1
|
Naz F, Hagspiel N, Xu F, Thompson B, Brett Moreau G, Young M, Herbein J, Fox CB, Petri WA, Abhyankar MM. Enhanced immunogenicity of a Clostridioides difficile TcdB vaccine adjuvanted with a synthetic dual-TLR ligand adjuvant. NPJ Vaccines 2025; 10:33. [PMID: 39966390 PMCID: PMC11836405 DOI: 10.1038/s41541-025-01075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/19/2025] [Indexed: 02/20/2025] Open
Abstract
We report a comprehensive evaluation of the toxin B (TcdB) vaccine adjuvanted with a dual Toll-like receptor ligand liposome adjuvant for Clostridioides difficile infection (CDI). The vaccine completely protected mice from a lethal infection. Compared to alum adjuvanted TcdB, it generated functionally superior systemic antibodies and supported strong memory B cell and gut IgA responses. This pharmaceutically acceptable adjuvant platform holds promise for developing a next-generation CDI vaccine.
Collapse
Affiliation(s)
- Farha Naz
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Nicholas Hagspiel
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Feifan Xu
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Brandon Thompson
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - G Brett Moreau
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Mary Young
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | | | | | - William A Petri
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Mayuresh M Abhyankar
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA.
| |
Collapse
|
2
|
Uyar‐Aydin Z, Kadler S, Lauster R, Bartfeld S, Rosowski M. Survival of Human Bone Marrow Plasma Cells In Vitro Depends on the Support of the Stromal Cells, PI3K, and Canonical NF-kappaB Signaling. Eur J Immunol 2025; 55:e202451358. [PMID: 39777683 PMCID: PMC11708448 DOI: 10.1002/eji.202451358] [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: 07/05/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Contrary to short-lived plasma cells, which survive only 3-5 days, long-lived plasma cells (LLPCs) contribute to the humoral memory of the body and thus also to many antibody-related diseases. The ability of plasma cells to persist over months, years, and even a lifetime has been demonstrated in vivo. Yet, the in vitro culture of human primary bone marrow-derived plasma cells has been limited to a few days. Here, we establish culture conditions for human primary bone marrow-derived plasma cells for 21 days. Plasma cells and stromal cells are isolated from human bone marrow and cultured in 2D or a 3D ceramic scaffold. The plasma cells' survival and antibody secretion depend on direct contact with stromal cells. The culture promotes CD19-negative PCs. Inhibition of the PI3K or NF-kappaB pathways using chemical inhibitors reduced the survival of the plasma cells. These results underline the supportive role of the stromal cells for the survival of the LLPC and confirm mechanisms that were identified in mouse LLPCs also for human LLPCs. The culture described here will promote further studies to deepen our understanding of the human LLPC.
Collapse
Affiliation(s)
- Zehra Uyar‐Aydin
- Department Medical BiotechnologyInstitute of BiotechnologyTechnische Universität BerlinBerlinGermany
| | - Shirin Kadler
- Si‐M/Der Simulierte MenschTechnische Universität Berlin and Charité Universitätsmedizin BerlinBerlinGermany
| | - Roland Lauster
- Department Medical BiotechnologyInstitute of BiotechnologyTechnische Universität BerlinBerlinGermany
- Si‐M/Der Simulierte MenschTechnische Universität Berlin and Charité Universitätsmedizin BerlinBerlinGermany
| | - Sina Bartfeld
- Department Medical BiotechnologyInstitute of BiotechnologyTechnische Universität BerlinBerlinGermany
- Si‐M/Der Simulierte MenschTechnische Universität Berlin and Charité Universitätsmedizin BerlinBerlinGermany
| | - Mark Rosowski
- Department Medical BiotechnologyInstitute of BiotechnologyTechnische Universität BerlinBerlinGermany
- Si‐M/Der Simulierte MenschTechnische Universität Berlin and Charité Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
3
|
Naz F, Hagspiel N, Xu F, Thompson B, Moreau GB, Young M, Herbein J, Fox CB, Petri WA, Abhyankar MM. Enhanced immunogenicity of a Clostridioides difficile TcdB vaccine adjuvanted with a synthetic dual-TLR ligand adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625229. [PMID: 39651154 PMCID: PMC11623652 DOI: 10.1101/2024.11.25.625229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
We report a comprehensive evaluation of the toxin B (TcdB) vaccine adjuvanted with a dual Toll-like receptor ligand liposome adjuvant for Clostridioides difficile infection (CDI). The vaccine completely protected mice from a lethal infection. Compared to alum adjuvanted TcdB, it generated functionally superior systemic antibodies and supported strong memory B cell and gut IgA responses. This pharmaceutically acceptable adjuvant platform holds promise for developing a next-generation CDI vaccine.
Collapse
|
4
|
Min EK, Kim SR, Lee CM, Na KH, Park CH, Oh BC, Jung Y, Hong IS. Identification of memory mechanism in tissue-resident stem cells via ANGPTL4 beyond immune cells upon viral antigen exposure. Mol Ther 2024; 32:3042-3058. [PMID: 38582960 PMCID: PMC11403228 DOI: 10.1016/j.ymthe.2024.04.006] [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: 01/11/2024] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Although memory functions of immune cells characterized by increased resistance to subsequent infections after initial pathogen exposure are well-established, it remains unclear whether non-immune cells, especially tissue-resident stem cells, exhibit similar memory mechanisms. The present study revealed that detrimental effects of initial viral antigen exposure (human papillomavirus [HPV]) on diverse stem cell functions were significantly exacerbated upon subsequent secondary exposure both in vitro and in vivo. Importantly, endometrial stem cells exhibited robust memory functions following consecutive HPV antigen exposures, whereas fully differentiated cells such as fibroblasts and vesicular cells did not show corresponding changes in response to the same antigen exposures. Deficiency of angiopoietin-like 4 (ANGPTL4) achieved through small hairpin RNA knockdown in vitro and knockout (KO) mice in vivo highlighted the critical role of ANGPTL4 in governing memory functions associated with various stem cell processes. This regulation occurred through histone H3 methylation alterations and PI3K/Akt signaling pathways in response to successive HPV antigen exposures. Furthermore, memory functions associated with various stem cell functions that were evident in wild-type mice following consecutive exposures to HPV antigen were not observed in ANGPTL4 KO mice. In summary, our findings strongly support the presence of memory mechanism in non-immune cells, particularly tissue-resident stem cells.
Collapse
Affiliation(s)
- Eun-Kyung Min
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Soo-Rim Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Choon-Mi Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Kun-Hee Na
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Chan Hum Park
- Department of Otolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Byung-Chul Oh
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - YunJae Jung
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Korea.
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea.
| |
Collapse
|
5
|
Park S, Min E, Kim S, Kim S, Na K, Park CH, Jung Y, Oh B, Hong I. Exploring Memory Function Beyond Immune Cells: ANGPTL4-Mediated Memory Functions in Tissue Resident Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307545. [PMID: 38666393 PMCID: PMC11267307 DOI: 10.1002/advs.202307545] [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: 10/10/2023] [Revised: 04/15/2024] [Indexed: 07/25/2024]
Abstract
Adapted immune cells are known to develop memory functions that increase resistance to subsequent infections after initial pathogen exposure, however, it is unclear whether non-immune cells, like tissue-resident stem cells, have similar memory functions. Here, it is found that tissue-resident stem cells crucial for tissue regeneration show diminished adverse effects on diverse stem cell functions against successive exposure to foreign antigen (β-glucan) to maintain tissue homeostasis and stability both in vitro and in vivo. These data suggest that endometrial stem cells may possess a robust memory function, in contrast, fully differentiated cells like fibroblasts and vesicular cells do not show these memory mechanisms upon consecutive antigen exposure. Moreover, the pivotal role of Angiopoietin-like 4 (ANGPTL4) in regulating the memory functions of endometrial stem cells is identified through specific shRNA knockdown in vitro and knockout mice in vivo experiments. ANGPTL4 is associated with the alteration of diverse stem cell functions and epigenetic modifications, notably through histone H3 methylation changes and two pathways (i.e., PI3K/Akt and FAK/ERK1/2 signaling) upon consecutive antigen exposure. These findings imply the existence of inherent self-defense mechanisms through which local stem cells can adapt and protect themselves from recurrent antigenic challenges, ultimately mitigating adverse consequences.
Collapse
Affiliation(s)
- Se‐Ra Park
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheon406–840Republic of Korea
| | - Eun‐kyung Min
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheon406–840Republic of Korea
| | - Soo‐Rim Kim
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheon406–840Republic of Korea
| | - Suk‐Kyung Kim
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheon406–840Republic of Korea
| | - Kun‐Hee Na
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Microbiology, College of MedicineGachon UniversityIncheon21999Republic of Korea
| | - Chan Hum Park
- Department of Otolaryngology‐Head and Neck Surgery, Chuncheon Sacred Heart HospitalHallym University College of MedicineChuncheon24201Republic of Korea
| | - YunJae Jung
- Department of Microbiology, College of MedicineGachon UniversityIncheon21999Republic of Korea
| | - Byung‐Chul Oh
- Department of Physiology, Lee Gil Ya Cancer and Diabetes InstituteGachon University College of MedicineIncheon21999Republic of Korea
| | - In‐Sun Hong
- Department of Health Sciences and Technology, GAIHSTGachon UniversityIncheon21999Republic of Korea
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheon406–840Republic of Korea
| |
Collapse
|
6
|
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
|
7
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
8
|
Ben Hamza A, Welters C, Stadler S, Brüggemann M, Dietze K, Brauns O, Brümmendorf TH, Winkler T, Bullinger L, Blankenstein T, Rosenberger L, Leisegang M, Kammertöns T, Herr W, Moosmann A, Strobel J, Hackstein H, Dornmair K, Beier F, Hansmann L. Virus-reactive T cells expanded in aplastic anemia eliminate hematopoietic progenitor cells by molecular mimicry. Blood 2024; 143:1365-1378. [PMID: 38277625 DOI: 10.1182/blood.2023023142] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
ABSTRACT Acquired aplastic anemia is a bone marrow failure syndrome characterized by hypocellular bone marrow and peripheral blood pancytopenia. Frequent clinical responses to calcineurin inhibition and antithymocyte globulin strongly suggest critical roles for hematopoietic stem/progenitor cell-reactive T-cell clones in disease pathophysiology; however, their exact contribution and antigen specificities remain unclear. We determined differentiation states and targets of dominant T-cell clones along with their potential to eliminate hematopoietic progenitor cells in the bone marrow of 15 patients with acquired aplastic anemia. Single-cell sequencing and immunophenotyping revealed oligoclonal expansion and effector differentiation of CD8+ T-cell compartments. We reexpressed 28 dominant T-cell receptors (TCRs) of 9 patients in reporter cell lines to determine reactivity with (1) in vitro-expanded CD34+ bone marrow, (2) CD34- bone marrow, or (3) peptide pools covering immunodominant epitopes of highly prevalent viruses. Besides 5 cytomegalovirus-reactive TCRs, we identified 3 TCRs that recognized antigen presented on hematopoietic progenitor cells. T cells transduced with these TCRs eliminated hematopoietic progenitor cells of the respective patients in vitro. One progenitor cell-reactive TCR (11A5) also recognized an epitope of the Epstein-Barr virus-derived latent membrane protein 1 (LMP1) presented on HLA-A∗02:01. We identified 2 LMP1-related mimotopes within the human proteome as activating targets of TCR 11A5, providing proof of concept that molecular mimicry of viral and self-epitopes can drive T cell-mediated elimination of hematopoietic progenitor cells in aplastic anemia.
Collapse
Affiliation(s)
- Amin Ben Hamza
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carlotta Welters
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Serena Stadler
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
| | - Monika Brüggemann
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig Holstein, Kiel, Germany
| | - Kerstin Dietze
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Brauns
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | - Thomas Winkler
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Leonie Rosenberger
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Leisegang
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL
| | - Thomas Kammertöns
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Moosmann
- Department of Medicine III, Klinikum der Universität München, Munich, Germany
- German Center for Infection Research, Munich, Germany
- Helmholtz Munich, Munich, Germany
| | - Julian Strobel
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | - Leo Hansmann
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
9
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
10
|
Khan S, Chakraborty M, Wu F, Chen N, Wang T, Chan YT, Sayad A, Vásquez JDS, Kotlyar M, Nguyen K, Huang Y, Alibhai FJ, Woo M, Li RK, Husain M, Jurisica I, Gehring AJ, Ohashi PS, Furman D, Tsai S, Winer S, Winer DA. B Cells Promote T Cell Immunosenescence and Mammalian Aging Parameters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.556363. [PMID: 38529494 PMCID: PMC10962733 DOI: 10.1101/2023.09.12.556363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
A dysregulated adaptive immune system is a key feature of aging, and is associated with age-related chronic diseases and mortality. Most notably, aging is linked to a loss in the diversity of the T cell repertoire and expansion of activated inflammatory age-related T cell subsets, though the main drivers of these processes are largely unknown. Here, we find that T cell aging is directly influenced by B cells. Using multiple models of B cell manipulation and single-cell omics, we find B cells to be a major cell type that is largely responsible for the age-related reduction of naive T cells, their associated differentiation towards pathogenic immunosenescent T cell subsets, and for the clonal restriction of their T cell receptor (TCR). Accordingly, we find that these pathogenic shifts can be therapeutically targeted via CD20 monoclonal antibody treatment. Mechanistically, we uncover a new role for insulin receptor signaling in influencing age-related B cell pathogenicity that in turn induces T cell dysfunction and a decline in healthspan parameters. These results establish B cells as a pivotal force contributing to age-associated adaptive immune dysfunction and healthspan outcomes, and suggest new modalities to manage aging and related multi-morbidity.
Collapse
Affiliation(s)
- Saad Khan
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Mainak Chakraborty
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Fei Wu
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Nan Chen
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Tao Wang
- Department of Physiology, University of Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Ted Rogers Centre for Heart Research, Toronto, ON, M5G 1X8, Canada
| | - Yi Tao Chan
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Azin Sayad
- Princess Margaret Cancer Centre, University Health Network, ON M5G 2C1, Canada
| | - Juan Diego Sánchez Vásquez
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
| | - Max Kotlyar
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
| | - Khiem Nguyen
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Yingxiang Huang
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Faisal J. Alibhai
- Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
| | - Minna Woo
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, University of Toronto, ON M5G 1L7, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Division of Cardiac Surgery, University Health Network, University of Toronto, ON M5G IL7, Canada
| | - Mansoor Husain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Ted Rogers Centre for Heart Research, Toronto, ON, M5G 1X8, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
- Departments of Medical Biophysics and Computer Science, and Faculty of Dentistry, University of Toronto, ON M5S 2E4, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Adam J. Gehring
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto Center for Liver Disease & Schwartz Reisman Liver Research Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, ON M5G 2C1, Canada
| | - David Furman
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2RS, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Daniel A. Winer
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| |
Collapse
|
11
|
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
|
12
|
van de Ven K, Lanfermeijer J, van Dijken H, Muramatsu H, Vilas Boas de Melo C, Lenz S, Peters F, Beattie MB, Lin PJC, Ferreira JA, van den Brand J, van Baarle D, Pardi N, de Jonge J. A universal influenza mRNA vaccine candidate boosts T cell responses and reduces zoonotic influenza virus disease in ferrets. SCIENCE ADVANCES 2022; 8:eadc9937. [PMID: 36516261 PMCID: PMC9750153 DOI: 10.1126/sciadv.adc9937] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/04/2022] [Indexed: 06/17/2023]
Abstract
Universal influenza vaccines should protect against continuously evolving and newly emerging influenza viruses. T cells may be an essential target of such vaccines, as they can clear infected cells through recognition of conserved influenza virus epitopes. We evaluated a novel T cell-inducing nucleoside-modified messenger RNA (mRNA) vaccine that encodes the conserved nucleoprotein, matrix protein 1, and polymerase basic protein 1 of an H1N1 influenza virus. To mimic the human situation, we applied the mRNA vaccine as a prime-boost regimen in naïve ferrets (mimicking young children) and as a booster in influenza-experienced ferrets (mimicking adults). The vaccine induced and boosted broadly reactive T cells in the circulation, bone marrow, and respiratory tract. Booster vaccination enhanced protection against heterosubtypic infection with a potential pandemic H7N9 influenza virus in influenza-experienced ferrets. Our findings show that mRNA vaccines encoding internal influenza virus proteins represent a promising strategy to induce broadly protective T cell immunity against influenza viruses.
Collapse
Affiliation(s)
- Koen van de Ven
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josien Lanfermeijer
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Harry van Dijken
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Vilas Boas de Melo
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Stefanie Lenz
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Florence Peters
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | | | | | - José A. Ferreira
- Department of Statistics, Informatics and Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Judith van den Brand
- Division of Pathology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Debbie van Baarle
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology Research Group, University Medical Center Groningen, Groningen, Netherlands
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jørgen de Jonge
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
13
|
Welters C, Lammoglia Cobo MF, Stein CA, Hsu MT, Ben Hamza A, Penter L, Chen X, Buccitelli C, Popp O, Mertins P, Dietze K, Bullinger L, Moosmann A, Blanc E, Beule D, Gerbitz A, Strobel J, Hackstein H, Rahn HP, Dornmair K, Blankenstein T, Hansmann L. Immune Phenotypes and Target Antigens of Clonally Expanded Bone Marrow T Cells in Treatment-Naïve Multiple Myeloma. Cancer Immunol Res 2022; 10:1407-1419. [PMID: 36122410 PMCID: PMC9627264 DOI: 10.1158/2326-6066.cir-22-0434] [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: 06/01/2022] [Revised: 07/23/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Multiple myeloma is a hematologic malignancy of monoclonal plasma cells that accumulate in the bone marrow. Despite their clinical and pathophysiologic relevance, the roles of bone marrow-infiltrating T cells in treatment-naïve patients are incompletely understood. We investigated whether clonally expanded T cells (i) were detectable in multiple myeloma bone marrow, (ii) showed characteristic immune phenotypes, and (iii) whether dominant clones recognized antigens selectively presented on multiple myeloma cells. Single-cell index sorting and T-cell receptor (TCR) αβ sequencing of bone marrow T cells from 13 treatment-naïve patients showed dominant clonal expansion within CD8+ cytolytic effector compartments, and only a minority of expanded T-cell clones expressed the classic immune-checkpoint molecules PD-1, CTLA-4, or TIM-3. To identify their molecular targets, TCRs of 68 dominant bone marrow clones from five selected patients were reexpressed and incubated with multiple myeloma and non-multiple myeloma cells from corresponding patients. Only 1 of 68 TCRs recognized antigen presented on multiple myeloma cells. This TCR was HLA-C-restricted, self-peptide-specific and could be activated by multiple myeloma cells of multiple patients. The remaining dominant T-cell clones did not recognize multiple myeloma cells and were, in part, specific for antigens associated with chronic viral infections. In conclusion, we showed that dominant bone marrow T-cell clones in treatment-naïve patients rarely recognize antigens presented on multiple myeloma cells and exhibit low expression of classic immune-checkpoint molecules. Our data provide experimental context for experiences from clinical immune-checkpoint inhibition trials and will inform future T cell-dependent therapeutic strategies.
Collapse
Affiliation(s)
- Carlotta Welters
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - María Fernanda Lammoglia Cobo
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Alexander Stein
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Meng-Tung Hsu
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Amin Ben Hamza
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaojing Chen
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Christopher Buccitelli
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Kerstin Dietze
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Moosmann
- Department of Medicine III, Klinikum der Universität München, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Armin Gerbitz
- Hans Messner Allogeneic Stem Cell Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Julian Strobel
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Hans-Peter Rahn
- Preparative Flow Cytometry, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Leo Hansmann
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Corresponding Author: Leo Hansmann, Charité–Universitätsmedizin Berlin (CVK), Department of Hematology, Oncology, and Tumor Immunology, Augustenburger Platz 1, 13353 Berlin, Germany. Phone: 49-(0)30-450-665238; Fax: 49-(0)30-450-553914; E-mail:
| |
Collapse
|
14
|
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
|
15
|
Swain AC, Borghans JA, de Boer RJ. Effect of cellular aging on memory T-cell homeostasis. Front Immunol 2022; 13:947242. [PMID: 36059495 PMCID: PMC9429809 DOI: 10.3389/fimmu.2022.947242] [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: 05/18/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
The fact that T-cell numbers remain relatively stable throughout life, and that T-cell proliferation rates increase during lymphopenia, has led to the consensus that T-cell numbers are regulated in a density-dependent manner. Competition for resources among memory T cells has been proposed to underlie this ‘homeostatic’ regulation. We first review how two classic models of resource competition affect the T-cell receptor (TCR) diversity of the memory T-cell pool. First, ‘global’ competition for cytokines leads to a skewed repertoire that tends to be dominated by the very first immune response. Second, additional ‘cognate’ competition for specific antigens results in a very diverse and stable memory T-cell pool, allowing every antigen to be remembered, which we therefore define as the ‘gold-standard’. Because there is limited evidence that memory T cells of the same specificity compete more strongly with each other than with memory T cells of different specificities, i.e., for ‘cognate’ competition, we investigate whether cellular aging could account for a similar level of TCR diversity. We define cellular aging as a declining cellular fitness due to reduced proliferation. We find that the gradual erosion of previous T-cell memories due to cellular aging allows for better establishment of novel memories and for a much higher level of TCR diversity compared to global competition. A small continual source (either from stem-cell-like memory T-cells or from naive T-cells due to repeated antigen exposure) improves the diversity of the memory T-cell pool, but remarkably, only in the cellular aging model. We further show that the presence of a source keeps the inflation of chronic memory responses in check by maintaining the immune memories to non-chronic antigens. We conclude that cellular aging along with a small source provides a novel and immunologically realistic mechanism to achieve and maintain the ‘gold-standard’ level of TCR diversity in the memory T-cell pool.
Collapse
Affiliation(s)
- Arpit C. Swain
- Theoretical Biology, Utrecht University, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: Arpit C. Swain,
| | - José A.M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rob J. de Boer
- Theoretical Biology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
16
|
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
|
17
|
Tomasin R, Bruni-Cardoso A. The role of cellular quiescence in cancer - beyond a quiet passenger. J Cell Sci 2022; 135:276213. [PMID: 35929545 DOI: 10.1242/jcs.259676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Quiescence, the ability to temporarily halt proliferation, is a conserved process that initially allowed survival of unicellular organisms during inhospitable times and later contributed to the rise of multicellular organisms, becoming key for cell differentiation, size control and tissue homeostasis. In this Review, we explore the concept of cancer as a disease that involves abnormal regulation of cellular quiescence at every step, from malignant transformation to metastatic outgrowth. Indeed, disrupted quiescence regulation can be linked to each of the so-called 'hallmarks of cancer'. As we argue here, quiescence induction contributes to immune evasion and resistance against cell death. In contrast, loss of quiescence underlies sustained proliferative signalling, evasion of growth suppressors, pro-tumorigenic inflammation, angiogenesis and genomic instability. Finally, both acquisition and loss of quiescence are involved in replicative immortality, metastasis and deregulated cellular energetics. We believe that a viewpoint that considers quiescence abnormalities that occur during oncogenesis might change the way we ask fundamental questions and the experimental approaches we take, potentially contributing to novel discoveries that might help to alter the course of cancer therapy.
Collapse
Affiliation(s)
- Rebeka Tomasin
- e-signal Lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Ave Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Alexandre Bruni-Cardoso
- e-signal Lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Ave Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
18
|
Infantino M, Manfredi M, Stacchini L, Cosma C, Grossi V, Lari B, Russo E, Amedei A, Benucci M, Veneziani F, Casprini P, Catalano CM, Cirrincione G, Bonaccorsi G, Pompetti A. The role of neutralizing antibodies by sVNT after two doses of BNT162b2 mRNA vaccine in a cohort of Italian healthcare workers. Clin Chem Lab Med 2022; 60:934-940. [PMID: 35303766 DOI: 10.1515/cclm-2022-0170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Evaluating anti-SARS-CoV-2 antibody levels is a current priority to drive immunization, as well as to predict when a vaccine booster dose may be required and for which priority groups. The aim of our study was to investigate the kinetics of anti-SARS-CoV-2 Spike S1 protein IgG (anti-S1 IgG) antibodies and neutralizing antibodies (NAbs) in an Italian cohort of healthcare workers (HCWs), following the Pfizer/BNT162b2 mRNA vaccine, over a period of up to six months after the second dose. METHODS We enrolled 57 HCWs, without clinical history of COVID-19 infection. Fluoroenzyme-immunoassay was used for the quantitative anti-S1 IgG antibodies at different time points T1 (one month), T3 (three months) and T6 (six months) following the second vaccine shot. Simultaneously, a commercial surrogate virus neutralization test (sVNT) was used for the determination of NAbs, expressed as inhibition percentage (% IH). RESULTS Median values of anti-S1 IgG antibodies decreased from T1 (1,452 BAU/mL) to T6 (104 BAU/mL) with a percent variation of 92.8% while the sVNT showed a percent variation of 34.3% for the same time frame. The decline in anti-S1 IgG antibodies from T1 to T6 was not accompanied by a loss of the neutralizing capacity of antibodies. In fact at T6 a neutralization percentage <20% IH was observed only in 3.51% of HCWs. CONCLUSIONS Our findings reveal that the decrease of anti-S1 IgG levels do not correspond in parallel to a decrease of NAbs over time, which highlights the necessity of using both assays to assess vaccination effectiveness.
Collapse
Affiliation(s)
- Maria Infantino
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Mariangela Manfredi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Lorenzo Stacchini
- Department of Health Science, University of Florence, Florence, Italy
| | - Claudia Cosma
- Department of Health Science, University of Florence, Florence, Italy
| | - Valentina Grossi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Barbara Lari
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Francesca Veneziani
- Clinical Pathology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Patrizia Casprini
- Clinical Pathology Laboratory Unit, S. Giovanni di Dio Hospital, Florence, Italy
| | - Cateno Mario Catalano
- Department of Technical Health Services, Preventive Medicine, S. Giovanni di Dio Hospital, Florence, Italy
| | - Giuseppe Cirrincione
- Department of Technical Health Services, Preventive Medicine, S. Giovanni di Dio Hospital, Florence, Italy
| | | | - Adolfo Pompetti
- SOC Clinical Assistance Governance, SOS Preventive Medicine Unit, S. Giovanni di Dio Hospital, Florence, Italy
| |
Collapse
|
19
|
Abstract
Purpose of review Immunological memory is an important evolutionary adaptation of the immune system. Previously restricted to the adaptive immune system, the concept of memory has recently been broadened to the innate immune system. This review summarizes recent studies that highlight the contribution of the hematopoietic stem cells (HSCs) in supporting immunological memory. Recent findings Short-lived innate immune cells can build a long-lasting memory of infection to improve their response to secondary challenges. Studies show that these unexpected properties of the innate immune system are sustained by epigenetic and metabolic changes in the HSC compartment. Summary HSCs are durably altered in response to pathogens and serve as long-term support for innate immune memory. Many questions remain regarding the mechanisms contributing to the induction and the maintenance of this immune memory in HSCs. Answering these questions will open new perspectives to understand how environmental factors shape the HSC activity over time.
Collapse
|
20
|
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: 18] [Impact Index Per Article: 4.5] [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
|
21
|
|