1
|
Duque-Wilckens N, Joseph D, Syed M, Smith B, Maradiaga N, Yeh SY, Srinivasan V, Sotomayor F, Durga K, Nestler E, Moesers AJ, Robison AJ. FosB/ΔFosB activation in mast cells regulates gene expression to modulate allergic inflammation in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.06.592755. [PMID: 38766119 PMCID: PMC11100602 DOI: 10.1101/2024.05.06.592755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Mast cells are innate immune cells that regulate physiological processes by releasing pre-stored and newly synthesized mediators in response to allergens, infection, and other stimuli. Dysregulated mast cell activity can lead to multisystemic pathologies, but the underlying regulatory mechanisms remain poorly understood. We found that FOSB and ΔFOSB, transcription factors encoded by the FosB gene, are robustly expressed in mast cells following IgE-antigen stimulation, suggesting a role in modulating stimulus-induced mast cell functions. Using phenotypic, gene binding, and gene expression analyses in wild-type and mast cell-specific FosB knockout male mice, we demonstrate that FOSB/ΔFOSB modulates mast cell functions by limiting reactivity to allergen-like stimuli both in vitro and in vivo . These effects seem to be mediated, at least in part, by FOSB/ΔFOSB-driven enhanced expression of DUSP4, a dual-specificity phosphatase that attenuates MAPK signaling. These findings highlight FOSB/ΔFOSB as critical regulators of mast cell activity and potential targets for therapeutic intervention.
Collapse
|
2
|
Pursell T, Reers A, Mikelov A, Kotagiri P, Ellison JA, Hutson CL, Boyd SD, Frank HK. Genetically and Functionally Distinct Immunoglobulin Heavy Chain Locus Duplication in Bats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.606892. [PMID: 39211187 PMCID: PMC11360916 DOI: 10.1101/2024.08.09.606892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The genetic locus encoding immunoglobulin heavy chains (IgH) is critical for vertebrate humoral immune responses and diverse antibody repertoires. Immunoglobulin and T cell receptor loci of most bat species have not been annotated, despite the recurrent role of bats as viral reservoirs and sources of zoonotic pathogens. We investigated the genetic structure and function of IgH loci across the largest bat family, Vespertilionidae, focusing on big brown bats (Eptesicus fuscus ). We discovered that E. fuscus and ten other species within Vespertilionidae have two complete, functional, and distinct immunoglobulin heavy chain loci on separate chromosomes. This locus organization is previously unknown in mammals, but is reminiscent of more limited duplicated loci in teleost fish. Single cell transcriptomic data validate functional rearrangement and expression of immunoglobulin heavy chains of both loci in the expressed repertoire of Eptesicus fuscus , with maintenance of allelic exclusion, bias of usage toward the smaller and more compact IgH locus, and evidence of differential selection of antigen-experienced B cells and plasma cells varying by IgH locus use. This represents a unique mechanism for mammalian humoral immunity and may contribute to bat resistance to viral pathogenesis.
Collapse
|
3
|
Liu J, Zhang H. Zinc Finger and BTB Domain-Containing 20: A Newly Emerging Player in Pathogenesis and Development of Human Cancers. Biomolecules 2024; 14:192. [PMID: 38397429 PMCID: PMC10887282 DOI: 10.3390/biom14020192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Zinc finger and BTB domain-containing 20 (ZBTB20), which was initially identified in human dendritic cells, belongs to a family of transcription factors (TFs) with an N-terminal BTB domain and one or more C-terminal DNA-binding zinc finger domains. Under physiological conditions, ZBTB20 acts as a transcriptional repressor in cellular development and differentiation, metabolism, and innate immunity. Interestingly, multiple lines of evidence from mice and human systems have revealed the importance of ZBTB20 in the pathogenesis and development of cancers. ZBTB20 is not only a hotspot of genetic variation or fusion in many types of human cancers, but also a key TF or intermediator involving in the dysregulation of cancer cells. Given the diverse functions of ZBTB20 in both health and disease, we herein summarize the structure and physiological roles of ZBTB20, with an emphasis on the latest findings on tumorigenesis and cancer progression.
Collapse
Affiliation(s)
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China;
| |
Collapse
|
4
|
Lende SSF, Barnkob NM, Hansen RW, Bansia H, Vestergaard M, Rothemejer FH, Worsaae A, Brown D, Pedersen ML, Rahimic AHF, Juhl AK, Gjetting T, Østergaard L, Georges AD, Vuillard LM, Schleimann MH, Koefoed K, Tolstrup M. Discovery of neutralizing SARS-CoV-2 antibodies enriched in a unique antigen specific B cell cluster. PLoS One 2023; 18:e0291131. [PMID: 37729215 PMCID: PMC10511142 DOI: 10.1371/journal.pone.0291131] [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: 05/03/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Despite development of effective SARS-CoV-2 vaccines, a sub-group of vaccine non-responders depends on therapeutic antibodies or small-molecule drugs in cases of severe disease. However, perpetual viral evolution has required continuous efficacy monitoring as well as exploration of new therapeutic antibodies, to circumvent resistance mutations arising in the viral population. We performed SARS-CoV-2-specific B cell sorting and subsequent single-cell sequencing on material from 15 SARS-CoV-2 convalescent participants. Through screening of 455 monoclonal antibodies for SARS-CoV-2 variant binding and virus neutralization, we identified a cluster of activated B cells highly enriched for SARS-CoV-2 neutralizing antibodies. Epitope binning and Cryo-EM structure analysis identified the majority of neutralizing antibodies having epitopes overlapping with the ACE2 receptor binding motif (class 1 binders). Extensive functional antibody characterization identified two potent neutralizing antibodies, one retaining SARS-CoV-1 neutralizing capability, while both bind major common variants of concern and display prophylactic efficacy in vivo. The transcriptomic signature of activated B cells harboring broadly binding neutralizing antibodies with therapeutic potential identified here, may be a guide in future efforts of rapid therapeutic antibody discovery.
Collapse
Affiliation(s)
- Stine Sofie Frank Lende
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Harsh Bansia
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, United States of America
| | | | - Frederik Holm Rothemejer
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Deijona Brown
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, United States of America
| | - Maria Lange Pedersen
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Anna Karina Juhl
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
| | - Torben Gjetting
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, United States of America
- Antibody Technology, Novo Nordisk A/S, Måløv, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Amédée Des Georges
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, United States of America
- Department of Chemistry and Biochemistry, City College of New York, New York, NY, United States of America
- PhD Programs in Biochemistry, and Chemistry, Graduate Center, City University of New York, New York, NY, United States of America
| | | | | | | | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Skejby, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Aranburu A, Engström E, Gerasimcik N, Alsén S, Camponeschi A, Yrlid U, Grimsholm O, Mårtensson IL. Clonal relationships of memory B cell subsets in autoimmune mice. Front Immunol 2023; 14:1129234. [PMID: 36936947 PMCID: PMC10015592 DOI: 10.3389/fimmu.2023.1129234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023] Open
Abstract
Immunological memory protects our body from re-infection and it is composed of a cellular and a humoral arm. The B-cell branch with its memory B cells (MBCs), plasma cells and antibodies, formed either in a germinal centre (GC) -dependent or -independent manner, ensure that we can rapidly mount a recall immune response. Previous work in immunised wildtype (WT) mice have identified several subsets of MBCs whereas less is known under autoimmune conditions. Here, we have investigated the heterogeneity of the MBC compartment in autoimmune mouse models and examined the clonal relationships between MBC subsets and GC B cells in one of the models. We demonstrate the presence of at least four different MBC subsets based on their differential expression pattern of CD73, CD80 and PD-L2 in surrogate light chain-deficient (SLC-/-), MRL+/+ and MRLlpr/lpr mice, where most of the MBCs express IgM. Likewise, four MBC subsets could be identified in WT immunised mice. In SLC-/- mice, high-throughput sequencing of Ig heavy chains demonstrates that the two CD73-positive subsets are generally more mutated. Lineage tree analyses on expanded clones show overlaps between all MBC subsets and GC B cells primarily in the IgM sequences. Moreover, each of the three IgM MBC subsets could be found both as ancestor and progeny to GC B cells. This was also observed in the IgG sequences except for the CD73-negative subset. Thus, our findings demonstrate that several MBC subsets are present in autoimmune and WT mice. In SLC-/- mice, these MBC subsets are clonally related to each other and to GC B cells. Our results also indicate that different MBC subsets can seed the GC reaction.
Collapse
Affiliation(s)
- Alaitz Aranburu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Engström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Natalija Gerasimcik
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Samuel Alsén
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ola Grimsholm
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Inga-Lill Mårtensson,
| |
Collapse
|
6
|
Lyu M, Shi X, Liu Y, Zhao H, Yuan Y, Xie R, Gu Y, Dong Y, Wang M. Single-Cell Transcriptome Analysis of H5N1-HA-Stimulated Alpaca PBMCs. Biomolecules 2022; 13:biom13010060. [PMID: 36671445 PMCID: PMC9855979 DOI: 10.3390/biom13010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
Avian influenza A virus H5N1 is a highly pathogenic and persistently a major threat to global health. Vaccines and antibodies targeting hemagglutinin (HA) protein are the primary management strategies for the epidemic virus. Although camelids possess unique immunological features, the immune response induced by specific antigens has not yet been thoroughly investigated. Herein, we immunized an alpaca with the HA antigen of the H5N1 virus and performed single-cell transcriptome profiling for analysis of longitudinal peripheral blood mononuclear cell (PBMCs) behavior using single-cell sequencing technology (scRNA-seq). We revealed multiple cellular immunities during the immunization. The monocytes continued to expand after immunization, while the plasma cells reached their peak three days after the second antigen stimulation. Both monocytes and B cells were stimulated by the HA antigen and produced cell-type-specific cytokines to participated in the immune response. To our knowledge, this is the first study to examine the HA-specific immunological dynamics of alpaca PBMCs at the single-cell level, which is beneficial for understanding the anti-viral immune system and facilitating the development of more potent vaccines and antibodies in camelid animals.
Collapse
Affiliation(s)
- Menghua Lyu
- BGI-Shenzhen, Shenzhen 518083, China
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | | | - Yang Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Yue Yuan
- BGI-Shenzhen, Shenzhen 518083, China
| | - Run Xie
- BGI-Shenzhen, Shenzhen 518083, China
| | - Ying Gu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | |
Collapse
|
7
|
Jiang X, Zhang X, Jiang N, Sun Y, Li T, Zhang J, Shen Y, Cao J. The single-cell landscape of cystic echinococcosis in different stages provided insights into endothelial and immune cell heterogeneity. Front Immunol 2022; 13:1067338. [PMID: 36569953 PMCID: PMC9772464 DOI: 10.3389/fimmu.2022.1067338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Hydatid cysts and angiogenesis are the key characteristics of cystic echinococcosis, with immune cells and endothelial cells mediating essential roles in disease progression. Recent single-cell analysis studies demonstrated immune cell infiltration after Echinococcus granulosus infection, highlighting the diagnostic and therapeutic potential of targeting certain cell types in the lesion microenvironment. However, more detailed immune mechanisms during different periods of E. granulosus infection were not elucidated. Methods Herein, we characterized immune and endothelial cells from the liver samples of mice in different stages by single-cell RNA sequencing. Results We profiled the transcriptomes of 45,199 cells from the liver samples of mice at 1, 3, and 6 months after infection (two replicates) and uninfected wild-type mice. The cells were categorized into 26 clusters with four distinct cell types: natural killer (NK)/T cells, B cells, myeloid cells, and endothelial cells. An SPP1+ macrophage subset with immunosuppressive and pro-angiogenic functions was identified in the late infection stage. Single-cell regulatory network inference and clustering (SCENIC) analysis suggested that Cebpe, Runx3, and Rora were the key regulators of the SPP1+ macrophages. Cell communication analysis revealed that the SPP1+ macrophages interacted with endothelial cells and had pro-angiogenic functions. There was an obvious communicative relationship between SPP1+ macrophages and endothelial cells via Vegfa-Vegfr1/Vegfr2, and SPP1+ macrophages interacted with other immune cells via specific ligand-receptor pairs, which might have contributed to their immunosuppressive function. Discussion Our comprehensive exploration of the cystic echinococcosis ecosystem and the first discovery of SPP1+ macrophages with infection period specificity provide deeper insights into angiogenesis and the immune evasion mechanisms associated with later stages of infection.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Xiaofan Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,Department of Laboratory Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nan Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Yeting Sun
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Teng Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Jing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,*Correspondence: Yujuan Shen, ; Jianping Cao,
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China,School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Yujuan Shen, ; Jianping Cao,
| |
Collapse
|
8
|
Bhattacharya D. Instructing durable humoral immunity for COVID-19 and other vaccinable diseases. Immunity 2022; 55:945-964. [PMID: 35637104 PMCID: PMC9085459 DOI: 10.1016/j.immuni.2022.05.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Many aspects of SARS-CoV-2 have fully conformed with the principles established by decades of viral immunology research, ultimately leading to the crowning achievement of highly effective COVID-19 vaccines. Nonetheless, the pandemic has also exposed areas where our fundamental knowledge is thinner. Some key unknowns are the duration of humoral immunity post-primary infection or vaccination and how long booster shots confer protection. As a corollary, if protection does not last as long as desired, what are some ways it can be improved? Here, I discuss lessons from other infections and vaccines that point to several key features that influence durable antibody production and the perseverance of immunity. These include (1) the specific innate sensors that are initially triggered, (2) the kinetics of antigen delivery and persistence, (3) the starting B cell receptor (BCR) avidity and antigen valency, and (4) the memory B cell subsets that are recalled by boosters. I further highlight the fundamental B cell-intrinsic and B cell-extrinsic pathways that, if understood better, would provide a rational framework for vaccines to reliably provide durable immunity.
Collapse
Affiliation(s)
- Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
9
|
Kodali S, Li M, Budai MM, Chen M, Wang J. Protection of Quiescence and Longevity of IgG Memory B Cells by Mitochondrial Autophagy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1085-1098. [PMID: 35101890 PMCID: PMC8887795 DOI: 10.4049/jimmunol.2100969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022]
Abstract
The development of long-lived immune memory cells against pathogens is critical for the success of vaccines to establish protection against future infections. However, the mechanisms governing the long-term survival of immune memory cells remain to be elucidated. In this article, we show that the maintenance mitochondrial homeostasis by autophagy is critical for restricting metabolic functions to protect IgG memory B cell survival. Knockout of mitochondrial autophagy genes, Nix and Bnip3, leads to mitochondrial accumulation and increases in oxidative phosphorylation and fatty acid synthesis, resulting in the loss of IgG+ memory B cells in mice. Inhibiting fatty acid synthesis or silencing necroptosis gene Ripk3 rescued Nix-/-Bnip3-/- IgG memory B cells, indicating that mitochondrial autophagy is important for limiting metabolic functions to prevent cell death. Our results suggest a critical role for mitochondrial autophagy in the maintenance of immunological memory by protecting the metabolic quiescence and longevity of memory B cells.
Collapse
Affiliation(s)
- Srikanth Kodali
- * Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Min Li
- * Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Marietta M. Budai
- * Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Min Chen
- † Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX; .,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX; and.,Department of Surgery, Weill Cornell Medical College, Cornell University, New York, NY
| |
Collapse
|
10
|
Khatri I, Diks AM, van den Akker EB, Oosten LEM, Zwaginga JJ, Reinders MJT, van Dongen JJM, Berkowska MA. Longitudinal Dynamics of Human B-Cell Response at the Single-Cell Level in Response to Tdap Vaccination. Vaccines (Basel) 2021; 9:1352. [PMID: 34835283 PMCID: PMC8617659 DOI: 10.3390/vaccines9111352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/08/2021] [Accepted: 11/13/2021] [Indexed: 01/28/2023] Open
Abstract
To mount an adequate immune response against pathogens, stepwise mutation and selection processes are crucial functions of the adaptive immune system. To better characterize a successful vaccination response, we performed longitudinal (days 0, 5, 7, 10, and 14 after Boostrix vaccination) analysis of the single-cell transcriptome as well as the B-cell receptor (BCR) repertoire (scBCR-rep) in plasma cells of an immunized donor and compared it with baseline B-cell characteristics as well as flow cytometry findings. Based on the flow cytometry knowledge and literature findings, we discriminated individual B-cell subsets in the transcriptomics data and traced over-time maturation of plasmablasts/plasma cells (PB/PCs) and identified the pathways associated with the plasma cell maturation. We observed that the repertoire in PB/PCs differed from the baseline B-cell repertoire e.g., regarding expansion of unique clones in post-vaccination visits, high usage of IGHG1 in expanded clones, increased class-switching events post-vaccination represented by clonotypes spanning multiple IGHC classes and positive selection of CDR3 sequences over time. Importantly, the Variable gene family-based clustering of BCRs represented a similar measure as the gene-based clustering, but certainly improved the clustering of BCRs, as BCRs from duplicated Variable gene families could be clustered together. Finally, we developed a query tool to dissect the immune response to the components of the Boostrix vaccine. Using this tool, we could identify the BCRs related to anti-tetanus and anti-pertussis toxoid BCRs. Collectively, we developed a bioinformatic workflow which allows description of the key features of an ongoing (longitudinal) immune response, such as activation of PB/PCs, Ig class switching, somatic hypermutation, and clonal expansion, all of which are hallmarks of antigen exposure, followed by mutation & selection processes.
Collapse
Affiliation(s)
- Indu Khatri
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (I.K.); (A.M.D.); (M.A.B.)
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (E.B.v.d.A.); (M.J.T.R.)
| | - Annieck M. Diks
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (I.K.); (A.M.D.); (M.A.B.)
| | - Erik B. van den Akker
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (E.B.v.d.A.); (M.J.T.R.)
- Department of Molecular Epidemiology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Liesbeth E. M. Oosten
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.E.M.O.); (J.J.Z.)
| | - Jaap Jan Zwaginga
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.E.M.O.); (J.J.Z.)
| | - Marcel J. T. Reinders
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (E.B.v.d.A.); (M.J.T.R.)
- Delft Bioinformatics Lab, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Jacques J. M. van Dongen
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (I.K.); (A.M.D.); (M.A.B.)
| | - Magdalena A. Berkowska
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (I.K.); (A.M.D.); (M.A.B.)
| |
Collapse
|
11
|
Limited access to antigen drives generation of early B cell memory while restraining the plasmablast response. Immunity 2021; 54:2005-2023.e10. [PMID: 34525339 PMCID: PMC7612941 DOI: 10.1016/j.immuni.2021.08.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 05/26/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.
Collapse
|
12
|
Cheng ZY, He TT, Gao XM, Zhao Y, Wang J. ZBTB Transcription Factors: Key Regulators of the Development, Differentiation and Effector Function of T Cells. Front Immunol 2021; 12:713294. [PMID: 34349770 PMCID: PMC8326903 DOI: 10.3389/fimmu.2021.713294] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The development and differentiation of T cells represents a long and highly coordinated, yet flexible at some points, pathway, along which the sequential and dynamic expressions of different transcriptional factors play prominent roles at multiple steps. The large ZBTB family comprises a diverse group of transcriptional factors, and many of them have emerged as critical factors that regulate the lineage commitment, differentiation and effector function of hematopoietic-derived cells as well as a variety of other developmental events. Within the T-cell lineage, several ZBTB proteins, including ZBTB1, ZBTB17, ZBTB7B (THPOK) and BCL6 (ZBTB27), mainly regulate the development and/or differentiation of conventional CD4/CD8 αβ+ T cells, whereas ZBTB16 (PLZF) is essential for the development and function of innate-like unconventional γδ+ T & invariant NKT cells. Given the critical role of T cells in host defenses against infections/tumors and in the pathogenesis of many inflammatory disorders, we herein summarize the roles of fourteen ZBTB family members in the development, differentiation and effector function of both conventional and unconventional T cells as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhong-Yan Cheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ting-Ting He
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Abstract
Memory B cells (MBCs) are critical for the rapid development of protective immunity following re-infection. MBCs capable of neutralizing distinct subclasses of pathogens, such as influenza and HIV, have been identified in humans. However, efforts to develop vaccines that induce broadly protective MBCs to rapidly mutating pathogens have not yet been successful. Better understanding of the signals regulating MBC development and function are essential to overcome current challenges hindering successful vaccine development. Here, we discuss recent advancements regarding the signals and transcription factors regulating germinal centre-derived MBC development and function.
Collapse
Affiliation(s)
- Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Horns F, Dekker CL, Quake SR. Memory B Cell Activation, Broad Anti-influenza Antibodies, and Bystander Activation Revealed by Single-Cell Transcriptomics. Cell Rep 2021; 30:905-913.e6. [PMID: 31968262 PMCID: PMC7891556 DOI: 10.1016/j.celrep.2019.12.063] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 01/08/2023] Open
Abstract
Antibody memory protects humans from many diseases. Protective antibody memory responses require activation of transcriptional programs, cell proliferation, and production of antigen-specific antibodies, but how these aspects of the response are coordinated is poorly understood. We profile the molecular and cellular features of the antibody response to influenza vaccination by integrating single-cell transcriptomics, longitudinal antibody repertoire sequencing, and antibody binding measurements. Single-cell transcriptional profiling reveals a program of memory B cell activation characterized by CD11c and T-bet expression associated with clonal expansion and differentiation toward effector function. Vaccination elicits an antibody clone, which rapidly acquired broad high-affinity hemagglutinin binding during affinity maturation. Unexpectedly, many antibody clones elicited by vaccination do not bind vaccine, demonstrating non-specific activation of bystander antibodies by influenza vaccination. These results offer insight into how molecular recognition, transcriptional programs, and clonal proliferation are coordinated in the human B cell repertoire during memory recall. Antibody memory requires coordination of molecular recognition, gene expression programs, and clonal dynamics. Horns et al. study the human antibody memory response using single-cell and repertoire sequencing, revealing a transcriptional program of memory B cell activation, broadly binding anti-influenza antibodies, and widespread bystander activation of non-vaccine-binding antibodies after influenza vaccination.
Collapse
Affiliation(s)
- Felix Horns
- Biophysics Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Cornelia L Dekker
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
15
|
Price MJ, Scharer CD, Kania AK, Randall TD, Boss JM. Conserved Epigenetic Programming and Enhanced Heme Metabolism Drive Memory B Cell Reactivation. THE JOURNAL OF IMMUNOLOGY 2021; 206:1493-1504. [PMID: 33627377 DOI: 10.4049/jimmunol.2000551] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/25/2021] [Indexed: 02/01/2023]
Abstract
Memory B cells (MBCs) have enhanced capabilities to differentiate to plasma cells and generate a rapid burst of Abs upon secondary stimulation. To determine if MBCs harbor an epigenetic landscape that contributes to increased differentiation potential, we derived the chromatin accessibility and transcriptomes of influenza-specific IgM and IgG MBCs compared with naive cells. MBCs possessed an accessible chromatin architecture surrounding plasma cell-specific genes, as well as altered expression of transcription factors and genes encoding cell cycle, chemotaxis, and signal transduction processes. Intriguingly, this MBC signature was conserved between humans and mice. MBCs of both species possessed a heightened heme signature compared with naive cells. Differentiation in the presence of hemin enhanced oxidative phosphorylation metabolism and MBC differentiation into Ab-secreting plasma cells. Thus, these data define conserved MBC transcriptional and epigenetic signatures that include a central role for heme and multiple other pathways in augmenting MBC reactivation potential.
Collapse
Affiliation(s)
- Madeline J Price
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Anna K Kania
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322; .,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
16
|
Ripperger TJ, Bhattacharya D. Transcriptional and Metabolic Control of Memory B Cells and Plasma Cells. Annu Rev Immunol 2021; 39:345-368. [PMID: 33556247 DOI: 10.1146/annurev-immunol-093019-125603] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
For many infections and almost all vaccines, neutralizing-antibody-mediated immunity is the primary basis and best functional correlate of immunological protection. Durable long-term humoral immunity is mediated by antibodies secreted by plasma cells that preexist subsequent exposures and by memory B cells that rapidly respond to infections once they have occurred. In the midst of the current pandemic of coronavirus disease 2019, it is important to define our current understanding of the unique roles of memory B cells and plasma cells in immunity and the factors that control the formation and persistence of these cell types. This fundamental knowledge is the basis to interpret findings from natural infections and vaccines. Here, we review transcriptional and metabolic programs that promote and support B cell fates and functions, suggesting points at which these pathways do and do not intersect.
Collapse
Affiliation(s)
- Tyler J Ripperger
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, Arizona 85724, USA; ,
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, Arizona 85724, USA; ,
| |
Collapse
|
17
|
Wiggins KJ, Scharer CD. Roadmap to a plasma cell: Epigenetic and transcriptional cues that guide B cell differentiation. Immunol Rev 2020; 300:54-64. [PMID: 33278036 DOI: 10.1111/imr.12934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
Antibody-secreting cells (ASCs) or plasma cells secrete antibodies and form a cornerstone of humoral immunity. B cells that receive activation signals in the presence or absence of T cells initiate a differentiation program that requires epigenetic and transcriptional reprogramming in order to ultimately form ASC. Reprogramming is accomplished through the interplay of transcription factors that initiate gene expression programs and epigenetic mechanisms that maintain these programs and cell fates. An important consideration is that all of these factors are operating in the context of cell division. Recent technical advances now allow mechanistic studies to move beyond genetic studies to identify the promoters and enhancer repertoires that are regulated by epigenetic mechanisms and transcription factors in rare cell types and differentiation stages in vivo. This review will detail efforts to integrate transcriptional and epigenetic changes during B cell differentiation with cell division in vivo. What has emerged is a multiphased differentiation model that requires distinct transcription factors and epigenetic programs at each step. The identification of markers that define each phase will help facilitate the manipulation of B cell differentiation for vaccine development or to treat diseases where antibodies are a component.
Collapse
Affiliation(s)
- Keenan J Wiggins
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Ripperger TJ, Uhrlaub JL, Watanabe M, Wong R, Castaneda Y, Pizzato HA, Thompson MR, Bradshaw C, Weinkauf CC, Bime C, Erickson HL, Knox K, Bixby B, Parthasarathy S, Chaudhary S, Natt B, Cristan E, El Aini T, Rischard F, Campion J, Chopra M, Insel M, Sam A, Knepler JL, Capaldi AP, Spier CM, Dake MD, Edwards T, Kaplan ME, Scott SJ, Hypes C, Mosier J, Harris DT, LaFleur BJ, Sprissler R, Nikolich-Žugich J, Bhattacharya D. Orthogonal SARS-CoV-2 Serological Assays Enable Surveillance of Low-Prevalence Communities and Reveal Durable Humoral Immunity. Immunity 2020; 53:925-933.e4. [PMID: 33129373 PMCID: PMC7554472 DOI: 10.1016/j.immuni.2020.10.004] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022]
Abstract
We conducted a serological study to define correlates of immunity against SARS-CoV-2. Compared to those with mild coronavirus disease 2019 (COVID-19) cases, individuals with severe disease exhibited elevated virus-neutralizing titers and antibodies against the nucleocapsid (N) and the receptor binding domain (RBD) of the spike protein. Age and sex played lesser roles. All cases, including asymptomatic individuals, seroconverted by 2 weeks after PCR confirmation. Spike RBD and S2 and neutralizing antibodies remained detectable through 5-7 months after onset, whereas α-N titers diminished. Testing 5,882 members of the local community revealed only 1 sample with seroreactivity to both RBD and S2 that lacked neutralizing antibodies. This fidelity could not be achieved with either RBD or S2 alone. Thus, inclusion of multiple independent assays improved the accuracy of antibody tests in low-seroprevalence communities and revealed differences in antibody kinetics depending on the antigen. We conclude that neutralizing antibodies are stably produced for at least 5-7 months after SARS-CoV-2 infection.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Arizona/epidemiology
- Betacoronavirus/immunology
- Betacoronavirus/isolation & purification
- COVID-19
- COVID-19 Testing
- Clinical Laboratory Techniques/methods
- Coronavirus Infections/blood
- Coronavirus Infections/diagnosis
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Nucleocapsid Proteins
- Female
- Humans
- Immunity, Humoral
- Male
- Middle Aged
- Nucleocapsid Proteins/immunology
- Pandemics
- Phosphoproteins
- Pneumonia, Viral/blood
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Prevalence
- Protein Interaction Domains and Motifs
- SARS-CoV-2
- Seroepidemiologic Studies
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Young Adult
Collapse
Affiliation(s)
- Tyler J Ripperger
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Rachel Wong
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Division of Biological and Biomedical Sciences, Washington University, St. Louis, MO, USA
| | - Yvonne Castaneda
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Hannah A Pizzato
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Division of Biological and Biomedical Sciences, Washington University, St. Louis, MO, USA
| | - Mallory R Thompson
- Department of Surgery, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Christine Bradshaw
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Craig C Weinkauf
- Department of Surgery, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Heidi L Erickson
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Kenneth Knox
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Department of Medicine, University of Arizona, Phoenix, Phoenix, AZ, USA
| | - Billie Bixby
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Sairam Parthasarathy
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Sachin Chaudhary
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Bhupinder Natt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Elaine Cristan
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Tammer El Aini
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Franz Rischard
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Janet Campion
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Madhav Chopra
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Michael Insel
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Afshin Sam
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - James L Knepler
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Andrew P Capaldi
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Tucson, AZ, USA; Functional Genomics Core, University of Arizona, Tucson, AZ, USA
| | - Catherine M Spier
- Department of Pathology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Michael D Dake
- Office of the Senior Vice-President for Health Sciences, University of Arizona, Tucson, AZ, USA
| | - Taylor Edwards
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Matthew E Kaplan
- Functional Genomics Core, University of Arizona, Tucson, AZ, USA
| | - Serena Jain Scott
- Division of Geriatrics, General Medicine and Palliative Care, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Cameron Hypes
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Department of Emergency Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - Jarrod Mosier
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; Department of Emergency Medicine, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA
| | - David T Harris
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Health Sciences Biobank, University of Arizona, Tucson, AZ, USA
| | | | - Ryan Sprissler
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
19
|
Baumgarth N, Nikolich-Žugich J, Lee FEH, Bhattacharya D. Antibody Responses to SARS-CoV-2: Let's Stick to Known Knowns. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:2342-2350. [PMID: 32887754 PMCID: PMC7578055 DOI: 10.4049/jimmunol.2000839] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
The scale of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has thrust immunology into the public spotlight in unprecedented ways. In this article, which is part opinion piece and part review, we argue that the normal cadence by which we discuss science with our colleagues failed to properly convey likelihoods of the immune response to SARS-CoV-2 to the public and the media. As a result, biologically implausible outcomes were given equal weight as the principles set by decades of viral immunology. Unsurprisingly, questionable results and alarmist news media articles have filled the void. We suggest an emphasis on setting expectations based on prior findings while avoiding the overused approach of assuming nothing. After reviewing Ab-mediated immunity after coronavirus and other acute viral infections, we posit that, with few exceptions, the development of protective humoral immunity of more than a year is the norm. Immunity to SARS-CoV-2 is likely to follow the same pattern.
Collapse
Affiliation(s)
- Nicole Baumgarth
- Center for Immunology and Infectious Diseases, Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322
- Lowance Center for Human Immunology, Department of Medicine, Emory University, Atlanta, GA 30322; and
- Lowance Center for Human Immunology, Department of Pediatrics, Emory University, Atlanta, GA 30322
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724;
| |
Collapse
|
20
|
Moroney JB, Vasudev A, Pertsemlidis A, Zan H, Casali P. Integrative transcriptome and chromatin landscape analysis reveals distinct epigenetic regulations in human memory B cells. Nat Commun 2020; 11:5435. [PMID: 33116135 PMCID: PMC7595102 DOI: 10.1038/s41467-020-19242-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023] Open
Abstract
Memory B cells (MBCs) are long-lived and produce high-affinity, generally, class-switched antibodies. Here, we use a multiparameter approach involving CD27 to segregate naïve B cells (NBC), IgD+ unswitched (unsw)MBCs and IgG+ or IgA+ class-switched (sw)MBCs from humans of different age, sex and race. Conserved antibody variable gene expression indicates that MBCs emerge through unbiased selection from NBCs. Integrative analyses of mRNAs, miRNAs, lncRNAs, chromatin accessibility and cis-regulatory elements uncover a core mRNA-ncRNA transcriptional signature shared by IgG+ and IgA+ swMBCs and distinct from NBCs, while unswMBCs display a transitional transcriptome. Some swMBC transcriptional signature loci are accessible but not expressed in NBCs. Profiling miRNAs reveals downregulated MIR181, and concomitantly upregulated MIR181 target genes such as RASSF6, TOX, TRERF1, TRPV3 and RORα, in swMBCs. Finally, lncRNAs differentially expressed in swMBCs cluster proximal to the IgH chain locus on chromosome 14. Our findings thus provide new insights into MBC transcriptional programs and epigenetic regulation, opening new investigative avenues on these critical cell elements in human health and disease. Human memory B cells differentiate from naïve B cells and can express different immunoglobulin (Ig) isotypes resulted from class-switch recombination. Here the authors describe, using transcriptional and epigenetic data from human memory B cells and integrated multi-omics analyses, the differentiation regulation and trajectory of IgG+, IgA+ and IgD+ memory B cells.
Collapse
Affiliation(s)
- Justin B Moroney
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Anusha Vasudev
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Hong Zan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Paolo Casali
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
21
|
Wong R, Belk JA, Govero J, Uhrlaub JL, Reinartz D, Zhao H, Errico JM, D'Souza L, Ripperger TJ, Nikolich-Zugich J, Shlomchik MJ, Satpathy AT, Fremont DH, Diamond MS, Bhattacharya D. Affinity-Restricted Memory B Cells Dominate Recall Responses to Heterologous Flaviviruses. Immunity 2020; 53:1078-1094.e7. [PMID: 33010224 DOI: 10.1016/j.immuni.2020.09.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/11/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
Memory B cells (MBCs) can respond to heterologous antigens either by molding new specificities through secondary germinal centers (GCs) or by selecting preexisting clones without further affinity maturation. To distinguish these mechanisms in flavivirus infections and immunizations, we studied recall responses to envelope protein domain III (DIII). Conditional deletion of activation-induced cytidine deaminase (AID) between heterologous challenges of West Nile, Japanese encephalitis, Zika, and dengue viruses did not affect recall responses. DIII-specific MBCs were contained mostly within the plasma-cell-biased CD80+ subset, and few GCs arose following heterologous boosters, demonstrating that recall responses are confined by preexisting clonal diversity. Measurement of monoclonal antibody (mAb) binding affinity to DIII proteins, timed AID deletion, single-cell RNA sequencing, and lineage tracing experiments point to selection of relatively low-affinity MBCs as a mechanism to promote diversity. Engineering immunogens to avoid this MBC diversity may facilitate flavivirus-type-specific vaccines with minimized potential for infection enhancement.
Collapse
Affiliation(s)
- Rachel Wong
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Immunobiology, University of Arizona, Tucson, AZ 85724, USA
| | - Julia A Belk
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, USA
| | - Dakota Reinartz
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, USA
| | - Haiyan Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - John M Errico
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Lucas D'Souza
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, USA
| | - Tyler J Ripperger
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, USA
| | | | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | |
Collapse
|
22
|
Wong R, Bhattacharya D. ZBTB38 is dispensable for antibody responses. PLoS One 2020; 15:e0235183. [PMID: 32956421 PMCID: PMC7505459 DOI: 10.1371/journal.pone.0235183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Members of the broad complex, tram track, bric-a-brac and zinc finger (BTB-ZF) family of transcription factors, such as BCL-6, ZBTB20, and ZBTB32, regulate antigen-specific B cell differentiation, plasma cell longevity, and the duration of antibody production. We found that ZBTB38, a different member of the BTB-ZF family that binds methylated DNA at CpG motifs, is highly expressed by germinal center B cells and plasma cells. To define the functional role of ZBTB38 in B cell responses, we generated mice conditionally deficient in this transcription factor. Germinal center B cells lacking ZBTB38 dysregulated very few genes relative to wild-type and heterozygous littermate controls. Accordingly, mice with hematopoietic-specific deletion of Zbtb38 showed normal germinal center B cell numbers and antibody responses following immunization with hapten-protein conjugates. Memory B cells from these animals functioned normally in secondary recall responses. Despite expression of ZBTB38 in hematopoietic stem cells, progenitors and mature myeloid and lymphoid lineages were also present in normal numbers in mutant mice. These data demonstrate that ZBTB38 is dispensable for hematopoiesis and antibody responses. These conditional knockout mice may instead be useful in defining the functional importance of ZBTB38 in other cell types and contexts.
Collapse
Affiliation(s)
- Rachel Wong
- Division of Biological and Biomedical Sciences, Washington University in St. Louis, Saint Louis, MO, United States of America
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
- * E-mail:
| |
Collapse
|
23
|
Ripperger TJ, Uhrlaub JL, Watanabe M, Wong R, Castaneda Y, Pizzato HA, Thompson MR, Bradshaw C, Weinkauf CC, Bime C, Erickson HL, Knox K, Bixby B, Parthasarathy S, Chaudhary S, Natt B, Cristan E, Aini TE, Rischard F, Campion J, Chopra M, Insel M, Sam A, Knepler JL, Capaldi AP, Spier CM, Dake MD, Edwards T, Kaplan ME, Scott SJ, Hypes C, Mosier J, Harris DT, LaFleur BJ, Sprissler R, Nikolich-Žugich J, Bhattacharya D. Detection, prevalence, and duration of humoral responses to SARS-CoV-2 under conditions of limited population exposure. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.08.14.20174490. [PMID: 32817969 PMCID: PMC7430613 DOI: 10.1101/2020.08.14.20174490] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We conducted an extensive serological study to quantify population-level exposure and define correlates of immunity against SARS-CoV-2. We found that relative to mild COVID-19 cases, individuals with severe disease exhibited elevated authentic virus-neutralizing titers and antibody levels against nucleocapsid (N) and the receptor binding domain (RBD) and the S2 region of spike protein. Unlike disease severity, age and sex played lesser roles in serological responses. All cases, including asymptomatic individuals, seroconverted by 2 weeks post-PCR confirmation. RBD- and S2-specific and neutralizing antibody titers remained elevated and stable for at least 2-3 months post-onset, whereas those against N were more variable with rapid declines in many samples. Testing of 5882 self-recruited members of the local community demonstrated that 1.24% of individuals showed antibody reactivity to RBD. However, 18% (13/73) of these putative seropositive samples failed to neutralize authentic SARS-CoV-2 virus. Each of the neutralizing, but only 1 of the non-neutralizing samples, also displayed potent reactivity to S2. Thus, inclusion of multiple independent assays markedly improved the accuracy of antibody tests in low seroprevalence communities and revealed differences in antibody kinetics depending on the viral antigen. In contrast to other reports, we conclude that immunity is durable for at least several months after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Tyler J. Ripperger
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Jennifer L. Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Rachel Wong
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Division of Biological and Biomedical Sciences, Washington University, St. Louis, MO, USA
| | - Yvonne Castaneda
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Hannah A. Pizzato
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Division of Biological and Biomedical Sciences, Washington University, St. Louis, MO, USA
| | - Mallory R. Thompson
- Department of Surgery, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Christine Bradshaw
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Craig C. Weinkauf
- Department of Surgery, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Heidi L. Erickson
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Kenneth Knox
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Department of Medicine, University of Arizona-Phoenix, Phoenix, AZ
| | - Billie Bixby
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Sairam Parthasarathy
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Sachin Chaudhary
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Bhupinder Natt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Elaine Cristan
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Tammer El Aini
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Franz Rischard
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Janet Campion
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Madhav Chopra
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Michael Insel
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Afshin Sam
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - James L. Knepler
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Andrew P. Capaldi
- Department of Molecular and Cellular Biology, University of Arizona – Tucson, Tucson, AZ, USA
- Functional Genomics Core, University of Arizona, Tucson, AZ
| | - Catherine M. Spier
- Department of Pathology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
| | - Michael D. Dake
- Office of the Senior Vice-President for Health Sciences, University of Arizona, Tucson, USA
| | - Taylor Edwards
- University of Arizona Genomics Core, University of Arizona, Tucson, AZ and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | | | - Serena Jain Scott
- Division of Geriatrics, General Medicine and Palliative Care, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, USA
| | - Cameron Hypes
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Department of Emergency Medicine, University of Arizona College of Medicine-Tucson, Tucson, USA
| | - Jarrod Mosier
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- Department of Emergency Medicine, University of Arizona College of Medicine-Tucson, Tucson, USA
| | - David T. Harris
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Health Sciences Biobank, University of Arizona, Tucson, USA
| | | | - Ryan Sprissler
- University of Arizona Genomics Core, University of Arizona, Tucson, AZ and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine – Tucson, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, USA
| |
Collapse
|
24
|
Robinson JE, Greiner TC, Bouska AC, Iqbal J, Cutucache CE. Identification of a Splenic Marginal Zone Lymphoma Signature: Preliminary Findings With Diagnostic Potential. Front Oncol 2020; 10:640. [PMID: 32457837 PMCID: PMC7225304 DOI: 10.3389/fonc.2020.00640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Splenic marginal zone lymphoma (SMZL) is a rare, indolent non-Hodgkin's lymphoma that affects 0. 13 per 100,000 persons annually. Overall survival of SMZL is estimated to reach 8-11 years in most cases, but up to 30% of SMZL cases develop aggressive presentations resulting in greatly diminished time of survival. SMZL presents with a very heterogeneous molecular profile, making diagnosis problematic, and accurate prognosis even less likely. The study herein has identified a potential diagnostic gene expression signature with highly specific predictive utility, coined the SMZL-specific Gene Expression Signature (SSGES). Additionally, five of the most impactful markers identified within the SSGES were selected for a five-protein panel, for further evaluation among control and SMZL patient samples. These markers included EME2, ERCC5, SETBP1, USP24, and ZBTB32. When compared with control spleen and other B-cell lymphoma subtypes, significantly higher expression was noticed in SMZL samples when stained for EME2 and USP24. Additionally, ERCC5, SETBP1, USP24, and ZBTB32 staining displayed indications of prognostic value for SMZL patients. Delineation of the SSGES offers a unique SMZL signature that could provide diagnostic utility for a malignancy that has historically been difficult to identify, and the five-marker protein panel provides additional support for such findings. These results should be further investigated and validated in subsequent molecular investigations of SMZL so it may be potentially incorporated into standard oncology practice for improving the understanding and outlook for SMZL patients.
Collapse
Affiliation(s)
- Jacob E Robinson
- Department of Biology, University of Nebraska Omaha, Omaha, NE, United States
| | - Timothy C Greiner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Alyssa C Bouska
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | | |
Collapse
|
25
|
D'Souza L, Bhattacharya D. Plasma cells: You are what you eat. Immunol Rev 2019; 288:161-177. [PMID: 30874356 DOI: 10.1111/imr.12732] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022]
Abstract
Plasma cells are terminally differentiated B lymphocytes that constitutively secrete antibodies. These antibodies can provide protection against pathogens, and their quantity and quality are the best clinical correlates of vaccine efficacy. As such, plasma cell lifespan is the primary determinant of the duration of humoral immunity. Yet dysregulation of plasma cell function can cause autoimmunity or multiple myeloma. The longevity of plasma cells is primarily dictated by nutrient uptake and non-transcriptionally regulated metabolic pathways. We have previously shown a positive effect of glucose uptake and catabolism on plasma cell longevity and function. In this review, we discuss these findings with an emphasis on nutrient uptake and its effects on respiratory capacity, lifespan, endoplasmic reticulum stress, and antibody secretion in plasma cells. We further discuss how some of these pathways may be dysregulated in multiple myeloma, potentially providing new therapeutic targets. Finally, we speculate on the connection between plasma cell intrinsic metabolism and systemic changes in nutrient availability and metabolic diseases.
Collapse
Affiliation(s)
- Lucas D'Souza
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
26
|
Jash A, Zhou YW, Gerardo DK, Ripperger TJ, Parikh BA, Piersma S, Jamwal DR, Kiela PR, Boon ACM, Yokoyama WM, Hsieh CS, Bhattacharya D. ZBTB32 restrains antibody responses to murine cytomegalovirus infections, but not other repetitive challenges. Sci Rep 2019; 9:15257. [PMID: 31649328 PMCID: PMC6813321 DOI: 10.1038/s41598-019-51860-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 10/01/2019] [Indexed: 02/06/2023] Open
Abstract
ZBTB32 is a transcription factor that is highly expressed by a subset of memory B cells and restrains the magnitude and duration of recall responses against hapten-protein conjugates. To define physiological contexts in which ZBTB32 acts, we assessed responses by Zbtb32-/- mice or bone marrow chimeras against a panel of chronic and acute challenges. Mixed bone marrow chimeras were established in which all B cells were derived from either Zbtb32-/- mice or control littermates. Chronic infection of Zbtb32-/- chimeras with murine cytomegalovirus led to nearly 20-fold higher antigen-specific IgG2b levels relative to controls by week 9 post-infection, despite similar viral loads. In contrast, IgA responses and specificities in the intestine, where memory B cells are repeatedly stimulated by commensal bacteria, were similar between Zbtb32-/- mice and control littermates. Finally, an infection and heterologous booster vaccination model revealed no role for ZBTB32 in restraining primary or recall antibody responses against influenza viruses. Thus, ZBTB32 does not limit recall responses to a number of physiological acute challenges, but does restrict antibody levels during chronic viral infections that periodically engage memory B cells. This restriction might selectively prevent recall responses against chronic infections from progressively overwhelming other antibody specificities.
Collapse
Affiliation(s)
- Arijita Jash
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - You W Zhou
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Diana K Gerardo
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Tyler J Ripperger
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Bijal A Parikh
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Sytse Piersma
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Deepa R Jamwal
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Pawel R Kiela
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Wayne M Yokoyama
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Chyi S Hsieh
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America.,Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America. .,Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA.
| |
Collapse
|
27
|
Wong R, Bhattacharya D. Basics of memory B-cell responses: lessons from and for the real world. Immunology 2019; 156:120-129. [PMID: 30488482 PMCID: PMC6328991 DOI: 10.1111/imm.13019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/22/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
The production of pathogen-specific B cells and antibodies underlies protective immunity elicited by most vaccines and many infections. Humoral immunity follows a regulated process by which high-affinity antibody-secreting plasma cells and memory B cells are generated. Yet for certain pathogens, protective immunity is inefficiently generated and/or maintained. For example, Dengue virus infections lead to lasting immunity against re-infection by the same serotype. However, if infected with a different Dengue serotype, the individual is predisposed to more severe disease than if he/she was completely naive. As another example, both natural infections with or vaccination against malaria do not necessarily lead to lasting immunity, as the same individual can be re-infected many times over the course of a lifetime. In this review, we discuss how these real-world problems can both instruct and be informed by recent basic studies using model organisms and antigens. An emphasis is placed on protective epitopes and functional distinctions between memory B-cell subsets in both mice and humans. Using flavivirus and Plasmodium infections as examples, we also speculate on the differences between ineffective B-cell responses that actually occur in the real world, and perfect-world responses that would generate lasting immunity.
Collapse
Affiliation(s)
- Rachel Wong
- Division of Biological and Biomedical SciencesWashington UniversitySt LouisMOUSA
- Department of ImmunobiologyUniversity of Arizona College of MedicineTucsonAZUSA
| | - Deepta Bhattacharya
- Department of ImmunobiologyUniversity of Arizona College of MedicineTucsonAZUSA
| |
Collapse
|
28
|
Abstract
In this review, Boothby et al. summarize some salient advances toward elucidation of the molecular programming of the fate choices and function of B cells in the periphery. They also note unanswered questions that pertain to differences among subsets of B lymphocytes and plasma cells. Mature B lymphocytes are crucial components of adaptive immunity, a system essential for the evolutionary fitness of mammals. Adaptive lymphocyte function requires an initially naïve cell to proliferate extensively and its progeny to have the capacity to assume a variety of fates. These include either terminal differentiation (the long-lived plasma cell) or metastable transcriptional reprogramming (germinal center and memory B cells). In this review, we focus principally on the regulation of differentiation and functional diversification of the “B2” subset. An overview is combined with an account of more recent advances, including initial work on mechanisms that eliminate DNA methylation and potential links between intracellular metabolites and chromatin editing.
Collapse
|
29
|
Tellier J, Nutt SL. Plasma cells: The programming of an antibody‐secreting machine. Eur J Immunol 2018; 49:30-37. [DOI: 10.1002/eji.201847517] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/28/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Julie Tellier
- The Walter and Eliza Hall Institute of Medical Research Parkville Victoria Australia
- Department of Medical Biology University of Melbourne Parkville Victoria Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research Parkville Victoria Australia
- Department of Medical Biology University of Melbourne Parkville Victoria Australia
| |
Collapse
|
30
|
Lam WY, Jash A, Yao CH, D'Souza L, Wong R, Nunley RM, Meares GP, Patti GJ, Bhattacharya D. Metabolic and Transcriptional Modules Independently Diversify Plasma Cell Lifespan and Function. Cell Rep 2018; 24:2479-2492.e6. [PMID: 30157439 PMCID: PMC6172041 DOI: 10.1016/j.celrep.2018.07.084] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/15/2018] [Accepted: 07/25/2018] [Indexed: 01/12/2023] Open
Abstract
Plasma cell survival and the consequent duration of immunity vary widely with infection or vaccination. Using fluorescent glucose analog uptake, we defined multiple developmentally independent mouse plasma cell populations with varying lifespans. Long-lived plasma cells imported more fluorescent glucose analog, expressed higher surface levels of the amino acid transporter CD98, and had more autophagosome mass than did short-lived cells. Low amino acid concentrations triggered reductions in both antibody secretion and mitochondrial respiration, especially by short-lived plasma cells. To explain these observations, we found that glutamine was used for both mitochondrial respiration and anaplerotic reactions, yielding glutamate and aspartate for antibody synthesis. Endoplasmic reticulum (ER) stress responses, which link metabolism to transcriptional outcomes, were similar between long- and short-lived subsets. Accordingly, population and single-cell transcriptional comparisons across mouse and human plasma cell subsets revealed few consistent and conserved differences. Thus, plasma cell antibody secretion and lifespan are primarily defined by non-transcriptional metabolic traits.
Collapse
Affiliation(s)
- Wing Y Lam
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Arijita Jash
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cong-Hui Yao
- Department of Chemistry, Washington University, St. Louis, MO 63110, USA
| | - Lucas D'Souza
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Rachel Wong
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Ryan M Nunley
- Washington University Orthopedics, Barnes Jewish Hospital, St. Louis, MO 63110, USA
| | - Gordon P Meares
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63110, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
31
|
Zhu C, Chen G, Zhao Y, Gao XM, Wang J. Regulation of the Development and Function of B Cells by ZBTB Transcription Factors. Front Immunol 2018; 9:580. [PMID: 29616049 PMCID: PMC5869932 DOI: 10.3389/fimmu.2018.00580] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/07/2018] [Indexed: 12/20/2022] Open
Abstract
The large ZBTB family comprises a diverse group of transcriptional factors. Several ZBTB proteins have emerged as critical factors that regulate the lineage commitment, differentiation, and function of lymphoid cells as well as many other developmental events. For instance, dysfunctions of ZBTB20 or ZBTB24 have been linked to multisystem failures in humans. Within the B-cell lineage, BCL6, ZBTB7A, ZBTB17, and ZBTB1 regulate the development/differentiation of B cells in both bone marrow and peripheral lymphoid organs, while ZBTB20 and ZBTB32 seem to mainly impact the maintenance of terminal plasma cells. Given the importance of B cells in the prevention and treatment of infectious or autoimmune disorders, we herein summarize the roles of seven ZBTB family members (BCL6, ZBTB7A, ZBTB17, ZBTB20, ZBTB32, ZBTB1, and ZBTB24) in the development, differentiation, and function of B cells as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Can Zhu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ge Chen
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
32
|
Coley WD, Zhao Y, Benck CJ, Liu Y, Hotta-Iwamura C, Rahman MJ, Tarbell KV. Loss of Zbtb32 in NOD mice does not significantly alter T cell responses. F1000Res 2018; 7:318. [PMID: 29707204 PMCID: PMC5909056 DOI: 10.12688/f1000research.13864.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Background: We previously identified the transcriptional regulator Zbtb32 as a factor that can promote T cell tolerance in the Non-Obese Diabetic (NOD) mouse, a model of Type 1 diabetes. Antigen targeted to DCIR2
+ dendritic cells (DCs)
in vivo inhibited both diabetes and effector T cell expansion in NOD mice. Furthermore, Zbtb32 was preferentially induced in autoreactive CD4 T cells stimulated by these tolerogenic DCIR2
+ DCs, and overexpression of Zbtb32 in islet-specific T cells inhibited the diabetes development by limiting T cell proliferation and cytokine production. Methods: To further understand the role of Zbtb32 in T cell tolerance induction, we have now used CRISPR to target the Zbtb32 gene for deletion directly in NOD mice and characterized the mutant mice. We hypothesized that the systemic loss of Zbtb32 in NOD mice would lead to increased T cell activation and increased diabetes pathogenesis. Results: Although NOD.Zbtb32
-/- male NOD mice showed a trend towards increased diabetes incidence compared to littermate controls, the difference was not significant. Furthermore, no significant alteration in lymphocyte number or function was observed. Importantly,
in vitro stimulation of lymphocytes from NOD.Zbtb32
-/- mice did not produce the expected hypersensitive phenotype observed in other genetic strains, potentially due to compensation by homologous genes. Conclusions: The loss of Zbtb32 in the NOD background does not result in the expected T cell activation phenotype.
Collapse
Affiliation(s)
- William D Coley
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - Yongge Zhao
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - Charles J Benck
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - Yi Liu
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - Chie Hotta-Iwamura
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - M Jubayer Rahman
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA
| | - Kristin V Tarbell
- Immune Tolerance Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda , MD, 20892, USA.,Department of Inflammation and Oncology, Amgen, Inc, South San Francisco, CA, USA
| |
Collapse
|
33
|
Sandoval H, Kodali S, Wang J. Regulation of B cell fate, survival, and function by mitochondria and autophagy. Mitochondrion 2017; 41:58-65. [PMID: 29175010 DOI: 10.1016/j.mito.2017.11.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/13/2017] [Accepted: 11/19/2017] [Indexed: 01/31/2023]
Abstract
B cells are responsible for protective antibody production after differentiation into antibody-secreting cells during humoral immune responses. From early B cell development in the bone marrow, to their maturation in the periphery, activation in the germinal center, and differentiation into plasma cells or memory B cells, B cells display ever-changing functions and properties. Autophagy and mitochondria play important roles in B cell development, activation, and differentiation to accommodate the phenotypic and environmental changes encountered over the lifetime of the cell. Among their many functions, mitochondria and autophagy generate energy, mediate cell survival, and produce/eliminate reactive oxygen species that can serve as signal molecules to regulate differentiation. As B cells mature and differentiate into plasma or memory cells, both autophagic and mitochondrial functions undergo significant changes. In this review, we aim to provide an overview of the role of the autophagosome and mitochondria in regulating B cell fate, survival, and function. Moreover, we will discuss the interplay between these two highly metabolic organelles during B cell development, maturation, and differentiation.
Collapse
Affiliation(s)
- Hector Sandoval
- Immunobiology and Transplant Research Section, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Srikanth Kodali
- Immunobiology and Transplant Research Section, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jin Wang
- Immunobiology and Transplant Research Section, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Sintes J, Gentile M, Zhang S, Garcia-Carmona Y, Magri G, Cassis L, Segura-Garzón D, Ciociola A, Grasset EK, Bascones S, Comerma L, Pybus M, Lligé D, Puga I, Gutzeit C, He B, DuBois W, Crespo M, Pascual J, Mensa A, Aróstegui JI, Juan M, Yagüe J, Serrano S, Lloreta J, Meffre E, Hahne M, Cunningham-Rundles C, Mock BA, Cerutti A. mTOR intersects antibody-inducing signals from TACI in marginal zone B cells. Nat Commun 2017; 8:1462. [PMID: 29133782 PMCID: PMC5684130 DOI: 10.1038/s41467-017-01602-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) enhances immunity in addition to orchestrating metabolism. Here we show that mTOR coordinates immunometabolic reconfiguration of marginal zone (MZ) B cells, a pre-activated lymphocyte subset that mounts antibody responses to T-cell-independent antigens through a Toll-like receptor (TLR)-amplified pathway involving transmembrane activator and CAML interactor (TACI). This receptor interacts with mTOR via the TLR adapter MyD88. The resulting mTOR activation instigates MZ B-cell proliferation, immunoglobulin G (IgG) class switching, and plasmablast differentiation through a rapamycin-sensitive pathway that integrates metabolic and antibody-inducing transcription programs, including NF-κB. Disruption of TACI-mTOR interaction by rapamycin, truncation of the MyD88-binding domain of TACI, or B-cell-conditional mTOR deficiency interrupts TACI signaling via NF-κB and cooperation with TLRs, thereby hampering IgG production to T-cell-independent antigens but not B-cell survival. Thus, mTOR drives innate-like antibody responses by linking proximal TACI signaling events with distal immunometabolic transcription programs.
Collapse
Affiliation(s)
- Jordi Sintes
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain.
| | - Maurizio Gentile
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yolanda Garcia-Carmona
- Department of Medicine and Pediatrics, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Giuliana Magri
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Linda Cassis
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Daniel Segura-Garzón
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Alessandra Ciociola
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Emilie K Grasset
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Center for Molecular Medicine at Karolinska University Hospital, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Sabrina Bascones
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Laura Comerma
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Marc Pybus
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - David Lligé
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Irene Puga
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Cindy Gutzeit
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bing He
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wendy DuBois
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marta Crespo
- Department of Nephrology, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Julio Pascual
- Department of Nephrology, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain
| | - Anna Mensa
- Immunology Service, Hospital Clínic of Barcelona, Barcelona, 08036, Spain
| | | | - Manel Juan
- Immunology Service, Hospital Clínic of Barcelona, Barcelona, 08036, Spain
| | - Jordi Yagüe
- Immunology Service, Hospital Clínic of Barcelona, Barcelona, 08036, Spain
| | - Sergi Serrano
- Department of Pathology, Hospital del Mar, Barcelona, 08003, Spain
- Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Josep Lloreta
- Department of Pathology, Hospital del Mar, Barcelona, 08003, Spain
- Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Eric Meffre
- Department of Immunobiology, Yale University, New Haven, CT, 06511, USA
| | - Michael Hahne
- Molecular Genetics Institute of Montpellier, Montpellier, 34293, France
| | - Charlotte Cunningham-Rundles
- Department of Medicine and Pediatrics, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrea Cerutti
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, 08003, Spain.
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, 08003, Spain.
| |
Collapse
|
35
|
Lam WY, Bhattacharya D. Metabolic Links between Plasma Cell Survival, Secretion, and Stress. Trends Immunol 2017; 39:19-27. [PMID: 28919256 DOI: 10.1016/j.it.2017.08.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 01/12/2023]
Abstract
Humoral immunity is generated and maintained by antigen-specific antibodies that counter infectious pathogens. Plasma cells are the major producers of antibodies during and after infections, and each plasma cell produces some thousands of antibody molecules per second. This magnitude of secretion requires enormous quantities of amino acids and glycosylation sugars to properly build and fold antibodies, biosynthetic substrates to fuel endoplasmic reticulum (ER) biogenesis, and additional carbon sources to generate energy. Many of these processes are likely to be linked, thereby affording possibilities to improve vaccine design and to develop new therapies for autoimmunity. We review here aspects of plasma cell biology with an emphasis on recent studies and the relationships between intermediary metabolism, antibody production, and lifespan.
Collapse
Affiliation(s)
- Wing Y Lam
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Current address: Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
36
|
Shin HM, Kapoor VN, Kim G, Li P, Kim HR, Suresh M, Kaech SM, Wherry EJ, Selin LK, Leonard WJ, Welsh RM, Berg LJ. Transient expression of ZBTB32 in anti-viral CD8+ T cells limits the magnitude of the effector response and the generation of memory. PLoS Pathog 2017; 13:e1006544. [PMID: 28827827 PMCID: PMC5578684 DOI: 10.1371/journal.ppat.1006544] [Citation(s) in RCA: 21] [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: 04/25/2017] [Revised: 08/31/2017] [Accepted: 07/20/2017] [Indexed: 01/02/2023] Open
Abstract
Virus infections induce CD8+ T cell responses comprised of a large population of terminal effector cells and a smaller subset of long-lived memory cells. The transcription factors regulating the relative expansion versus the long-term survival potential of anti-viral CD8+ T cells are not completely understood. We identified ZBTB32 as a transcription factor that is transiently expressed in effector CD8+ T cells. After acute virus infection, CD8+ T cells deficient in ZBTB32 showed enhanced virus-specific CD8+ T cell responses, and generated increased numbers of virus-specific memory cells; in contrast, persistent expression of ZBTB32 suppressed memory cell formation. The dysregulation of CD8+ T cell responses in the absence of ZBTB32 was catastrophic, as Zbtb32-/- mice succumbed to a systemic viral infection and showed evidence of severe lung pathology. We found that ZBTB32 and Blimp-1 were co-expressed following CD8+ T cell activation, bound to each other, and cooperatively regulated Blimp-1 target genes Eomes and Cd27. These findings demonstrate that ZBTB32 is a key transcription factor in CD8+ effector T cells that is required for the balanced regulation of effector versus memory responses to infection. CD8+ T lymphocytes are essential for immune protection against viruses. In response to an infection, these cells are activated, proliferate, and generate antiviral effector cells that eradicate the infection. Following this, the majority of these effector cells die, leaving a small subset of long-lived virus-specific memory T cells. Our study identifies a transcription factor, ZBTB32, that is required for the regulation of CD8+ T cell responses. In its absence, antiviral CD8+ T cell numbers increase to abnormally high levels, and generate an overabundance of memory T cells. When this dysregulated response occurs following infection with a virus that cannot be rapidly eliminated by the immune system, the infected animals die from immune-mediated tissue damage, indicating the importance of this pathway.
Collapse
Affiliation(s)
- Hyun Mu Shin
- Dept of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Anatomy and Cell Biology, Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Varun N. Kapoor
- Dept of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Gwanghun Kim
- Department of Anatomy and Cell Biology, Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Peng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hang-Rae Kim
- Department of Anatomy and Cell Biology, Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - M. Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Susan M. Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - E. John Wherry
- Department of Microbiology and Institute for Immunology, University of Pennsylvania Perelman School Medicine, Philadelphia, Pennsylvania, United States of America
| | - Liisa K. Selin
- Dept of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Warren J. Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, United States of America
| | - Raymond M. Welsh
- Dept of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Leslie J. Berg
- Dept of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
37
|
Magri G, Comerma L, Pybus M, Sintes J, Lligé D, Segura-Garzón D, Bascones S, Yeste A, Grasset EK, Gutzeit C, Uzzan M, Ramanujam M, van Zelm MC, Albero-González R, Vazquez I, Iglesias M, Serrano S, Márquez L, Mercade E, Mehandru S, Cerutti A. Human Secretory IgM Emerges from Plasma Cells Clonally Related to Gut Memory B Cells and Targets Highly Diverse Commensals. Immunity 2017; 47:118-134.e8. [PMID: 28709802 PMCID: PMC5519504 DOI: 10.1016/j.immuni.2017.06.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/07/2017] [Accepted: 06/16/2017] [Indexed: 12/16/2022]
Abstract
Secretory immunoglobulin A (SIgA) enhances host-microbiota symbiosis, whereas SIgM remains poorly understood. We found that gut IgM+ plasma cells (PCs) were more abundant in humans than mice and clonally related to a large repertoire of memory IgM+ B cells disseminated throughout the intestine but rare in systemic lymphoid organs. In addition to sharing a gut-specific gene signature with memory IgA+ B cells, memory IgM+ B cells were related to some IgA+ clonotypes and switched to IgA in response to T cell-independent or T cell-dependent signals. These signals induced abundant IgM which, together with SIgM from clonally affiliated PCs, recognized mucus-embedded commensals. Bacteria recognized by human SIgM were dually coated by SIgA and showed increased richness and diversity compared to IgA-only-coated or uncoated bacteria. Thus, SIgM may emerge from pre-existing memory rather than newly activated naive IgM+ B cells and could help SIgA to anchor highly diverse commensal communities to mucus. IgM+ PCs generating SIgM are relatively abundant in human but not mouse gut IgM+ PCs clonally relate to a large gut repertoire of memory IgM+ B cells Gut memory IgM+ B cells express a tissue-specific signature and can switch to IgA Human but not mouse SIgM binds a highly diverse microbiota dually coated by SIgA
Collapse
Affiliation(s)
- Giuliana Magri
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain.
| | - Laura Comerma
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Marc Pybus
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Jordi Sintes
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - David Lligé
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Daniel Segura-Garzón
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Sabrina Bascones
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Ada Yeste
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Emilie K Grasset
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Cindy Gutzeit
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mathieu Uzzan
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Meera Ramanujam
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | | | - Ivonne Vazquez
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain
| | - Mar Iglesias
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain; Universitat Autònoma de Barcelona, Barcelona 08003, Spain
| | - Sergi Serrano
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain; Universitat Autònoma de Barcelona, Barcelona 08003, Spain
| | - Lucía Márquez
- Department of Gastroenterology, Hospital del Mar, Barcelona 08003, Spain
| | - Elena Mercade
- Department of Biology, Health and Environment, University of Barcelona, Barcelona 08028, Spain
| | - Saurabh Mehandru
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea Cerutti
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain; Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Catalan Institute for Research and Advanced Studies (ICREA), Barcelona 08003, Spain.
| |
Collapse
|
38
|
Abstract
We comprehensively review memory B cells (MBCs), covering the definition of MBCs and their identities and subsets, how MBCs are generated, where they are localized, how they are maintained, and how they are reactivated. Whereas naive B cells adopt multiple fates upon stimulation, MBCs are more restricted in their responses. Evolving work reveals that the MBC compartment in mice and humans consists of distinct subpopulations with differing effector functions. We discuss the various approaches to define subsets and subset-specific roles. A major theme is the need to both deliver faster effector function upon reexposure and readapt to antigenically variant pathogens while avoiding burnout, which would be the result if all MBCs generated only terminal effector function. We discuss cell-intrinsic differences in gene expression and signaling that underlie differences in function between MBCs and naive B cells and among MBC subsets and how this leads to memory responses.
Collapse
Affiliation(s)
- Florian Weisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| | - Mark Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| |
Collapse
|