 2012. In addition, we provide a potential mechanism for the maintenance of long-lived antibodies after the induction of PvMSP1-42. The results indicate that PvMSP1-42 induces more CD73+CD80+ memory B cells (MBCs) compared to P. vivax GPI-anchored micronemal antigen (PvGAMA), allowing IgG anti-PvMSP1-42 antibodies to be maintained for a long time.
Collapse
Affiliation(s)
- Feng Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiahui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yaobao Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Zhenyu Ren
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Junhu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Weijuan Gong
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yi Yin
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yinyue Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Li Qian
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xinlong He
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiu Han
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhijie Lin
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jingyuan Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Wenwen Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiali Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Didier Menard
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
- Université de Strasbourg, UR 3073—Pathogens Host Arthropods Vectors Interactions Unit, Malaria Genetics and Resistance Team (MEGATEAM), Strasbourg, France
- CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, Strasbourg, France
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| |
Collapse
|
8
|
Wang L, Jin G, Zhou Q, Liu Y, Zhao X, Li Z, Yin N, Peng M. Induction of immortal-like and functional CAR T cells by defined factors. J Exp Med 2024; 221:e20232368. [PMID: 38530240 PMCID: PMC10965394 DOI: 10.1084/jem.20232368] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/10/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Long-term antitumor efficacy of chimeric antigen receptor (CAR) T cells depends on their functional persistence in vivo. T cells with stem-like properties show better persistence, but factors conferring bona fide stemness to T cells remain to be determined. Here, we demonstrate the induction of CAR T cells into an immortal-like and functional state, termed TIF. The induction of CARTIF cells depends on the repression of two factors, BCOR and ZC3H12A, and requires antigen or CAR tonic signaling. Reprogrammed CARTIF cells possess almost infinite stemness, similar to induced pluripotent stem cells while retaining the functionality of mature T cells, resulting in superior antitumor effects. Following the elimination of target cells, CARTIF cells enter a metabolically dormant state, persisting in vivo with a saturable niche and providing memory protection. TIF represents a novel state of T cells with unprecedented stemness, which confers long-term functional persistence of CAR T cells in vivo and holds broad potential in T cell therapies.
Collapse
Affiliation(s)
- Lixia Wang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Qiuping Zhou
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yanyan Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
9
|
Kashimura M. Blood defense system - Proposal for a new concept of an immune system against blood borne pathogens comprising the liver, spleen and bone marrow. Scand J Immunol 2024; 99:e13363. [PMID: 38605529 DOI: 10.1111/sji.13363] [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: 08/22/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 04/13/2024]
Abstract
Blood-borne pathogen (BBP) infections can rapidly progress to life-threatening sepsis and must therefore be promptly eliminated by the host's immune system. Intravascular macrophages of the liver sinusoid, splenic marginal zone and red pulp and perisinusoidal macrophage protrusions in the bone marrow (BM) directly phagocytose BBPs in the blood as an innate immune response. The liver, spleen and BM thereby work together as the blood defence system (BDS) in response to BBPs by exerting their different immunological roles. The liver removes the vast majority of these invading organisms via innate immunity, but their complete elimination is not possible without the actions of antibodies. Splenic marginal zone B cells promptly produce IgM and IgG antibodies against BBPs. The splenic marginal zone transports antigenic information from the innate to the adaptive immune systems. The white pulp of the spleen functions as adaptive immune tissue and produces specific and high-affinity antibodies with an immune memory against BBPs. The BM works to maintain immune memory by supporting the survival of memory B cells, memory T cells and long-lived plasma cells (LLPCs), all of which have dedicated niches. Furthermore, BM perisinusoidal naïve follicular B cells promptly produce IgM antibodies against BBPs in the BM sinusoid and the IgG memory B cells residing in the BM rapidly transform to plasma cells which produce high-affinity IgG antibodies upon reinfection. This review describes the complete immune defence characteristics of the BDS against BBPs through the collaboration of the liver, spleen and BM with combined different immunological roles.
Collapse
Affiliation(s)
- Makoto Kashimura
- Department of Hematology, Shinmatsudo Central General Hospital, Matsudo, Japan
| |
Collapse
|
10
|
Rupp L, Dietsche I, Kießler M, Sommer U, Muckenhuber A, Steiger K, van Eijck CWF, Richter L, Istvanffy R, Jäger C, Friess H, van Eijck CHJ, Demir IE, Reyes CM, Schmitz M. Neoadjuvant chemotherapy is associated with suppression of the B cell-centered immune landscape in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1378190. [PMID: 38629072 PMCID: PMC11018975 DOI: 10.3389/fimmu.2024.1378190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at advanced stages and associated with early distant metastasis and poor survival. Besides clinical factors, the tumor microenvironment (TME) emerged as a crucial determinant of patient survival and therapy response in many tumors, including PDAC. Thus, the presence of tumor-infiltrating lymphocytes and the formation of tertiary lymphoid structures (TLS) is associated with longer survival in PDAC. Although neoadjuvant therapy (NeoTx) has improved the management of locally advanced tumors, detailed insight into its effect on various TME components is limited. While a remodeling towards a proinflammatory state was reported for PDAC-infiltrating T cells, the effect of NeoTx on B cell subsets, including plasma cells, and TLS formation is widely unclear. We thus investigated the frequency, composition, and spatial distribution of PDAC-infiltrating B cells in primary resected (PR) versus neoadjuvant-treated patients using a novel multiplex immunohistochemistry panel. The NeoTx group displayed significantly lower frequencies of pan B cells, GC B cells, plasmablasts, and plasma cells, accompanied by a reduced abundance of TLS. This finding was supported by bulk RNA-sequencing analysis of an independent fresh frozen tissue cohort, which revealed that major B cell pathways were downregulated in the NeoTx group. We further observed that plasma cells frequently formed aggregates that localized close to TLS and that TLS+ patients displayed significantly higher plasma cell frequencies compared to TLS- patients in the PR group. Additionally, high densities of CD20+ intratumoral B cells were significantly associated with longer overall survival in the PR group. While CD20+ B cells held no prognostic value for NeoTx patients, an increased frequency of proliferating CD20+Ki67+ B cells emerged as an independent prognostic factor for longer survival in the NeoTx group. These results indicate that NeoTx differentially affects PDAC-infiltrating immune cells and may have detrimental effects on the existing B cell landscape and the formation of TLS. Gaining further insight into the underlying molecular mechanisms is crucial to overcome the intrinsic immunotherapy resistance of PDAC and develop novel strategies to improve the long-term outcome of PDAC patients.
Collapse
Affiliation(s)
- Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ina Dietsche
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Maximilian Kießler
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Alexander Muckenhuber
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Casper W. F. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Leonard Richter
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Casper H. J. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of General Surgery, Hepato-Pancreato-Biliary (HPB) Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Technical University of Munich, Munich, Germany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Mertens TF, Liebheit AT, Ehl J, Köhler R, Rakhymzhan A, Woehler A, Katthän L, Ebel G, Liublin W, Kasapi A, Triantafyllopoulou A, Schulz TJ, Niesner RA, Hauser AE. MarShie: a clearing protocol for 3D analysis of single cells throughout the bone marrow at subcellular resolution. Nat Commun 2024; 15:1764. [PMID: 38409121 PMCID: PMC10897183 DOI: 10.1038/s41467-024-45827-6] [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: 03/06/2023] [Accepted: 02/01/2024] [Indexed: 02/28/2024] Open
Abstract
Analyzing immune cell interactions in the bone marrow is vital for understanding hematopoiesis and bone homeostasis. Three-dimensional analysis of the complete, intact bone marrow within the cortex of whole long bones remains a challenge, especially at subcellular resolution. We present a method that stabilizes the marrow and provides subcellular resolution of fluorescent signals throughout the murine femur, enabling identification and spatial characterization of hematopoietic and stromal cell subsets. By combining a pre-processing algorithm for stripe artifact removal with a machine-learning approach, we demonstrate reliable cell segmentation down to the deepest bone marrow regions. This reveals age-related changes in the marrow. It highlights the interaction between CX3CR1+ cells and the vascular system in homeostasis, in contrast to other myeloid cell types, and reveals their spatial characteristics after injury. The broad applicability of this method will contribute to a better understanding of bone marrow biology.
Collapse
Affiliation(s)
- Till Fabian Mertens
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Alina Tabea Liebheit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Johanna Ehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ralf Köhler
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Asylkhan Rakhymzhan
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Andrew Woehler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115, Berlin, Germany
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Lukas Katthän
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Gernot Ebel
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Wjatscheslaw Liublin
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ana Kasapi
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Tim Julius Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), 85764, Munich-Neuherberg, Germany
| | - Raluca Aura Niesner
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Dynamic and Functional in vivo Imaging, Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
12
|
Kared H, Jyssum I, Alirezaylavasani A, Egner IM, The Tran T, Tietze L, Lund KP, Tveter AT, Provan SA, Ørbo H, Haavardsholm EA, Vaage JT, Jørgensen K, Syversen SW, Lund-Johansen F, Goll GL, Munthe LA. Dynamics of SARS-CoV-2 immunity after vaccination and breakthrough infection in rituximab-treated rheumatoid arthritis patients: a prospective cohort study. Front Immunol 2024; 15:1296273. [PMID: 38455062 PMCID: PMC10917913 DOI: 10.3389/fimmu.2024.1296273] [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: 09/18/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Background SARS-CoV-2 vaccination in rheumatoid arthritis (RA) patients treated with B cell-depleting drugs induced limited seroconversion but robust cellular response. We aimed to document specific T and B cell immunity in response to vaccine booster doses and breakthrough infection (BTI). Methods We included 76 RA patients treated with rituximab who received up to four SARS-CoV-2 vaccine doses or three doses plus BTI, in addition to vaccinated healthy donors (HD) and control patients treated with tumor necrosis factor inhibitor (TNFi). We quantified anti-SARS-CoV-2 receptor-binding domain (RBD) Spike IgG, anti-nucleocapsid (NC) IgG, 92 circulating inflammatory proteins, Spike-binding B cells, and Spike-specific T cells along with comprehensive high-dimensional phenotyping and functional assays. Findings The time since the last rituximab infusion, persistent inflammation, and age were associated with the anti-SARS-CoV-2 RBD IgG seroconversion. The vaccine-elicited serological response was accompanied by an incomplete induction of peripheral Spike-specific memory B cells but occurred independently of T cell responses. Vaccine- and BTI-elicited cellular immunity was similar between RA and HD ex vivo in terms of frequency or phenotype of Spike-specific cytotoxic T cells and in vitro in terms of the functionality and differentiation profile of Spike-specific T cells. Interpretation SARS-CoV-2 vaccination in RA can induce persistent effector T-cell responses that are reactivated by BTI. Paused rituximab medication allowed serological responses after a booster dose (D4), especially in RA with lower inflammation, enabling efficient humoral and cellular immunity after BTI, and contributed overall to the development of potential durable immunity.
Collapse
Affiliation(s)
- Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Jyssum
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid M. Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trung The Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Katrine Persgård Lund
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Therese Tveter
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Sella A. Provan
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Hilde Ørbo
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Espen A. Haavardsholm
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Kristin Jørgensen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ludvig A. Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Little JS, Oakley MS, Thorner AR, Johnston D, Majam V, Liakos AD, Novack LA, Zheng H, Meredith S, Chou CK, Newton BR, Soiffer RJ, Krause PJ, Baden LR, Kumar S. Immune Control in Repeated Babesia microti Infection in a Patient With B-Cell Deficiency. Open Forum Infect Dis 2024; 11:ofad568. [PMID: 38213635 PMCID: PMC10783156 DOI: 10.1093/ofid/ofad568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/09/2023] [Indexed: 01/13/2024] Open
Abstract
The immunology of human babesiosis is poorly investigated. We present a comprehensive investigation of a 75-year-old man with B-cell deficiency who experienced 3 episodes of babesiosis over a 6-year period. Slowly evolving clinical immunity was observed, as evidenced by milder clinical symptoms and lower peak parasite burden after each subsequent babesiosis episode. The patient exhibited several striking immunologic findings. First, the patient had exceptionally high Babesia microti-specific antibodies despite very few circulating B cells, which predominantly coexpressed CD27 (memory marker) and CD95 (death receptor). Second, we demonstrated the presence of long-lasting NK cells and expansion of T memory stem cells. Third, levels of the IP-10 cytokine directly correlated with parasite burden. These results raise fundamental questions on the priming, maintenance, and location of a B-cell population that produces high antibody levels in the face of severe B-cell deficiency. Our results should invoke interest among researchers to study the immunology and pathogenesis of human babesiosis.
Collapse
Affiliation(s)
- Jessica S Little
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Miranda S Oakley
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anna R Thorner
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Demerise Johnston
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Victoria Majam
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Alexis D Liakos
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Lewis A Novack
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hong Zheng
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Scott Meredith
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Chao-Kai Chou
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Benjamin R Newton
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J Soiffer
- Harvard Medical School, Boston, Massachusetts, USA
- Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Peter J Krause
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health and Yale School of Medicine, New Haven, Connecticut, USA
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sanjai Kumar
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
14
|
Elizaldi SR, Hawes CE, Verma A, Dinasarapu AR, Lakshmanappa YS, Schlegel BT, Rajasundaram D, Li J, Durbin-Johnson BP, Ma ZM, Beckman D, Ott S, Lifson J, Morrison JH, Iyer SS. CCR7+ CD4 T Cell Immunosurveillance Disrupted in Chronic SIV-Induced Neuroinflammation in Rhesus Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555037. [PMID: 37693567 PMCID: PMC10491118 DOI: 10.1101/2023.08.28.555037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
CD4 T cells survey and maintain immune homeostasis in the brain, yet their differentiation states and functional capabilities remain unclear. Our approach, combining single-cell transcriptomic analysis, ATAC-seq, spatial transcriptomics, and flow cytometry, revealed a distinct subset of CCR7+ CD4 T cells resembling lymph node central memory (T CM ) cells. We observed chromatin accessibility at the CCR7, CD28, and BCL-6 loci, defining molecular features of T CM . Brain CCR7+ CD4 T cells exhibited recall proliferation and interleukin-2 production ex vivo, showcasing their functional competence. We identified the skull bone marrow as a local niche for these cells alongside other CNS border tissues. Sequestering T CM cells in lymph nodes using FTY720 led to reduced CCR7+ CD4 T cell frequencies in the cerebrospinal fluid, accompanied by increased monocyte levels and soluble markers indicating immune activation. In macaques chronically infected with SIVCL57 and experiencing viral rebound due to cessation of antiretroviral therapy, a decrease in brain CCR7+ CD4 T cells was observed, along with increased microglial activation and initiation of neurodegenerative pathways. Our findings highlight a role for CCR7+ CD4 T cells in CNS immune surveillance and their decline during chronic SIV-induced neuroinflammation highlights their responsiveness to neuroinflammatory processes. GRAPHICAL ABSTRACT In Brief Utilizing single-cell and spatial transcriptomics on adult rhesus brain, we uncover a unique CCR7+ CD4 T cell subset resembling central memory T cells (T CM ) within brain and border tissues, including skull bone marrow. Our findings show decreased frequencies of this subset during SIV- induced chronic neuroinflammation, emphasizing responsiveness of CCR7+ CD4 T cells to CNS disruptions. Highlights CCR7+ CD4 T cells survey border and parenchymal CNS compartments during homeostasis; reduced presence of CCR7+ CD4 T cells in cerebrospinal fluid leads to immune activation, implying a role in neuroimmune homeostasis. CNS CCR7+ CD4 T cells exhibit phenotypic and functional features of central memory T cells (T CM ) including production of interleukin 2 and the capacity for rapid recall proliferation. Furthermore, CCR7+ CD4 T cells reside in the skull bone marrow. CCR7+ CD4 T cells are markedly decreased within the brain parenchyma during chronic viral neuroinflammation.
Collapse
|
15
|
Abstract
Bone marrow is known as the site of hematopoiesis. What is not being described in textbooks of immunology is the fact that bone marrow is not only a generative, but also an antigen-responsive, immune organ. It is also a major storage site for antigen-specific memory B and T cells. That bone marrow is a priming site for T cell responses to blood borne antigens was discovered exactly 20 years ago. This review celebrates this important discovery. The review provides a number of examples of medical relevance of bone marrow as a central immune system, including cancer, microbial infections, autoimmune reactions, and bone marrow transplantation. Bone marrow mesenchymal stem cell-derived stromal cells provide distinct bone marrow niches for stem cells and immune cells. By transmitting anti-inflammatory dampening effects, facilitating wound healing and tissue regeneration mesenchymal stem cells contribute to homeostasis of bone and other tissues. Based on the evidence presented, the review proposes that bone marrow is a multifunctional and protective immune system. In an analogy to the central nervous system, it is suggested that bone marrow be designated as the central immune system.
Collapse
|
16
|
Costa MD, Donner S, Bertrand J, Pop OL, Lohmann CH. Hypersensitivity and lymphocyte activation after total hip arthroplasty. ORTHOPADIE (HEIDELBERG, GERMANY) 2023; 52:214-221. [PMID: 36820851 DOI: 10.1007/s00132-023-04349-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/24/2023]
Abstract
In the last decades total hip arthroplasty (THA) has become a standard procedure with many benefits but also a few still unsolved complications, which can lead to surgical revision in 19-23% of cases. Thus, aseptic loosening and metal hypersensitivity remain challenges. The phenomenon of wear debris causes chronic inflammation, which produces osteolysis and aseptic loosening. Wear debris promotes osteoclast production and inhibits osteoblasts by secretion of pro-inflammatory cytokines. Micro-abrasions can be induced by abrasive, adhesive and fatigue wear and cause a liberation of metal ions, which lead to another immune response elicited mostly by macrophages. Another reaction in the neocapsule can be a type IV hypersensitivity reaction to various alloys, containing metals such as nickel, cobalt and chromium. Patch testing and the lymphocyte transformation test (LTT) are not the best diagnostic possibilities to exclude a postoperative hypersensitivity reaction, because of the different alignment of the epicutaneous cells compared to the periprosthetic deep tissue. This hypersensitivity reaction is mostly induced by cytokines, which are secreted by macrophages rather than lymphocytes. In cell cultures and in animal studies, multipotent mesenchymal stem cells (MSC) have been shown to play a role in improving initial implant integration, to limit periprosthetic osteolysis and also to reconstitute peri-implant bone stock during implant revision. Thus, MSC might be used in the future to prolong the durability of THA. A better understanding of the interactions between primary chronic inflammation, corrosion, osteolysis and hypersensitivity is mandatory to develop new therapeutic strategies, aiming at the reduction of the incidence of implant failures. In this article the underlying immunological mechanisms to aseptic loosening are presented.
Collapse
Affiliation(s)
- Maximilian D Costa
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Department of Morphological Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Stefanie Donner
- Centre for Musculoskeletal Surgery, Charité-University Medicine, Berlin, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Ovidiu-Laurean Pop
- Department of Morphological Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Christoph H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| |
Collapse
|
17
|
Milota T, Smetanova J, Skotnicova A, Rataj M, Lastovicka J, Zelena H, Parackova Z, Fejtkova M, Kanderova V, Fronkova E, Rejlova K, Sediva A, Kalina T. Clinical Outcomes, Immunogenicity, and Safety of BNT162b2 Vaccine in Primary Antibody Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:306-314.e2. [PMID: 36379409 DOI: 10.1016/j.jaip.2022.10.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is characterized by an impaired postvaccination response, high susceptibility to respiratory tract infections, and a broad spectrum of noninfectious complications. Thus, patients with CVID may be at high risk for COVID-19, and vaccination's role in prevention is questionable. OBJECTIVE We evaluated the clinical outcomes, safety, and dynamics of humoral and T-cell immune responses induced by the mRNA vaccine BNT162b2 in CVID. METHODS This prospective observational cohort study focused on the clinical outcomes (proportion of infected patients and disease severity), safety (incidences of adverse events and changes in laboratory parameters), and dynamics of humoral (specific postvaccination and virus-neutralizing antibody assessment) and T-cell immune responses (anti-SARS-CoV-2-specific T-cell detection) in 21 patients with CVID after a two-dose administration of BNT162b2. The patients were observed for 6 months. RESULTS Humoral response was observed in 52% of patients (11 of 21) at month 1 after vaccination but continuously decreased to 33.3% at month 6 (five of 15). Nevertheless, they had a remarkably lower anti-SARS-CoV-2 neutralizing antibody titer compared with healthy controls. The T-cell response was measurable in 46% of patients with CVID (six of 13) at month 1 and persisted over the study period. Mild infection occurred in three patients within the follow-up period (14.3%). The vaccine also exhibited a favorable safety profile. CONCLUSIONS The BNT162b2 vaccine elicited a measurable antibody response in a high proportion of patients, but it was limited by low titer of virus-neutralizing antibodies and rapid waning of anti-receptor-binding domain SARS-CoV-2-specific antibodies. T-cell response was detected in one-third of patients and remained stable within the follow-up period. Vaccination has favorable safety and clinical-related outcomes in preventing severe COVID-19.
Collapse
Affiliation(s)
- Tomas Milota
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic.
| | - Jitka Smetanova
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Aneta Skotnicova
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Michal Rataj
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jan Lastovicka
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Hana Zelena
- Department of Virology, Public Health Institute, Ostrava, Czech Republic
| | - Zuzana Parackova
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Martina Fejtkova
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Veronika Kanderova
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Eva Fronkova
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Katerina Rejlova
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Anna Sediva
- Department of Immunology, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Tomas Kalina
- Childhood Leukemia Investigation Prague, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
18
|
Massalska M, Ciechomska M, Kuca-Warnawin E, Burakowski T, Kornatka A, Radzikowska A, Pawlak D, Muz B, Loniewska-Lwowska A, Palucha A, Maldyk P, Maslinski W. Effectiveness of Soluble CTLA-4-Fc in the Inhibition of Bone Marrow T-Cell Activation in Context of Indoleamine 2.3-Dioxygenase (IDO) and CD4 +Foxp3 + Treg Induction. J Inflamm Res 2022; 15:6813-6829. [PMID: 36578517 PMCID: PMC9792113 DOI: 10.2147/jir.s359775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease with systemic inflammation finally resulting in damaged joints. One of the RA development models suggests bone marrow (BM) as a place of inflammation development further leading to disease progression. We aimed to investigate the potential of CTLA-4-Fc molecule in inducing tolerogenic milieu in BM measured as indoleamine 2,3-dioxygenase (IDO) expression, CD4+Foxp3+ Treg induction, and T cell activation control. The expression of IDO-pathway genes was also examined in monocytes to estimate the tolerogenic potential in the periphery. Methods Bone marrow mononuclear cells (BMMC) were stimulated by pro-inflammatory cytokines and CTLA-4-Fc. Next IDO expression, CD4+CD69+ and CD4+Foxp3+ percentage were estimated by PCR and FACS staining, respectively. Enzymatic activity of IDO was confirmed by HPLC in BM plasma and blood plasma. Genes expressed in IDO-pathway were analyzed by NGS in peripheral monocytes isolated from RA patients and healthy controls. Results We found that CTLA-4-Fc and IFN-γ stimulation results in IDO production by BMMC. CTLA-4-Fc induced tryptophan catabolism can inhibit mitogen-induced CD4+ T cells activation without influencing CD8+ cells, but did not control CD25 nor Foxp3 expression in BM cells. Significantly higher expression of selected IDO-pathway genes was detected on peripheral monocytes isolated from RA as compared to healthy controls. Conclusion This study sheds light on some immunosuppression aspects present or induced in BM. The potential of IDO-mediated pathways were confirmed in the periphery, what may represent the promising candidates for therapeutic strategies in RA.
Collapse
Affiliation(s)
- Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland,Correspondence: Magdalena Massalska, Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, Spartanska 1, Warsaw, 02-637, Poland, Tel/Fax +48 22 670 94 94, Email
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| | - Tomasz Burakowski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| | - Anna Radzikowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, 15-222, Poland
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | | | | | - Pawel Maldyk
- Department of Rheumoorthopaedic Surgery, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland,Clinical Department of Orthopaedic and Traumatology of Locomotor System, Enfant-Jesus Clinical Hospital, Warsaw, 02-005, Poland
| | - Wlodzimierz Maslinski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, 02-637, Poland
| |
Collapse
|
19
|
Shi L, Liu C, Xiong H, Shi D. Elevation of IgE in patients with psoriasis: Is it a paradoxical phenomenon? Front Med (Lausanne) 2022; 9:1007892. [PMID: 36314037 PMCID: PMC9606585 DOI: 10.3389/fmed.2022.1007892] [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: 07/31/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Immunoglobulin E (IgE) elevation is a hallmark of allergic conditions such as atopic dermatitis (AD). The pathogenesis of AD is typically associated with high levels of IL-4 and IL-13 produced by activated T helper 2 (Th2) cells. Psoriasis, on the other hand, is an inflammatory skin disease mainly driven by Th17 cells and their related cytokines. Although the immunopathologic reactions and clinical manifestations are often easily distinguished in the two skin conditions, patients with psoriasis may sometimes exhibit AD-like manifestations, such as elevated IgE and persistent pruritic lesions. Given the fact that the effective T cells have great plasticity to re-differentiate in response to innate and environmental factors, this unusual skin condition could be a consequence of a cross-reaction between distinct arms of T-cell and humoral immunity. Here we review the literature concerning the roles of IgE in the development of AD and psoriasis, showing that elevated IgE seems to be an important indicator for this non-typical psoriasis.
Collapse
Affiliation(s)
- Leyao Shi
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China,The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China
| | - Chen Liu
- The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China
| | - Huabao Xiong
- Basic Medical School, Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China,Huabao Xiong
| | - Dongmei Shi
- The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China,Department of Dermatology, Jining No.1 People's Hospital, Jining, China,*Correspondence: Dongmei Shi
| |
Collapse
|
20
|
Li J, Reinke S, Shen Y, Schollmeyer L, Liu YC, Wang Z, Hardt S, Hipfl C, Hoffmann U, Frischbutter S, Chang HD, Alexander T, Perka C, Radbruch H, Qin Z, Radbruch A, Dong J. A ubiquitous bone marrow reservoir of preexisting SARS-CoV-2-reactive memory CD4+ T lymphocytes in unexposed individuals. Front Immunol 2022; 13:1004656. [PMID: 36268016 PMCID: PMC9576920 DOI: 10.3389/fimmu.2022.1004656] [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: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating, blood-borne SARS-CoV-2-reactive memory T cells in persons so far unexposed to SARS-CoV-2 or the vaccines have been described in 20-100% of the adult population. They are credited with determining the efficacy of the immune response in COVID-19. Here, we demonstrate the presence of preexisting memory CD4+ T cells reacting to peptides of the spike, membrane, or nucleocapsid proteins of SARS-CoV-2 in the bone marrow of all 17 persons investigated that had previously not been exposed to SARS-CoV-2 or one of the vaccines targeting it, with only 15 of these persons also having such cells detectable circulating in the blood. The preexisting SARS-CoV-2-reactive memory CD4+ T cells of the bone marrow are abundant and polyfunctional, with the phenotype of central memory T cells. They are tissue-resident, at least in those persons who do not have such cells in the blood, and about 30% of them express CD69. Bone marrow resident SARS-CoV-2-reactive memory CD4+ memory T cells are also abundant in vaccinated persons analyzed 10-168 days after 1°-4° vaccination. Apart from securing the bone marrow, preexisting cross-reactive memory CD4+ T cells may play an important role in shaping the systemic immune response to SARS-CoV-2 and the vaccines, and contribute essentially to the rapid establishment of long-lasting immunity provided by memory plasma cells, already upon primary infection.
Collapse
Affiliation(s)
- Jinchan Li
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Simon Reinke
- Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Yu Shen
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Lena Schollmeyer
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Yuk-Chien Liu
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Zixu Wang
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christian Hipfl
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ute Hoffmann
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
- Schwiete-Laboratory for Microbiota and Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Stefan Frischbutter
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Berlin, Germany
| | - Hyun-Dong Chang
- Schwiete-Laboratory for Microbiota and Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Radbruch
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
- *Correspondence: Jun Dong,
| |
Collapse
|
21
|
Aradottir Pind AA, Thorsdottir S, Magnusdottir G, Meinke A, Del Giudice G, Jonsdottir I, Bjarnarson SP. A comparative study of adjuvants effects on neonatal plasma cell survival niche in bone marrow and persistence of humoral immune responses. Front Immunol 2022; 13:904415. [PMID: 35990686 PMCID: PMC9381929 DOI: 10.3389/fimmu.2022.904415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
The neonatal immune system is distinct from the immune system of older individuals rendering neonates vulnerable to infections and poor responders to vaccination. Adjuvants can be used as tools to enhance immune responses to co-administered antigens. Antibody (Ab) persistence is mediated by long-lived plasma cells that reside in specialized survival niches in the bone marrow, and transient Ab responses in early life have been associated with decreased survival of plasma cells, possibly due to lack of survival factors. Various cells can secrete these factors and which cells are the main producers is still up for debate, especially in early life where this has not been fully addressed. The receptor BCMA and its ligand APRIL have been shown to be important in the maintenance of plasma cells and Abs. Herein, we assessed age-dependent maturation of a broad range of bone marrow accessory cells and their expression of the survival factors APRIL and IL-6. Furthermore, we performed a comparative analysis of the potential of 5 different adjuvants; LT-K63, mmCT, MF59, IC31 and alum, to enhance expression of survival factors and BCMA following immunization of neonatal mice with tetanus toxoid (TT) vaccine. We found that APRIL expression was reduced in the bone marrow of young mice whereas IL-6 expression was higher. Eosinophils, macrophages, megakaryocytes, monocytes and lymphocytes were important secretors of survival factors in early life but undefined cells also constituted a large fraction of secretors. Immunization and adjuvants enhanced APRIL expression but decreased IL-6 expression in bone marrow cells early after immunization. Furthermore, neonatal immunization with adjuvants enhanced the proportion of plasmablasts and plasma cells that expressed BCMA both in spleen and bone marrow. Enhanced BCMA expression correlated with enhanced vaccine-specific humoral responses, even though the effect of alum on BCMA was less pronounced than those of the other adjuvants at later time points. We propose that low APRIL expression in bone marrow as well as low BCMA expression of plasmablasts/plasma cells in early life together cause transient Ab responses and could represent targets to be triggered by vaccine adjuvants to induce persistent humoral immune responses in this age group.
Collapse
Affiliation(s)
- Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sigrun Thorsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Gudbjorg Julia Magnusdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- *Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|
22
|
Barakos GP, Hatzimichael E. Microenvironmental Features Driving Immune Evasion in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Diseases 2022; 10:diseases10020033. [PMID: 35735633 PMCID: PMC9221594 DOI: 10.3390/diseases10020033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
Bone marrow, besides the known functions of hematopoiesis, is an active organ of the immune system, functioning as a sanctuary for several mature immune cells. Moreover, evidence suggests that hematopoietic stem cells (the bone marrow’s functional unit) are capable of directly sensing and responding to an array of exogenous stimuli. This chronic immune stimulation is harmful to normal hematopoietic stem cells, while essential for the propagation of myeloid diseases, which show a dysregulated immune microenvironment. The bone marrow microenvironment in myelodysplastic syndromes (MDS) is characterized by chronic inflammatory activity and immune dysfunction, that drive excessive cellular death and through immune evasion assist in cancer cell expansion. Acute myeloid leukemia (AML) is another example of immune response failure, with features that augment immune evasion and suppression. In this review, we will outline some of the functions of the bone marrow with immunological significance and describe the alterations in the immune landscape of MDS and AML that drive disease progression.
Collapse
Affiliation(s)
- Georgios Petros Barakos
- First Department of Internal Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece;
| | - Eleftheria Hatzimichael
- Department of Haematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece
- Correspondence:
| |
Collapse
|
23
|
CD4+ and CD8+ T-cell responses in bone marrow to fatty acids in high-fat diets. J Nutr Biochem 2022; 107:109057. [DOI: 10.1016/j.jnutbio.2022.109057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/23/2022] [Accepted: 04/06/2022] [Indexed: 12/30/2022]
|
24
|
Efficient human-like antibody repertoire and hybridoma production in trans-chromosomic mice carrying megabase-sized human immunoglobulin loci. Nat Commun 2022; 13:1841. [PMID: 35383174 PMCID: PMC8983744 DOI: 10.1038/s41467-022-29421-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/15/2022] [Indexed: 11/15/2022] Open
Abstract
Trans-chromosomic (Tc) mice carrying mini-chromosomes with megabase-sized human immunoglobulin (Ig) loci have contributed to the development of fully human therapeutic monoclonal antibodies, but mitotic instability of human mini-chromosomes in mice may limit the efficiency of hybridoma production. Here, we establish human antibody-producing Tc mice (TC-mAb mice) that stably maintain a mouse-derived, engineered chromosome containing the entire human Ig heavy and kappa chain loci in a mouse Ig-knockout background. Comprehensive, high-throughput DNA sequencing shows that the human Ig repertoire, including variable gene usage, is well recapitulated in TC-mAb mice. Despite slightly altered B cell development and a delayed immune response, TC-mAb mice have more subsets of antigen-specific plasmablast and plasma cells than wild-type mice, leading to efficient hybridoma production. Our results thus suggest that TC-mAb mice offer a valuable platform for obtaining fully human therapeutic antibodies, and a useful model for elucidating the regulation of human Ig repertoire formation. Trans-chromosomic (Tc) mice have helped the development of therapeutic antibodies, but chromosome instability limits its application. Here the authors develop a new line of Tc mice with full human Ig heavy and kappa loci integrated into the mouse artificial chromosome for stable passage, and confirm efficient generation of B cell responses and specific antibodies.
Collapse
|
25
|
Knop L, Spanier J, Larsen PK, Witte A, Bank U, Dunay IR, Kalinke U, Schüler T. IFNAR signaling in fibroblastic reticular cells can modulate CD8 + memory fate decision. Eur J Immunol 2022; 52:895-906. [PMID: 35365883 DOI: 10.1002/eji.202149760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022]
Abstract
CD8+ memory T cells (TM ) are crucial for the long-term protection from infections and cancer. Multiple cell types and cytokines are involved in the regulation of CD8+ T cell responses and subsequent TM formation. Besides their direct antiviral effects, type I interferons (IFN-α/β) modulate CD8+ T cell immunity via their action on several immune cell subsets. However, it is largely unclear how non-immune cells are involved in this multicellular network modulating CD8+ TM formation. Fibroblastic reticular cells (FRCs), form the three-dimensional scaffold of secondary lymphoid organs, express the IFN-α/β receptor (IFNAR) and modulate adaptive immune responses. However, it is unclear whether and how early IFNAR signals in lymph node (LN) FRCs affect CD8+ TM differentiation. Using peptide vaccination and viral infection, we studied CD8+ TM differentiation in mice with a FRC-specific IFNAR deletion (FRCΔIFNAR ). We show here that the differentiation of CD8+ TCR-transgenic T cells into central memory cells (TCM ) is enhanced in peptide-vaccinated FRCΔIFNAR mice. Conversely, vesicular stomatitis virus (VSV) infection of FRCΔIFNAR mice is associated with impaired TCM formation and the accumulation of VSV-specific double-positive (dp) CD127lo KLRG-1hi effector memory T cells. In summary, we provide evidence for a context-dependent contribution of FRC-specific IFNAR signaling to CD8+ TM differentiation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laura Knop
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Amelie Witte
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ute Bank
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| |
Collapse
|
26
|
Oncolytic Vaccinia Virus Augments T Cell Factor 1-Positive Stem-like CD8+ T Cells, Which Underlies the Efficacy of Anti-PD-1 Combination Immunotherapy. Biomedicines 2022; 10:biomedicines10040805. [PMID: 35453555 PMCID: PMC9027961 DOI: 10.3390/biomedicines10040805] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 12/28/2022] Open
Abstract
Oncolytic virotherapy has garnered attention as an antigen-agnostic therapeutic cancer vaccine that induces cancer-specific T cell responses without additional antigen loading. As anticancer immune responses are compromised by a lack of antigenicity and chronic immunosuppressive microenvironments, an effective immuno-oncology modality that converts cold tumors into hot tumors is crucial. To evaluate the immune-activating characteristics of oncolytic vaccinia virus (VACV; JX-594, pexastimogene devacirepvec), diverse murine syngeneic cancer models with different tissue types and immune microenvironments were used. Intratumorally administered mJX-594, a murine variant of JX-594, potently increased CD8+ T cells, including antigen-specific cancer CD8+ T cells, and decreased immunosuppressive cells irrespective of tissue type or therapeutic efficacy. Remodeling of tumors into inflamed ones by mJX-594 led to a response to combined anti-PD-1 treatment, but not to mJX-594 or anti-PD-1 monotherapy. mJX-594 treatment increased T cell factor 1-positive stem-like T cells among cancer-specific CD8+ T cells, and anti-PD-1 combination treatment further increased proliferation of these cells, which was important for therapeutic efficacy. The presence of functional cancer-specific CD8+ T cells in the spleen and bone marrow for an extended period, which proliferated upon encountering cancer antigen-loaded splenic dendritic cells, further indicated that long-term durable anticancer immunity was elicited by oncolytic VACV.
Collapse
|
27
|
Cendón C, Du W, Durek P, Liu YC, Alexander T, Serene L, Yang X, Gasparoni G, Salhab A, Nordström K, Lai T, Schulz AR, Rao A, Heinz GA, Stefanski AL, Claußnitzer A, Siewert K, Dörner T, Chang HD, Volk HD, Romagnani C, Qin Z, Hardt S, Perka C, Reinke S, Walter J, Mashreghi MF, Thurley K, Radbruch A, Dong J. Resident memory CD4+ T lymphocytes mobilize from bone marrow to contribute to a systemic secondary immune reaction. Eur J Immunol 2022; 52:737-752. [PMID: 35245389 DOI: 10.1002/eji.202149726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/27/2022] [Accepted: 03/01/2022] [Indexed: 11/12/2022]
Abstract
Resident memory T lymphocytes (TRM ) of epithelial tissues and the bone marrow protect their host tissue. To what extent these cells are mobilized and contribute to systemic immune reactions is less clear. Here we show that in secondary immune reactions to the measles-mumps-rubella (MMR) vaccine, CD4+ TRM are mobilized into the blood within 16 to 48 hours after immunization in humans. This mobilization of TRM is cognate: TRM recognizing other antigens are not mobilized, unless they cross-react with the vaccine. We also demonstrate through methylome analyses that TRM are mobilized from the bone marrow. These mobilized cells make significant contribution to the systemic immune reaction, as evidenced by their T-cell receptor Vβ clonotypes represented among the newly generated circulating memory T-cells, 14 days after vaccination. Thus, TRM of the bone marrow confer not only local, but also systemic immune memory. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Carla Cendón
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Weijie Du
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Pawel Durek
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Yuk-Chien Liu
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lindsay Serene
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Xinyi Yang
- Otto-Warburg-Laboratory, Computational Epigenomics, Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
| | - Gilles Gasparoni
- Department of Genetics, University of Saarland (UdS), Campus, Saarbrücken, 66123, Germany
| | - Abdulrahman Salhab
- Department of Genetics, University of Saarland (UdS), Campus, Saarbrücken, 66123, Germany
| | - Karl Nordström
- Department of Genetics, University of Saarland (UdS), Campus, Saarbrücken, 66123, Germany
| | - Tina Lai
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Axel R Schulz
- Mass Cytometry, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Anna Rao
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Gitta A Heinz
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Ana L Stefanski
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anne Claußnitzer
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katherina Siewert
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hyun-Dong Chang
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.,Schwiete-Laboratory for Microbiota and Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.,Medical Department / Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Bejing, China.,University of Chinese Academy of Sciences, Bejing, China.,Zhengzhou University, Zhengzhou, China
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Reinke
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn Walter
- Department of Genetics, University of Saarland (UdS), Campus, Saarbrücken, 66123, Germany
| | - Mir-F Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kevin Thurley
- Systems Biology of Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.,Institute for Theoretical Biology, Humboldt University Berlin, Germany
| | - Andreas Radbruch
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| |
Collapse
|
28
|
Meylan M, Petitprez F, Becht E, Bougoüin A, Pupier G, Calvez A, Giglioli I, Verkarre V, Lacroix G, Verneau J, Sun CM, Laurent-Puig P, Vano YA, Elaïdi R, Méjean A, Sanchez-Salas R, Barret E, Cathelineau X, Oudard S, Reynaud CA, de Reyniès A, Sautès-Fridman C, Fridman WH. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity 2022; 55:527-541.e5. [PMID: 35231421 DOI: 10.1016/j.immuni.2022.02.001] [Citation(s) in RCA: 374] [Impact Index Per Article: 124.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/09/2021] [Accepted: 02/02/2022] [Indexed: 12/30/2022]
Abstract
The presence of intratumoral tertiary lymphoid structures (TLS) is associated with positive clinical outcomes and responses to immunotherapy in cancer. Here, we used spatial transcriptomics to examine the nature of B cell responses within TLS in renal cell carcinoma (RCC). B cells were enriched in TLS, and therein, we could identify all B cell maturation stages toward plasma cell (PC) formation. B cell repertoire analysis revealed clonal diversification, selection, expansion in TLS, and the presence of fully mature clonotypes at distance. In TLS+ tumors, IgG- and IgA-producing PCs disseminated into the tumor beds along fibroblastic tracks. TLS+ tumors exhibited high frequencies of IgG-producing PCs and IgG-stained and apoptotic malignant cells, suggestive of anti-tumor effector activity. Therapeutic responses and progression-free survival correlated with IgG-stained tumor cells in RCC patients treated with immune checkpoint inhibitors. Thus, intratumoral TLS sustains B cell maturation and antibody production that is associated with response to immunotherapy, potentially via direct anti-tumor effects.
Collapse
Affiliation(s)
- Maxime Meylan
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Florent Petitprez
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, 75013 Paris, France; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Etienne Becht
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, 75013 Paris, France
| | - Antoine Bougoüin
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Guilhem Pupier
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Anne Calvez
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Ilenia Giglioli
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Virginie Verkarre
- Département de pathologie, Hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris - Paris Centre, 75015 Paris, France; Université de Paris, 75006 Paris, France; PARCC, INSERM, Equipe Labellisée Ligue contre le Cancer, 75015 Paris, France
| | - Guillaume Lacroix
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Johanna Verneau
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Chen-Ming Sun
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, 75006 Paris, France
| | - Yann-Alexandre Vano
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France; Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, 75013 Paris, France; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; Département d'oncologie médicale, Hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris - Paris Centre, F-75015 Paris, France; Université de Paris, 75006 Paris, France; PARCC, INSERM, Equipe Labellisée Ligue contre le Cancer, 75015 Paris, France
| | - Reza Elaïdi
- Association pour la Recherche de Thérapeutiques Innovantes en Cancérologie, 75015 Paris, France
| | - Arnaud Méjean
- Département d'urologie, Hôpital européen Georges Pompidou, Université de Paris, 75015 Paris, France
| | - Rafaël Sanchez-Salas
- Département d'urologie, Institut Mutualiste Montsouris, Université de Paris, 75014 Paris, France
| | - Eric Barret
- Département d'urologie, Institut Mutualiste Montsouris, Université de Paris, 75014 Paris, France
| | - Xavier Cathelineau
- Département d'urologie, Institut Mutualiste Montsouris, Université de Paris, 75014 Paris, France
| | - Stephane Oudard
- Département d'oncologie médicale, Hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris - Paris Centre, F-75015 Paris, France; Université de Paris, 75006 Paris, France
| | - Claude-Agnès Reynaud
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMRS8253, Université de Paris, 75015 Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, 75013 Paris, France; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, 75006 Paris, France; Université de Paris, 75006 Paris, France
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Wolf Herman Fridman
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France.
| |
Collapse
|
29
|
Mun Y, Fazio S, Arrieta CN. Remodeling of the Bone Marrow Stromal Microenvironment During Pathogenic Infections. Curr Top Microbiol Immunol 2021; 434:55-81. [PMID: 34850282 DOI: 10.1007/978-3-030-86016-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The bone marrow (BM) is the primary hematopoietic organ and a hub in which organismal demands for blood cellular output are systematically monitored. BM tissues are additionally home to a plethora of mature immune cell types, providing functional environments for the activation of immune responses and acting as preferred anatomical reservoirs for cells involved in immunological memory. Stromal cells of the BM microenvironment crucially govern different aspects of organ function, by structuring tissue microanatomy and by directly providing essential regulatory cues to hematopoietic and immune components in distinct niches. Emerging evidence demonstrates that stromal networks are endowed with remarkable functional and structural plasticity. Stress-induced adaptations of stromal cells translate into demand-driven hematopoiesis. Furthermore, aberrations of stromal integrity arising from pathological conditions critically contribute to the dysregulation of BM function. Here, we summarize our current understanding of the alterations that pathogenic infections and ensuing inflammatory conditions elicit on the global topography of the BM microenvironment, the integrity of anatomical niches and cellular interactions, and ultimately, on the regulatory function of diverse stromal subsets.
Collapse
Affiliation(s)
- YeVin Mun
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland
| | - Serena Fazio
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland
| | - César Nombela Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland.
| |
Collapse
|
30
|
Cell-based therapeutics for the treatment of hematologic diseases inside the bone marrow. J Control Release 2021; 339:1-13. [PMID: 34536449 DOI: 10.1016/j.jconrel.2021.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
Cell-based therapies could overcome the limitations of traditional drugs for the treatment of refractory diseases. Cell exchange between the bone marrow and blood is bidirectional. Several kinds of cells in the blood have the capability to enter the bone marrow by interacting with sinusoidal cells under specific physiological or pathological conditions. These cells are the potential living therapeutics or delivery vehicles to treat or prevent bone marrow-related hematologic diseases. In this review, we summarized the in vivo molecular mechanisms and kinetics of these cells in entering the bone marrow. The advances in the fabrication of living cell drugs and the strategies to design cell-based carriers into the bone marrow were discussed. The latest studies on how to use blood cells as living drugs or as drug carriers to improve therapeutic outcomes of hematologic diseases inside the bone marrow were highlighted.
Collapse
|
31
|
Ise W, Kurosaki T. Plasma cell generation during T cell-dependent immune responses. Int Immunol 2021; 33:797-801. [PMID: 34536284 DOI: 10.1093/intimm/dxab071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Plasma cells are terminally differentiated from activated B cells and are specialized for secreting antibodies, which are essential effector molecules in humoral immunity to neutralize invading pathogens. Upon challenge with T cell-dependent antigens, plasma cells can be generated during the primary extrafollicular response, the germinal center (GC) response or the secondary memory response. Recent studies have revealed that plasma cell generation is regulated not only by several key transcription factors, but also by epigenetic modifications. In addition, the differentiation of GC B cells toward a plasma cell fate is associated with affinity for antigens and is determined by the strength of contact with T follicular helper cells.
Collapse
Affiliation(s)
- Wataru Ise
- Regulation of host defense team, Division of microbiology and Immunology, Center for infectious Disease Education and Research, Osaka University, Osaka, Japan.,Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of microbiology and Immunology, Center for infectious Disease Education and Research, Osaka University, Osaka, Japan.,Laboratory of Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, Japan
| |
Collapse
|
32
|
Müller-Winkler J, Mitter R, Rappe JCF, Vanes L, Schweighoffer E, Mohammadi H, Wack A, Tybulewicz VLJ. Critical requirement for BCR, BAFF, and BAFFR in memory B cell survival. J Exp Med 2021; 218:211510. [PMID: 33119032 PMCID: PMC7604764 DOI: 10.1084/jem.20191393] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 07/22/2020] [Accepted: 09/14/2020] [Indexed: 01/23/2023] Open
Abstract
Memory B cells (MBCs) are long-lived cells that form a critical part of immunological memory, providing rapid antibody responses to recurring infections. However, very little is known about signals controlling MBC survival. Previous work has shown that antigen is not required for MBC survival, but a requirement for the B cell antigen receptor (BCR) has not been tested. Other studies have shown that, unlike naive B cells, MBCs do not express BAFFR and their survival is independent of BAFF, the ligand for BAFFR. Here, using inducible genetic ablation, we show that survival of MBCs is critically dependent on the BCR and on signaling through the associated CD79A protein. Unexpectedly, we found that MBCs express BAFFR and that their survival requires BAFF and BAFFR; hence, loss of BAFF or BAFFR impairs recall responses. Finally, we show that MBC survival requires IKK2, a kinase that transduces BAFFR signals. Thus, MBC survival is critically dependent on signaling from BCR and BAFFR.
Collapse
|
33
|
Cornelis R, Chang HD, Radbruch A. Keeping up with the stress of antibody production: BAFF and APRIL maintain memory plasma cells. Curr Opin Immunol 2021; 71:97-102. [PMID: 34303157 DOI: 10.1016/j.coi.2021.06.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022]
Abstract
Memory plasma cells, also called long-lived plasma cells, provide 'humoral immunity' by continued secretion of protective antibodies against pathogens, which the immune system has once encountered. They are maintained mainly in the bone marrow, docking on to stromal cells individually. In those niches they can apparently persist for decades (Chang et al., 2018 [1]). Integrin-mediated contact to the stromal cell provides an essential survival signal to the plasma cell, activating the PI3K signalling pathway, downregulating FoxO1/3a and repressing the activation of caspases 3 and 7. In a redundant form, the cytokines BAFF and APRIL, ligands of the plasma cell receptors TACI and BCMA, provide a second essential survival signal, preventing activation of caspase 12, as triggered by endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Rebecca Cornelis
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Germany
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Germany; Institute of Biotechnology, Technische Universität Berlin, Germany.
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Germany
| |
Collapse
|
34
|
The Bone Marrow as Sanctuary for Plasma Cells and Memory T-Cells: Implications for Adaptive Immunity and Vaccinology. Cells 2021; 10:cells10061508. [PMID: 34203839 PMCID: PMC8232593 DOI: 10.3390/cells10061508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
The bone marrow (BM) is key to protective immunological memory because it harbors a major fraction of the body’s plasma cells, memory CD4+ and memory CD8+ T-cells. Despite its paramount significance for the human immune system, many aspects of how the BM enables decade-long immunity against pathogens are still poorly understood. In this review, we discuss the relationship between BM survival niches and long-lasting humoral immunity, how intrinsic and extrinsic factors define memory cell longevity and show that the BM is also capable of adopting many responsibilities of a secondary lymphoid organ. Additionally, with more and more data on the differentiation and maintenance of memory T-cells and plasma cells upon vaccination in humans being reported, we discuss what factors determine the establishment of long-lasting immunological memory in the BM and what we can learn for vaccination technologies and antigen design. Finally, using these insights, we touch on how this holistic understanding of the BM is necessary for the development of modern and efficient vaccines against the pandemic SARS-CoV-2.
Collapse
|
35
|
Stromal Cell-Contact Dependent PI3K and APRIL Induced NF-κB Signaling Prevent Mitochondrial- and ER Stress Induced Death of Memory Plasma Cells. Cell Rep 2021; 32:107982. [PMID: 32755576 PMCID: PMC7408492 DOI: 10.1016/j.celrep.2020.107982] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/25/2020] [Accepted: 07/09/2020] [Indexed: 01/06/2023] Open
Abstract
The persistence of long-lived memory plasma cells in the bone marrow depends on survival factors available in the bone marrow, which are provided in niches organized by stromal cells. Using an ex vivo system in which we supply the known survival signals, direct cell contact to stromal cells, and the soluble cytokine a proliferation-inducing ligand (APRIL), we have elucidated the critical signaling pathways required for the survival of long-lived plasma cells. Integrin-mediated contact of bone marrow plasma cells with stromal cells activates the phosphatidylinositol 3-kinase (PI3K) signaling pathway, leading to critical inactivation of Forkhead-Box-Protein O1/3 (FoxO1/3) and preventing the activation of mitochondrial stress-associated effector caspases 3 and 7. Accordingly, inhibition of PI3K signaling in vivo ablates bone marrow plasma cells. APRIL signaling, by the nuclear factor κB (NF-κB) pathway, blocks activation of the endoplasmic-reticulum-stress-associated initiator caspase 12. Thus, stromal-cell-contact-induced PI3K and APRIL-induced NF-κB signaling provide the necessary and complementary signals to maintain bone marrow memory plasma cells.
Collapse
|
36
|
Nikolaou C, Muehle K, Schlickeiser S, Japp AS, Matzmohr N, Kunkel D, Frentsch M, Thiel A. High-dimensional single cell mass cytometry analysis of the murine hematopoietic system reveals signatures induced by ageing and physiological pathogen challenges. IMMUNITY & AGEING 2021; 18:20. [PMID: 33879187 PMCID: PMC8056611 DOI: 10.1186/s12979-021-00230-3] [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: 10/16/2020] [Accepted: 03/26/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Immune ageing is a result of repetitive microbial challenges along with cell intrinsic or systemic changes occurring during ageing. Mice under 'specific-pathogen-free' (SPF) conditions are frequently used to assess immune ageing in long-term experiments. However, physiological pathogenic challenges are reduced in SPF mice. The question arises to what extent murine experiments performed under SPF conditions are suited to analyze immune ageing in mice and serve as models for human immune ageing. Our previous comparisons of same aged mice with different microbial exposures, unambiguously identified distinct clusters of immune cells characteristic for numerous previous pathogen encounters in particular in pet shop mice. RESULTS We here performed single cell mass cytometry assessing splenic as secondary and bone marrow as primary lymphoid organ-derived leukocytes isolated from young versus aged SPF mice in order to delineate alterations of the murine hematopoietic system induced during ageing. We then compared immune clusters from young and aged SPF mice to pet shop mice in order to delineate alterations of the murine hematopoietic system induced by physiological pathogenic challenges and those caused by cell intrinsic or systemic changes during ageing. Notably, distinct immune signatures were similarly altered in both pet shop and aged SPF mice in comparison to young SPF mice, including increased frequencies of memory T lymphocytes, effector-cytokine producing T cells, plasma cells and mature NK cells. However, elevated frequencies of CD4+ T cells, total NK cells, granulocytes, pDCs, cDCs and decreased frequencies of naïve B cells were specifically identified only in pet shop mice. In aged SPF mice specifically the frequencies of splenic IgM+ plasma cells, CD8+ T cells and CD4+ CD25+ Treg were increased as compared to pet shop mice and young mice. CONCLUSIONS Our study dissects firstly how ageing impacts both innate and adaptive immune cells in primary and secondary lymphoid organs. Secondly, it partly distinguishes murine intrinsic immune ageing alterations from those induced by physiological pathogen challenges highlighting the importance of designing mouse models for their use in preclinical research including vaccines and immunotherapies.
Collapse
Affiliation(s)
- Christos Nikolaou
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany. .,Institute for Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany. .,Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - Kerstin Muehle
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Schlickeiser
- Institute for Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Flow & Mass Cytometry Core Facility, Charité - Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Alberto Sada Japp
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Matzmohr
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Desiree Kunkel
- Flow & Mass Cytometry Core Facility, Charité - Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Marco Frentsch
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Thiel
- Regenerative Immunology and Aging, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
37
|
Immunological memory in rheumatic inflammation - a roadblock to tolerance induction. Nat Rev Rheumatol 2021; 17:291-305. [PMID: 33824526 DOI: 10.1038/s41584-021-00601-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Why do we still have no cure for chronic inflammatory diseases? One reason could be that current therapies are based on the assumption that chronic inflammation is driven by persistent 'acute' immune reactions. Here we discuss a paradigm shift by suggesting that beyond these reactions, chronic inflammation is driven by imprinted, pathogenic 'memory' cells of the immune system. This rationale is based on the observation that in patients with chronic inflammatory rheumatic diseases refractory to conventional immunosuppressive therapies, therapy-free remission can be achieved by resetting the immune system; that is, by ablating immune cells and regenerating the immune system from stem cells. The success of this approach identifies antigen-experienced and imprinted immune cells as essential and sufficient drivers of inflammation. The 'dark side' of immunological memory primarily involves memory plasma cells secreting pathogenic antibodies and memory T lymphocytes secreting pathogenic cytokines and chemokines, but can also involve cells of innate immunity. New therapeutic strategies should address the persistence of these memory cells. Selective targeting of pathogenic immune memory cells could be based on their specificity, which is challenging, or on their lifestyle, which differs from that of protective immune memory cells, in particular for pathogenic T lymphocytes. The adaptations of such pathogenic memory cells to chronic inflammation offers entirely new therapeutic options for their selective ablation and the regeneration of immunological tolerance.
Collapse
|
38
|
Du W, Lenz D, Köhler R, Zhang E, Cendon C, Li J, Massoud M, Wachtlin J, Bodo J, Hauser AE, Radbruch A, Dong J. Rapid Isolation of Functional ex vivo Human Skin Tissue-Resident Memory T Lymphocytes. Front Immunol 2021; 12:624013. [PMID: 33828548 PMCID: PMC8019735 DOI: 10.3389/fimmu.2021.624013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/23/2021] [Indexed: 12/21/2022] Open
Abstract
Studies in animal models have shown that skin tissue-resident memory T (TRM) cells provide enhanced and immediate effector function at the site of infection. However, analyses of skin TRM cells in humans have been hindered by the lack of an optimized isolation protocol. Here, we present a combinatorial strategy-the 6-h collagenase IV digestion and gentle tissue dissociation – for rapid and efficient isolation of skin TRM cells with skin tissue-specific immune features. In comparison with paired blood circulating memory T cells, these ex vivo isolated skin T cells express typical TRM cell markers and display higher polyfunctional properties. Moreover, these isolated cells can also be assessed for longer periods of time in ex vivo cultures. Thus, the optimized isolation protocol provides a valuable tool for further understanding of human skin TRM cells, especially for direct comparison with peripheral blood T cells at the same sample collection time.
Collapse
Affiliation(s)
- Weijie Du
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Daniel Lenz
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Ralf Köhler
- Central Lab for Microscopy, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | | | - Carla Cendon
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Jinchan Li
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Mona Massoud
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Joachim Wachtlin
- Sankt Gertrauden Krankenhaus, Berlin, Germany.,Medizinische Hochschule Brandenburg, Neurrupin, Germany
| | - Juliane Bodo
- Plastische und Ästhetische Chirurgie, Berlin, Germany
| | - Anja E Hauser
- Central Lab for Microscopy, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany.,Immune Dynamics, Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Radbruch
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin, Institute of the Leibniz Association, Berlin, Germany
| |
Collapse
|
39
|
Stein JV, Ruef N, Wissmann S. Organ-Specific Surveillance and Long-Term Residency Strategies Adapted by Tissue-Resident Memory CD8 + T Cells. Front Immunol 2021; 12:626019. [PMID: 33659008 PMCID: PMC7917134 DOI: 10.3389/fimmu.2021.626019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/26/2021] [Indexed: 01/27/2023] Open
Abstract
Tissue-resident CD8+ T cells (CD8+ TRM) populate lymphoid and non-lymphoid tissues after infections as first line of defense against re-emerging pathogens. To achieve host protection, CD8+ TRM have developed surveillance strategies that combine dynamic interrogation of pMHC complexes on local stromal and hematopoietic cells with long-term residency. Factors mediating CD8+ TRM residency include CD69, a surface receptor opposing the egress-promoting S1P1, CD49a, a collagen-binding integrin, and CD103, which binds E-cadherin on epithelial cells. Moreover, the topography of the tissues of residency may influence TRM retention and surveillance strategies. Here, we provide a brief summary of these factors to examine how CD8+ TRM reconcile constant migratory behavior with their long-term commitment to local microenvironments, with a focus on epithelial barrier organs and exocrine glands with mixed connective-epithelial tissue composition.
Collapse
Affiliation(s)
- Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
40
|
Abstract
The soft marrow tissues, which are found disseminated throughout bone cavities, are prime sites for hematopoietic cell production, development, and control of immune responses, and regulation of skeletal metabolism. These essential functions are executed through the concerted and finely tuned interaction of a large variety of cell types of hematopoietic and nonhematopoietic origin, through yet largely unknown sophisticated molecular mechanisms. A fundamental insight of the biological underpinnings of organ function can be gained from the microscopic study of the bone marrow (BM), its complex structural organization and the existence of cell-specific spatial associations. Albeit the application of advanced imaging techniques to the analysis of BM has historically proved challenging, recent technological developments now enable the interrogation of organ-wide regions of marrow tissues in three dimensions at high resolution. Here, we provide a detailed experimental protocol for the generation of thick slices of BM from murine femoral cavities, the immunostaining of cellular and structural components within these samples, and their optical clearing, which enhances the depth at which optical sectioning can be performed with standard confocal microscopes. Collectively, the experimental pipeline here described allows for the rendering of single-cell resolution, multidimensional reconstructions of vast volumes of the complex BM microenvironment.
Collapse
|
41
|
Current Understanding of Myelomatous Mesenchymal Stromal Cells Extended through Advances in Experimental Methods. Cancers (Basel) 2020; 13:cancers13010025. [PMID: 33374627 PMCID: PMC7793501 DOI: 10.3390/cancers13010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/03/2023] Open
Abstract
Simple Summary As the amount of information available has grown, now it is known that many types of non-hematopoietic cells, including mesenchymal stem/progenitor cells, mature mesenchymal cells, and endothelial cells, as well as mature hematopoietic cells such as monocytes, macrophages, T-cells, and B-cells, have roles in the pathogenesis of multiple myeloma. This review focuses on the role of mesenchymal cells in the microenvironment of multiple myeloma. We summarize the experimental strategies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells. Abstract Multiple myeloma is an incurable cancer formed by malignant plasma cells. For the proliferation and survival of myeloma cells, as well as the occurrence of the complications, numerous intra- and extra-cellular mechanisms are involved. The interaction of myeloma cells with the microenvironment is known to be one of the most critical mechanisms. A specific microenvironment could affect the progression and growth of tumor cells, as well as drug resistance. Among various microenvironment components, such as hematological and non-hematological cells, and soluble factors (cytokines, chemokines, and extracellular matrix (ECM) proteins), in this review, we focus on the role of mesenchymal cells. We aimed to summarize the experimental strategies used for conducting studies and current understanding of the biological roles in the pathogenesis of myeloma. Furthermore, we discuss the possible clinical applications targeting mesenchymal cells.
Collapse
|
42
|
Miao R, Lim VY, Kothapalli N, Ma Y, Fossati J, Zehentmeier S, Sun R, Pereira JP. Hematopoietic Stem Cell Niches and Signals Controlling Immune Cell Development and Maintenance of Immunological Memory. Front Immunol 2020; 11:600127. [PMID: 33324418 PMCID: PMC7726109 DOI: 10.3389/fimmu.2020.600127] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Studies over the last couple of decades have shown that hematopoietic stem cells (HSCs) are critically dependent on cytokines such as Stem Cell Factor and other signals provided by bone marrow niches comprising of mesenchymal stem and progenitor cells (MSPCs) and endothelial cells (ECs). Because of their critical roles in HSC maintenance the niches formed by MSPCs and ECs are commonly referred to as HSC niches. For the most part, the signals required for HSC maintenance act in a short-range manner, which imposes the necessity for directional and positional cues in order for HSCs to localize and be retained properly in stem cell niches. The chemokine CXCL12 and its Gαi protein coupled receptor CXCR4, besides promoting HSC quiescence directly, also play instrumental roles in enabling HSCs to access bone marrow stem cell niches. Recent studies have revealed, however, that HSC niches also provide a constellation of hematopoietic cytokines that are critical for the production of most, if not all, blood cell types. Some hematopoietic cytokines, namely IL-7 and IL-15 produced by HSC niches, are not only required for lymphopoiesis but are also essential for memory T cell maintenance. Consequently, hematopoietic progenitors and differentiated immune cells, such as memory T cell subsets, also depend on the CXCL12/CXCR4 axis for migration into bone marrow and interactions with MSPCs and ECs. Similarly, subsets of antibody-secreting plasma cells also reside in close association with CXCL12-producing MSPCs in the bone marrow and require the CXCR4/CXCL12 axis for survival and long-term maintenance. Collectively, these studies demonstrate a broad range of key physiological roles, spanning blood cell production and maintenance of immunological memory, that are orchestrated by stem cell niches through a common and simple mechanism: CXCL12/CXCR4-mediated cell recruitment followed by receipt of a maintenance and/or instructive signal. A fundamental flaw of this type of cellular organization is revealed by myeloid and lymphoid leukemias, which target stem cell niches and induce profound transcriptomic changes that result in reduced hematopoietic activity and altered mesenchymal cell differentiation.
Collapse
Affiliation(s)
- Runfeng Miao
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Vivian Y Lim
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Neeharika Kothapalli
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Yifan Ma
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Julia Fossati
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Ruifeng Sun
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
43
|
Mitochondria at Work: New Insights into Regulation and Dysregulation of Cellular Energy Supply and Metabolism. Biomedicines 2020; 8:biomedicines8110526. [PMID: 33266387 PMCID: PMC7700424 DOI: 10.3390/biomedicines8110526] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are of great relevance to health, and their dysregulation is associated with major chronic diseases. Research on mitochondria-156 brand new publications from 2019 and 2020-have contributed to this review. Mitochondria have been fundamental for the evolution of complex organisms. As important and semi-autonomous organelles in cells, they can adapt their function to the needs of the respective organ. They can program their function to energy supply (e.g., to keep heart muscle cells going, life-long) or to metabolism (e.g., to support hepatocytes and liver function). The capacity of mitochondria to re-program between different options is important for all cell types that are capable of changing between a resting state and cell proliferation, such as stem cells and immune cells. Major chronic diseases are characterized by mitochondrial dysregulation. This will be exemplified by cardiovascular diseases, metabolic syndrome, neurodegenerative diseases, immune system disorders, and cancer. New strategies for intervention in chronic diseases will be presented. The tumor microenvironment can be considered a battlefield between cancer and immune defense, competing for energy supply and metabolism. Cancer cachexia is considered as a final stage of cancer progression. Nevertheless, the review will present an example of complete remission of cachexia via immune cell transfer. These findings should encourage studies along the lines of mitochondria, energy supply, and metabolism.
Collapse
|
44
|
Aradottir Pind AA, Molina Estupiñan JL, Magnusdottir GJ, Del Giudice G, Jonsdottir I, Bjarnarson SP. LT-K63 Enhances B Cell Activation and Survival Factors in Neonatal Mice That Translates Into Long-Lived Humoral Immunity. Front Immunol 2020; 11:527310. [PMID: 33193301 PMCID: PMC7644473 DOI: 10.3389/fimmu.2020.527310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Adjuvants enhance magnitude and duration of immune responses induced by vaccines. In this study we assessed in neonatal mice if and how the adjuvant LT-K63 given with a pneumococcal conjugate vaccine, Pnc1-TT, could affect the expression of tumor necrosis factor receptor (TNF-R) superfamily members, known to be involved in the initiation and maintenance of antibody responses; B cell activating factor receptor (BAFF-R) and B cell maturation antigen (BCMA) and their ligands, BAFF, and a proliferation inducing ligand (APRIL). Initially we assessed the maturation status of different B cell populations and their expression of BAFF-R and BCMA. Neonatal mice had dramatically fewer B cells than adult mice and the composition of different subsets within the B cell pool differed greatly. Proportionally newly formed B cells were most abundant, but they had diminished BAFF-R expression which could explain low proportions of marginal zone and follicular B cells observed. Limited BCMA expression was also detected in neonatal pre-plasmablasts/plasmablasts. LT-K63 enhanced vaccine-induced BAFF-R expression in splenic marginal zone, follicular and newly formed B cells, leading to increased plasmablast/plasma cells, and their enhanced expression of BCMA in spleen and bone marrow. Additionally, the induction of BAFF and APRIL expression occurred early in neonatal mice immunized with Pnc1-TT either with or without LT-K63. However, BAFF+ and APRIL+ cells in spleens were maintained at a higher level in mice that received the adjuvant. Furthermore, the early increase of APRIL+ cells in bone marrow was more profound in mice immunized with vaccine and adjuvant. Finally, we assessed, for the first time in neonatal mice, accessory cells of the plasma cell niche in bone marrow and their secretion of APRIL. We found that LT-K63 enhanced the frequency and APRIL expression of eosinophils, macrophages, and megakaryocytes, which likely contributed to plasma cell survival, even though APRIL+ cells showed a fast decline. All this was associated with enhanced, sustained vaccine-specific antibody-secreting cells in bone marrow and persisting vaccine-specific serum antibodies. Our study sheds light on the mechanisms behind the adjuvanticity of LT-K63 and identifies molecular pathways that should be triggered by vaccine adjuvants to induce sustained humoral immunity in early life.
Collapse
Affiliation(s)
- Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Jenny Lorena Molina Estupiñan
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gudbjorg Julia Magnusdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
45
|
Hojyo S, Tumes D, Murata A, Tokoyoda K. Multiple developmental pathways lead to the generation of CD4 T-cell memory. Int Immunol 2020; 32:589-595. [PMID: 32766843 DOI: 10.1093/intimm/dxaa051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Long-term immunological memory mediated by CD4 T cells provides a rapid protection against previously encountered pathogens or antigens. However, it is still controversial how memory CD4 T cells are generated and maintained. Unclear definitions of T-cell memory may be partially responsible for this controversy. It is becoming clear that diverse pathways are responsible for the differentiation and long-term persistence of memory T cells. We herein discuss the diversity of memory cell generation, describing a novel population of resting memory CD4 T cells and their precursors.
Collapse
Affiliation(s)
- Shintaro Hojyo
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Berlin, Germany.,Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Damon Tumes
- Centre for Cancer Biology, SA Pathology and The University of South Australia, Adelaide, South Australia, Australia
| | - Akihiko Murata
- Department of Immunology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Koji Tokoyoda
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Berlin, Germany.,Department of Immunology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| |
Collapse
|
46
|
Johnson BZ, McAlister S, McGuire HM, Palanivelu V, Stevenson A, Richmond P, Palmer DJ, Metcalfe J, Prescott SL, Wood FM, Fazekas de St Groth B, Linden MD, Fear MW, Fear VS. Pediatric Burn Survivors Have Long-Term Immune Dysfunction With Diminished Vaccine Response. Front Immunol 2020; 11:1481. [PMID: 32793203 PMCID: PMC7385079 DOI: 10.3389/fimmu.2020.01481] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/08/2020] [Indexed: 01/04/2023] Open
Abstract
Epidemiological studies have demonstrated that survivors of acute burn trauma are at long-term increased risk of developing a range of morbidities. The mechanisms underlying this increased risk remain unknown. This study aimed to determine whether burn injury leads to sustained immune dysfunction that may underpin long-term morbidity. Plasma and peripheral blood mononuclear cells were collected from 36 pediatric burn survivors >3 years after a non-severe burn injury (<10% total body surface area) and from age/sex-matched non-injured controls. Circulating cytokine and vaccine antibody levels were assessed using multiplex immunoassays and cell profiles compared using a panel of 40 metal-conjugated antibodies and mass cytometry. TNF-α (1.31-fold change from controls), IL-2 (1.18-fold), IL-7 (1.63-fold), and IFN-γ (1.18-fold) were all significantly elevated in the burn cohort. Additionally, burn survivors demonstrated diminished antibody responses to the diphtheria, tetanus, and pertussis vaccine antigens. Comparisons between groups using unsupervised clustering identified differences in proportions of clusters within T-cells, B-cells and myeloid cells. Manual gating confirmed increased memory T-regulatory and central memory CD4+ T-cells, with altered expression of T-cell, B-cell, and dendritic cell markers. Conclusions: This study demonstrates a lasting change to the immune profile of pediatric burn survivors, and highlights the need for further research into post-burn immune suppression and regulation.
Collapse
Affiliation(s)
- Blair Z Johnson
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Sonia McAlister
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Helen M McGuire
- Ramaciotti Facility for Human Systems Biology and the Charles Perkins Centre, Discipline of Pathology, The University of Sydney, Sydney, NSW, Australia
| | | | - Andrew Stevenson
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Peter Richmond
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Debra J Palmer
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Centre for Allergy and Immunology Research, Telethon Kids Institute, Perth, WA, Australia
| | - Jessica Metcalfe
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Centre for Allergy and Immunology Research, Telethon Kids Institute, Perth, WA, Australia
| | - Susan L Prescott
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Centre for Allergy and Immunology Research, Telethon Kids Institute, Perth, WA, Australia
| | - Fiona M Wood
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Department of Health WA, Perth, WA, Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Facility for Human Systems Biology and the Charles Perkins Centre, Discipline of Pathology, The University of Sydney, Sydney, NSW, Australia
| | - Matthew D Linden
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Mark W Fear
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Vanessa S Fear
- Genetic and Rare Diseases, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
47
|
Zens K, Münz C. Tissue resident T cell memory or how the magnificent seven are chilling in the bone. Eur J Immunol 2020; 49:849-852. [PMID: 31017296 DOI: 10.1002/eji.201948208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022]
Abstract
Following infection, tissue-resident memory T cells (Trm) are thought to be left behind at sites of antigen encounter to protect affected tissues against subsequent reinfection. In this issue of the European Journal of Immunology, however, Pascutti et al. demonstrate that both murine and human CD8+ Trm specific to seven different pathogens, including systemic, skin, and lung tissue-localized pathogens, accumulate in the bone marrow (BM). These cells have a CD69+ phenotype, develop independently of local antigen, and require IL-15, Blimp-1, and Hobit for their differentiation and maintenance. Following restimulation, these cells expand and rapidly produce cytokines. While some of these responses may protect the BM from infection, the consideration that some of these pathogens or their antigens might never reach the BM suggests additional functional roles of BM Trm, possibly in supporting hematopoietic functions via cytokine production following infection. It will be further interesting to determine whether BM Trm contribute to the circulating effector pool following reinfection with tissue-localized or systemic pathogens and whether these cells can be elicited by vaccination.
Collapse
Affiliation(s)
- Kyra Zens
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland
| |
Collapse
|
48
|
Riedel R, Addo R, Ferreira-Gomes M, Heinz GA, Heinrich F, Kummer J, Greiff V, Schulz D, Klaeden C, Cornelis R, Menzel U, Kröger S, Stervbo U, Köhler R, Haftmann C, Kühnel S, Lehmann K, Maschmeyer P, McGrath M, Naundorf S, Hahne S, Sercan-Alp Ö, Siracusa F, Stefanowski J, Weber M, Westendorf K, Zimmermann J, Hauser AE, Reddy ST, Durek P, Chang HD, Mashreghi MF, Radbruch A. Discrete populations of isotype-switched memory B lymphocytes are maintained in murine spleen and bone marrow. Nat Commun 2020; 11:2570. [PMID: 32444631 PMCID: PMC7244721 DOI: 10.1038/s41467-020-16464-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/03/2020] [Indexed: 12/15/2022] Open
Abstract
At present, it is not clear how memory B lymphocytes are maintained over time, and whether only as circulating cells or also residing in particular tissues. Here we describe distinct populations of isotype-switched memory B lymphocytes (Bsm) of murine spleen and bone marrow, identified according to individual transcriptional signature and B cell receptor repertoire. A population of marginal zone-like cells is located exclusively in the spleen, while a population of quiescent Bsm is found only in the bone marrow. Three further resident populations, present in spleen and bone marrow, represent transitional and follicular B cells and B1 cells, respectively. A population representing 10-20% of spleen and bone marrow memory B cells is the only one qualifying as circulating. In the bone marrow, all cells individually dock onto VCAM1+ stromal cells and, reminiscent of resident memory T and plasma cells, are void of activation, proliferation and mobility.
Collapse
Affiliation(s)
- René Riedel
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Evolutionary Genomics Group, Max Planck Institute for Evolutionary Biology, 24306, Plön, Germany
| | - Richard Addo
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Marta Ferreira-Gomes
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Gitta Anne Heinz
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Frederik Heinrich
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Jannis Kummer
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Victor Greiff
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH Zürich), CH-4058, Basel, Switzerland
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424, Oslo, Norway
| | - Daniel Schulz
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Cora Klaeden
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Rebecca Cornelis
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Ulrike Menzel
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH Zürich), CH-4058, Basel, Switzerland
| | - Stefan Kröger
- Knowledge Management in Bioinformatics, Humboldt-Universität zu Berlin, 12489, Berlin, Germany
- Department of Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Ulrik Stervbo
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Ralf Köhler
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Claudia Haftmann
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Institute of Experimental Immunology, Universitätsspital Zürich, 8057, Zürich, Switzerland
| | - Silvia Kühnel
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Katrin Lehmann
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Patrick Maschmeyer
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Mairi McGrath
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Sandra Naundorf
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Stefanie Hahne
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Özen Sercan-Alp
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- R&D, TA Immunology & Inflammation Research, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, 65926, Frankfurt am Main, Germany
| | - Francesco Siracusa
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jonathan Stefanowski
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Melanie Weber
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Kerstin Westendorf
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Jakob Zimmermann
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Department for BioMedical Research (DBMR), University of Bern, 3008, Bern, Switzerland
| | - Anja E Hauser
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH Zürich), CH-4058, Basel, Switzerland
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany.
- BCRT/DRFZ Single-Cell Laboratory for Advanced Cellular Therapies - Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany.
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117, Berlin, Germany.
| |
Collapse
|
49
|
Skirecki T, Swacha P, Hoser G, Golab J, Nowis D, Kozłowska E. Bone marrow is the preferred site of memory CD4+ T cell proliferation during recovery from sepsis. JCI Insight 2020; 5:134475. [PMID: 32434988 DOI: 10.1172/jci.insight.134475] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis survivors suffer from increased vulnerability to infections, and lymphopenia presumably contributes to this problem. The mechanisms of the recovery of memory CD4+ T cells after sepsis remain elusive. We used the cecal ligation and puncture mouse model of sepsis to study the restoration of the memory CD4+ T cells during recovery from sepsis. Then, adoptive transfer of antigen-specific naive CD4+ T cells followed by immunization and BrdU labeling were performed to trace the proliferation and migration of memory CD4+ T cells. We revealed that the bone marrow (BM) is the primary site of CD4+ memory T cell homing and proliferation after sepsis-induced lymphopenia. Of interest, BM CD4+ T cells had a higher basal proliferation rate in comparison with splenic T cells. These cells also show features of resident memory T cells yet have the capacity to migrate outside the BM niche and engraft secondary lymphoid organs. The BM niche also sustains viability and functionality of CD4+ T cells. We also identified IL-7 as the major inducer of proliferation of the BM memory CD4+ T cells and showed that recombinant IL-7 improves the recovery of these cells. Taken together, we provide data on the mechanism and location of memory CD4+ T cell proliferation during recovery from septic lymphopenia, which are of relevance in studying immunostimulatory therapies in sepsis.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Patrycja Swacha
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grażyna Hoser
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Warsaw, Poland.,Genomic Medicine, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Kozłowska
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
50
|
Lettau M, Wiedemann A, Schrezenmeier EV, Giesecke-Thiel C, Dörner T. Human CD27+ memory B cells colonize a superficial follicular zone in the palatine tonsils with similarities to the spleen. A multicolor immunofluorescence study of lymphoid tissue. PLoS One 2020; 15:e0229778. [PMID: 32187186 PMCID: PMC7080255 DOI: 10.1371/journal.pone.0229778] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Memory B cell (mBC) induction and maintenance is one of the keys to long-term protective humoral immunity. MBCs are fundamental to successful medical interventions such as vaccinations and therapy in autoimmunity. However, their lifestyle and anatomic residence remain enigmatic in humans. Extrapolation from animal studies serves as a conceptual basis but might be misleading due to major anatomical distinctions between species. METHODS AND FINDINGS Multicolor immunofluorescence stainings on fixed and unfixed frozen tissue sections were established using primary antibodies coupled to haptens and secondary signal amplification. The simultaneous detection of five different fluorescence signals enabled the localization and characterization of human CD27+CD20+Ki67- mBCs for the first time within one section using laser scanning microscopy. As a result, human tonsillar mBCs were initially identified within their complex microenvironment and their relative location to naïve B cells, plasma cells and T cells could be directly studied and compared to the human splenic mBC niche. In all investigated tonsils (n = 15), mBCs appeared to be not only located in a so far subepithelial defined area but were also follicle associated with a previous undescribed gradual decline towards the follicular mantle comparable to human spleen. However, mBC areas around secondary follicles with large germinal centers (GCs) in tonsils showed interruptions and a general widening towards the epithelium while in spleen the mBC-containing marginal zones (MZ) around smaller GCs were relatively broad and symmetrical. Considerably fewer IgM+IgD+/- pre-switch compared to IgA+ or IgG+ post-switch mBCs were detected in tonsils in contrast to spleen. CONCLUSIONS This study extends existing insights into the anatomic residence of human mBCs showing structural similarities of the superficial follicular area in human spleen and tonsil. Our data support the debate of renaming the human splenic MZ to 'superficial zone' in order to be aware of the differences in rodents and, moreover, to consider this term equally for the human palatine tonsil.
Collapse
Affiliation(s)
- Marie Lettau
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Annika Wiedemann
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Eva Vanessa Schrezenmeier
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| | - Claudia Giesecke-Thiel
- Department of Rheumatology and Clinical Immunology, Formerly at the Charité University Medicine Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- * E-mail:
| |
Collapse
|