401
|
Popescu M, Cabrera-Martinez B, Winslow GM. TNF-α Contributes to Lymphoid Tissue Disorganization and Germinal Center B Cell Suppression during Intracellular Bacterial Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:2415-2424. [PMID: 31570507 DOI: 10.4049/jimmunol.1900484] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
Bacterial, parasitic, and viral infections are well-known causes of lymphoid tissue disorganization, although the factors, both host and/or pathogen derived, that mediate these changes are largely unknown. Ehrlichia muris infection in mice causes a loss of germinal center (GC) B cells that is accompanied by the generation of extrafollicular T-bet+ CD11c+ plasmablasts and IgM memory B cells. We addressed a possible role for TNF-α in this process because this cytokine has been shown to regulate GC development. Ablation of TNF-α during infection resulted in an 8-fold expansion of GL7+ CD38lo CD95+ GC B cells, and a 2.5- and 5-fold expansion of CD138+ plasmablasts and T-bet+ memory cells, respectively. These changes were accompanied by a reduction in splenomegaly, more organized T and B cell zones, and an improved response to Ag challenge. CXCL13, the ligand for CXCR5, was detected at 6-fold higher levels following infection but was much reduced following TNF-α ablation, suggesting that CXCL13 dysregulation also contributes to loss of lymphoid tissue organization. T follicular helper cells, which also underwent expansion in infected TNF-α--deficient mice, may also have contributed to the expansion of T-bet+ B cells, as the latter are known to require T cell help. Our findings contrast with previously described roles for TNF-α in GCs and reveal how host-pathogen interactions can induce profound changes in cytokine and chemokine production that can alter lymphoid tissue organization, GC B cell development, and extrafollicular T-bet+ B cell generation.
Collapse
Affiliation(s)
- Maria Popescu
- Department of Microbiology and Immunology, Upstate Medical University, State University of New York, Syracuse, NY 13210
| | - Berenice Cabrera-Martinez
- Department of Microbiology and Immunology, Upstate Medical University, State University of New York, Syracuse, NY 13210
| | - Gary M Winslow
- Department of Microbiology and Immunology, Upstate Medical University, State University of New York, Syracuse, NY 13210
| |
Collapse
|
402
|
Hagen M, Derudder E. Inflammation and the Alteration of B-Cell Physiology in Aging. Gerontology 2019; 66:105-113. [PMID: 31553969 DOI: 10.1159/000501963] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 07/08/2019] [Indexed: 11/19/2022] Open
Abstract
Aging results for the immune system in a departure from the optimal homeostatic state seen in young organisms. This divergence regrettably contributes to a higher frequency of compromised responses to infections and inefficient classical vaccination in aged populations. In B cells, the cornerstone of humoral immunity, the development and distribution of the various mature B cell subsets are impacted by aging in both humans and mice. In addition, aged mature B cells demonstrate limited capacity to mount efficient antibody responses. An expected culprit for the decline in effective immunity is the rise of the systemic levels of pro-inflammatory molecules during aging, establishing a chronic low-grade inflammation. Indeed, numerous alterations affecting directly or indirectly B cells in old people and mice are reminiscent of various effects of acute inflammation on this cell type in young adults. The present mini-review will highlight the possible adverse contributions of the persistent low-level inflammation observed in susceptible older organisms to the inadequate B-cell physiology.
Collapse
Affiliation(s)
- Magdalena Hagen
- Institute for Biomedical Aging Research, University Innsbruck, Innsbruck, Austria
| | - Emmanuel Derudder
- Institute for Biomedical Aging Research, University Innsbruck, Innsbruck, Austria,
| |
Collapse
|
403
|
Excessive CD11c +Tbet + B cells promote aberrant T FH differentiation and affinity-based germinal center selection in lupus. Proc Natl Acad Sci U S A 2019; 116:18550-18560. [PMID: 31451659 DOI: 10.1073/pnas.1901340116] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Excessive self-reactive and inadequate affinity-matured antigen-specific antibody responses have been reported to coexist in lupus, with elusive cellular and molecular mechanisms. Here, we report that the antigen-specific germinal center (GC) response-a process critical for antibody affinity maturation-is compromised in murine lupus models. Importantly, this defect can be triggered by excessive autoimmunity-relevant CD11c+Tbet+ age-associated B cells (ABCs). In B cell-intrinsic Ship-deficient (ShipΔB) lupus mice, excessive CD11c+Tbet+ ABCs induce deregulated follicular T-helper (TFH) cell differentiation through their potent antigen-presenting function and consequently compromise affinity-based GC selection. Excessive CD11c+Tbet+ ABCs and deregulated TFH cell are also present in other lupus models and patients. Further, over-activated Toll-like receptor signaling in Ship-deficient B cells is critical for CD11c+Tbet+ ABC differentiation, and blocking CD11c+Tbet+ ABC differentiation in ShipΔB mice by ablating MyD88 normalizes TFH cell differentiation and rescues antigen-specific GC responses, as well as prevents autoantibody production. Our study suggests that excessive CD11c+Tbet+ ABCs not only contribute significantly to autoantibody production but also compromise antigen-specific GC B-cell responses and antibody-affinity maturation, providing a cellular link between the coexisting autoantibodies and inadequate affinity-matured antigen-specific antibodies in lupus models and a potential target for treating lupus.
Collapse
|
404
|
Johnson JL, Scholz JL, Marshak-Rothstein A, Cancro MP. Molecular pattern recognition in peripheral B cell tolerance: lessons from age-associated B cells. Curr Opin Immunol 2019; 61:33-38. [PMID: 31446338 DOI: 10.1016/j.coi.2019.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 01/06/2023]
Abstract
Although central tolerance mechanisms purge self-reactive B cells during development based on BCR signal strength, mechanisms that block the differentiation of autoreactive effector and memory B cells from mature pools remain poorly understood. Prior observations implicate nucleic acid sensing TLRs in autoimmunity, and more recent findings show that TLR9 is also involved in maintaining peripheral tolerance. Studies of the immunological changes that occur during aging revealed a subset of B cells denoted Age-associated B cells which expands in settings of aging and in autoimmunity. Further studies demonstrated that TLR9 signals poise activated B cells to adopt an Age-associated B cell phenotype, but BCR-delivered TLR9 signals cause programmed cell death that, if circumvented by costimulation, allows continued differentiation to the ABC fate. Together, these observations suggest molecular pattern recognition, rather than BCR epitope specificity per se, is a fundamental mediator of tolerogenic outcomes in the peripheral B cell activation.
Collapse
Affiliation(s)
- John L Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Jean L Scholz
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Ann Marshak-Rothstein
- Department of Medicine/Rheumatology, University of Massachusetts School of Medicine, Worcester, MA, United States
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| |
Collapse
|
405
|
Russell Knode LM, Park HS, Maul RW, Gearhart PJ. B cells from young and old mice switch isotypes with equal frequencies after ex vivo stimulation. Cell Immunol 2019; 345:103966. [PMID: 31447053 DOI: 10.1016/j.cellimm.2019.103966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 02/07/2023]
Abstract
To determine whether old B cells have the same capacity to switch isotypes as young cells, we purified splenic follicular, marginal zone, and age-associated B cell subsets from C57BL/6 mice. Cells were stimulated in culture with interleukin 4 and either lipopolysaccharide or anti-CD40, and switching to IgG1 was measured by flow cytometry of surface immunoglobulin. The results show that switching was robust in follicular and marginal zone B cells from old mice and was comparable to their young counterparts. However, age-associated B cells from old mice switched poorly relative to the other subsets. Expression of activation-induced deaminase, which initiates switching, was quantified by qPCR of mRNA, and it was equal between young and old follicular B cells. Thus, in this ex vivo system, the follicular and marginal zone cells from young and old mice behaved similarly, showing that the molecular machinery to perform switching is intact in old B cells.
Collapse
Affiliation(s)
- Lisa M Russell Knode
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States
| | - Han-Sol Park
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States
| | - Robert W Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States
| | - Patricia J Gearhart
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States.
| |
Collapse
|
406
|
Fraussen J, Marquez S, Takata K, Beckers L, Montes Diaz G, Zografou C, Van Wijmeersch B, Villar LM, O'Connor KC, Kleinstein SH, Somers V. Phenotypic and Ig Repertoire Analyses Indicate a Common Origin of IgD -CD27 - Double Negative B Cells in Healthy Individuals and Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2019; 203:1650-1664. [PMID: 31391234 DOI: 10.4049/jimmunol.1801236] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 07/11/2019] [Indexed: 11/19/2022]
Abstract
IgD-CD27- double negative (DN) B cells with proinflammatory characteristics are abnormally elevated in a proportion of multiple sclerosis (MS) patients. In this study, the origin and selection characteristics of DN B cells were studied in MS patients and healthy controls (HC). Expression of developmental markers on peripheral blood DN, IgD-CD27+ class-switched memory (CSM) and IgD+CD27- naive B cells of HC (n = 48) and MS patients (n = 96) was determined by flow cytometry. High-throughput adaptive immune receptor repertoire sequencing was performed on peripheral blood DN and CSM B cells of HC and MS patients (n = 3 each). DN B cells from HC and MS patients showed similar phenotypic and Ig repertoire characteristics. Phenotypic analysis indicated a mature state of DN B cells by low CD5, CD10, and CD38 expression. However, the frequency of CD95+ and IgA+ cells was lower in DN versus CSM B cells. DN B cells are Ag experienced, as shown by somatic hypermutation of their Ig genes in adaptive immune receptor repertoire sequencing, although they showed a lower mutation load than CSM B cells. Shared clones were found between DN and CSM B cells, although >95% of the clones were unique to each population, and differences in V(D)J usage and CDR3 physicochemical properties were found. Thus, DN B cells arise in HC and MS patients via a common developmental pathway that is probably linked to immune aging. However, DN and CSM B cells develop through unique differentiation pathways, with most DN B cells representing an earlier maturation state.
Collapse
Affiliation(s)
- Judith Fraussen
- Biomedical Research Institute, Hasselt University and School of Life Sciences, Transnational University Limburg, 3500 Hasselt, Belgium
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520
| | - Kazushiro Takata
- Department of Neurology, Yale School of Medicine, New Haven, CT 06519
| | - Lien Beckers
- Biomedical Research Institute, Hasselt University and School of Life Sciences, Transnational University Limburg, 3500 Hasselt, Belgium
| | - Gwendoline Montes Diaz
- Biomedical Research Institute, Hasselt University and School of Life Sciences, Transnational University Limburg, 3500 Hasselt, Belgium
| | | | - Bart Van Wijmeersch
- Biomedical Research Institute, Hasselt University and School of Life Sciences, Transnational University Limburg, 3500 Hasselt, Belgium.,Rehabilitation and MS-Center, 3900 Pelt, Belgium
| | - Luisa M Villar
- Departamento de Inmunologia, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Kevin C O'Connor
- Department of Neurology, Yale School of Medicine, New Haven, CT 06519; .,Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520; and
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520; .,Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520; and.,Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511
| | - Veerle Somers
- Biomedical Research Institute, Hasselt University and School of Life Sciences, Transnational University Limburg, 3500 Hasselt, Belgium
| |
Collapse
|
407
|
Wu C, Fu Q, Guo Q, Chen S, Goswami S, Sun S, Li T, Cao X, Chu F, Chen Z, Liu M, Liu Y, Fu T, Hao P, Hao Y, Shen N, Bao C, Zhang X. Lupus-associated atypical memory B cells are mTORC1-hyperactivated and functionally dysregulated. Ann Rheum Dis 2019; 78:1090-1100. [PMID: 31142473 PMCID: PMC6691860 DOI: 10.1136/annrheumdis-2019-215039] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES A population of atypical memory B cells (AtMs) are greatly expanded in patients with active lupus, but their generation and pathophysiological roles are poorly defined. The aim of this study was to comprehensively characterise lupus AtMs with a purpose to identify therapeutic clues to target this B cell population in lupus. METHODS Peripheral B cell subsets were measured by flow cytometry. Sorting-purified B cell subsets were subject to RNA sequencing and functional studies. Plasma cytokines and secreted immunoglobulins were detected by Luminex or ELISA. In situ renal B cells were detected by multiplexed immunohistochemistry. RESULTS CD24-CD20hi AtMs were strongly increased in two Chinese cohorts of patients with treatment-naïve lupus. Gene expression profile indicated that B cell signalling and activation, lipid/saccharide metabolism and endocytosis pathways were abnormally upregulated in lupus AtMs. In addition, the mammalian target of rapamycin complex 1 (mTORC1) pathway was remarkably activated in lupus AtMs, and blocking mTORC1 signalling by rapamycin abolished the generation of T-bet+ B cells and terminal differentiation of lupus AtMs. Furthermore, lupus AtMs displayed a dysfunctional phenotype, underwent accelerated apoptosis, poorly co-stimulated T cells and produced proinflammatory cytokines. Interestingly, lupus AtMs were in a paradoxically differentiated status with markers pro and against terminal differentiation and enriched with antinucleosome reactivity. Finally, AtMs were accumulated in the kidneys of patients with lupus nephritis and associated with disease severity. CONCLUSIONS These findings demonstrated that mTORC1-overactivated lupus AtMs are abnormally differentiated with metabolic and functional dysregulations. Inhibiting mTORC1 signalling might be an attractive option to target AtMs and to improve therapeutic effectiveness in patients with lupus.
Collapse
Affiliation(s)
- Chunmei Wu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qiong Fu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Guo
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Chen
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shyamal Goswami
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Shuhui Sun
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Teng Li
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xingjian Cao
- Department of Laboratory Medicine, The First Hospital of Nantong, Nantong, China
| | - Fuying Chu
- Department of Laboratory Medicine, The First Hospital of Nantong, Nantong, China
| | - Zechuan Chen
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Mei Liu
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yuanhua Liu
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Ting Fu
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Pei Hao
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yi Hao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Shen
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunde Bao
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
408
|
Age-associated changes in the impact of sex steroids on influenza vaccine responses in males and females. NPJ Vaccines 2019; 4:29. [PMID: 31312529 PMCID: PMC6626024 DOI: 10.1038/s41541-019-0124-6] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
Vaccine-induced immunity declines with age, which may differ between males and females. Using human sera collected before and 21 days after receipt of the monovalent A/Cal/09 H1N1 vaccine, we evaluated cytokine and antibody responses in adult (18-45 years) and aged (65+ years) individuals. After vaccination, adult females developed greater IL-6 and antibody responses than either adult males or aged females, with female antibody responses being positively associated with concentrations of estradiol. To test whether protection against influenza virus challenge was greater in females than males, we primed and boosted adult (8-10 weeks) and aged (68-70 weeks) male and female mice with an inactivated A/Cal/09 H1N1 vaccine or no vaccine and challenged with a drift variant A/Cal/09 virus. As compared with unvaccinated mice, vaccinated adult, but not aged, mice experienced less morbidity and better pulmonary viral clearance following challenge, regardless of sex. Vaccinated adult female mice developed antibody responses that were of greater quantity and quality and more protective than vaccinated adult males. Sex differences in vaccine efficacy diminished with age in mice. To determine the role of sex steroids in vaccine-induced immune responses, adult mice were gonadectomized and hormones (estradiol in females and testosterone in males) were replaced in subsets of animals before vaccination. Vaccine-induced antibody responses were increased in females by estradiol and decreased in males by testosterone. The benefit of elevated estradiol on antibody responses and protection against influenza in females is diminished with age in both mice and humans.
Collapse
|
409
|
Mácsik-Valent B, Nagy K, Fazekas L, Erdei A. Complement Receptor Type 1 (CR1, CD35), the Inhibitor of BCR-Mediated Human B Cell Activation, Differentially Regulates TLR7, and TLR9 Induced Responses. Front Immunol 2019; 10:1493. [PMID: 31312202 PMCID: PMC6614493 DOI: 10.3389/fimmu.2019.01493] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/14/2019] [Indexed: 11/13/2022] Open
Abstract
The complement system and Toll-like receptors (TLRs) are essential contributors of innate immunity. Separate activation of these systems has been shown to play a role in initiating and shaping the adaptive immune response, however the modulation of various B cell functions by the simultaneous involvement of these two systems has not yet been uncovered. We demonstrate here that occupancy of complement receptor type 1 (CR1, CD35) by its natural, complement component C3-derived ligand significantly and dose dependently reduces the TLR9-induced expression of activation markers, cytokine production, proliferation, and antibody production by human B cells, but has no effect on the TLR7-induced functions. The synergistic response to the simultaneous engagement of either TLR9 or TLR7 along with the BCR however, is significantly inhibited by CR1 occupancy. Our findings imply that both under physiological and pathological conditions, when complement- and TLR-activating microbial and damage products are present in the B cell environment, the cooperation between CR1 and TLR7 or TLR9 provides additional levels of the regulation of human B cell functions.
Collapse
Affiliation(s)
| | - Katinka Nagy
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | - László Fazekas
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
410
|
Epigenetic programming underpins B cell dysfunction in human SLE. Nat Immunol 2019; 20:1071-1082. [PMID: 31263277 PMCID: PMC6642679 DOI: 10.1038/s41590-019-0419-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is characterized by the expansion of extrafollicular pathogenic B cells derived from newly activated naïve cells. Although these cells express distinct markers, their epigenetic architecture and how it contributes to SLE remains poorly understood. To address this, we determined the DNA methylomes, chromatin accessibility and transcriptomes from five human B cell subsets, including a newly defined effector B cell subset from SLE and healthy subjects. Our data define a differentiation hierarchy between the subsets and elucidate the epigenetic and transcriptional differences between effector and memory B cells. Importantly, an SLE molecular signature was already established in resting naïve cells and was dominated by accessible chromatin enriched in AP-1 and EGR transcription factor motifs. Together, these factors acted in synergy with T-BET to shape the epigenome of expanded SLE effector B cell subsets. Thus, our data define the molecular foundation of pathogenic B cell dysfunction in SLE.
Collapse
|
411
|
Thorarinsdottir K, Camponeschi A, Jonsson C, Granhagen Önnheim K, Nilsson J, Forslind K, Visentini M, Jacobsson L, Mårtensson IL, Gjertsson I. CD21 -/low B cells associate with joint damage in rheumatoid arthritis patients. Scand J Immunol 2019; 90:e12792. [PMID: 31141193 DOI: 10.1111/sji.12792] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/03/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022]
Abstract
Depletion of B cells is beneficial in rheumatoid arthritis (RA) patients with autoantibodies to citrullinated proteins (ACPA) and/or the Fc portion of immunoglobulins (rheumatoid factor [RF]), suggesting a role for B cells in disease pathogenesis. To date, however, the identity of specifically pathogenic B cell subsets has not been discovered. One candidate population is identified by the low expression or absence of complement receptor 2 (CD21-/low B cells). In this study, we sought to determine whether there was any correlation between CD21-/low B cells and clinical outcome in patients with established RA, either ACPA+ /RF+ (n = 27) or ACPA- /RF- (n = 10). Healthy donors (n = 17) were included as controls. The proportion of the CD21-/low CD27- IgD- memory B cell subset in peripheral blood (PB) was significantly increased in ACPA+ /RF+ RA patients compared with healthy donors, and the frequency of this subset correlated with joint destruction (r = 0.57, P < 0.04). The levels of the chemokines CXCL-9 and CXCL-10 were higher in synovial fluid than in plasma, and PB CD21-/low cells expressed the receptor, CXCR3. In synovial fluid, most of the B cells were CD21-/low , approximately 40% of that population was CD27- IgD- , and a third of those expressed the pro-osteoclastogenic factor receptor activator of the nuclear factor κB ligand (RANKL). This subset also secreted RANKL, in addition to other factors such as IL-6, even in the absence of stimulation. We interpret these data as reason to propose the hypothesis that the CD27- IgD- subset of CD21-/low B cells may mediate joint destruction in patients with ACPA+ /RF+ RA.
Collapse
Affiliation(s)
- Katrin Thorarinsdottir
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Charlotte Jonsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Karin Granhagen Önnheim
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nilsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Forslind
- Section of Rheumatology, Department of Clinical Sciences, Lund University, Helsingborg, Sweden.,Section of Rheumatology, Department of Research and Education, Helsingborg's Hospital, Helsingborg, Sweden
| | - Marcella Visentini
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Lennart Jacobsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
412
|
Stone SL, Peel JN, Scharer CD, Risley CA, Chisolm DA, Schultz MD, Yu B, Ballesteros-Tato A, Wojciechowski W, Mousseau B, Misra RS, Hanidu A, Jiang H, Qi Z, Boss JM, Randall TD, Brodeur SR, Goldrath AW, Weinmann AS, Rosenberg AF, Lund FE. T-bet Transcription Factor Promotes Antibody-Secreting Cell Differentiation by Limiting the Inflammatory Effects of IFN-γ on B Cells. Immunity 2019; 50:1172-1187.e7. [PMID: 31076359 PMCID: PMC6929688 DOI: 10.1016/j.immuni.2019.04.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/04/2019] [Accepted: 04/12/2019] [Indexed: 01/07/2023]
Abstract
Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.
Collapse
Affiliation(s)
- Sara L Stone
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessica N Peel
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Christopher A Risley
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Danielle A Chisolm
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael D Schultz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bingfei Yu
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Wojciech Wojciechowski
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Betty Mousseau
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ravi S Misra
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Adedayo Hanidu
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Huiping Jiang
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Zhenhao Qi
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Troy D Randall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott R Brodeur
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Ananda W Goldrath
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Amy S Weinmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
413
|
Laffont S, Guéry JC. Deconstructing the sex bias in allergy and autoimmunity: From sex hormones and beyond. Adv Immunol 2019; 142:35-64. [PMID: 31296302 DOI: 10.1016/bs.ai.2019.04.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Men and women differ in their susceptibility to develop autoimmunity and allergy but also in their capacity to cope with infections. Mechanisms responsible for this sexual dimorphism are still poorly documented and probably multifactorial. This review discusses the recent development in our understanding of the cell-intrinsic actions of biological factors linked to sex, sex hormones and sex chromosome complement, on immune cells, which may account for the sex differences in the enhanced susceptibility of women to develop immunological disorders, such as allergic asthma or systemic lupus erythematosus (SLE). We choose to more specifically discuss the impact of sex hormones on the development and function of immune cell populations directly involved in type-2 immunity, and the role of the X-linked Toll like receptor 7 (TLR7) in anti-viral immunity and in SLE. We will also elaborate on the recent evidence demonstrating that TLR7 escapes from X chromosome inactivation in the immune cells of women, and how this may contribute to endow woman immune system with enhanced responsiveness to RNA-virus and susceptibility to SLE.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France.
| |
Collapse
|
414
|
Ambegaonkar AA, Nagata S, Pierce SK, Sohn H. The Differentiation in vitro of Human Tonsil B Cells With the Phenotypic and Functional Characteristics of T-bet+ Atypical Memory B Cells in Malaria. Front Immunol 2019; 10:852. [PMID: 31068937 PMCID: PMC6491666 DOI: 10.3389/fimmu.2019.00852] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/02/2019] [Indexed: 01/14/2023] Open
Abstract
Malaria is a deadly infectious disease associated with fundamental changes in the composition of the memory B cell (MBC) compartment, most notably a large expansion of T-bet+ MBCs, termed atypical MBCs. However, we know little about the precursors of atypical MBCs and the conditions that drive their differentiation. We compared the responses of human tonsil naïve B cells, MBCs, and germinal center B cells to a variety of stimulatory conditions. We determined that prolonged antigen presentation in the presence of CpG and IFN-γ induced maximal expression of T-bet and other phenotypic markers of malaria-associated atypical MBCs primarily in naïve B cells in vitro. Importantly T-bet+ naïve-derived B cells resembled atypical MBCs in their hypo-responsiveness to signaling through their B cell receptors. Thus, naïve B cells can be induced to differentiate into phenotypically and functionally atypical-like MBCs in vitro under conditions that may prevail in chronic infectious diseases in vivo.
Collapse
Affiliation(s)
- Abhijit A Ambegaonkar
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Satoshi Nagata
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
415
|
|
416
|
Abstract
A single exposure to many viral and bacterial pathogens typically induces life-long immunity, however, the development of the protective immunity to Plasmodium parasites is strikingly less efficient and achieves only partial protection, with adults residing in endemic areas often experiencing asymptomatic infections. Although naturally acquired immunity to malaria requires both cell-mediated and humoral immune responses, antibodies govern the control of malarial disease caused by the blood-stage form of the parasites. A large body of epidemiological evidence described that antibodies to Plasmodium antigens are inefficiently generated and rapidly lost without continued parasite exposure, suggesting that malaria is accompanied by defects in the development of immunological B cell memory. This topic has been of focus of recent studies of malaria infection in humans and mice. This review examines the main findings to date on the processes that modulate the acquisition of memory B cell responses to malaria, and highlights the importance of closing outstanding gaps of knowledge in the field for the rational design of next generation therapeutics against malaria.
Collapse
Affiliation(s)
- Ann Ly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Diana S Hansen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
417
|
Jenks SA, Cashman KS, Woodruff MC, Lee FEH, Sanz I. Extrafollicular responses in humans and SLE. Immunol Rev 2019; 288:136-148. [PMID: 30874345 PMCID: PMC6422038 DOI: 10.1111/imr.12741] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022]
Abstract
Chronic autoimmune diseases, and in particular Systemic Lupus Erythematosus (SLE), are endowed with a long-standing autoreactive B-cell compartment that is presumed to reactivate periodically leading to the generation of new bursts of pathogenic antibody-secreting cells (ASC). Moreover, pathogenic autoantibodies are typically characterized by a high load of somatic hypermutation and in some cases are highly stable even in the context of prolonged B-cell depletion. Long-lived, highly mutated antibodies are typically generated through T-cell-dependent germinal center (GC) reactions. Accordingly, an important role for GC reactions in the generation of pathogenic autoreactivity has been postulated in SLE. Nevertheless, pathogenic autoantibodies and autoimmune disease can be generated through B-cell extrafollicular (EF) reactions in multiple mouse models and human SLE flares are characterized by the expansion of naive-derived activated effector B cells of extrafollicular phenotype. In this review, we will discuss the properties of the EF B-cell pathway, its relationship to other effector B-cell populations, its role in autoimmune diseases, and its contribution to human SLE. Furthermore, we discuss the relationship of EF B cells with Age-Associated B cells (ABCs), a TLR-7-driven B-cell population that mediates murine autoimmune and antiviral responses.
Collapse
Affiliation(s)
- Scott A. Jenks
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Kevin S. Cashman
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Matthew C. Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - F. Eun-Hyung Lee
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Emory University, Atlanta, Georgia, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
418
|
Knox JJ, Myles A, Cancro MP. T-bet + memory B cells: Generation, function, and fate. Immunol Rev 2019; 288:149-160. [PMID: 30874358 PMCID: PMC6626622 DOI: 10.1111/imr.12736] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 12/16/2022]
Abstract
B cells expressing the transcription factor T-bet have emerged as participants in a number of protective and pathogenic immune responses. T-bet+ B cells characteristically differentiate in response to combined Toll-like receptor and cytokine signaling, contribute to protective immunity against intracellular pathogens via IgG2a/c production and antibody-independent mechanisms, and are prone to produce autoantibodies. Despite recent advances, a number of questions remain regarding the basic biology of T-bet+ B cells and their functional niche within the immune system. Herein, we review the discovery and defining characteristics of the T-bet+ B cell subset in both mice and humans. We further discuss their origins, the basis for their persistence, and their potential fate in vivo. Evidence indicates that T-bet+ B cells represent a distinct, germinal center-derived memory population that may serve as an important therapeutic target for the improvement of humoral immunity and prevention of autoimmunity.
Collapse
Affiliation(s)
- James J. Knox
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Arpita Myles
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael P. Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
419
|
Minto H, Mensah KA, Reynolds PR, Meffre E, Rubtsova K, Gelfand EW. A novel ATM mutation associated with elevated atypical lymphocyte populations, hyper-IgM, and cutaneous granulomas. Clin Immunol 2019; 200:55-63. [PMID: 30639167 PMCID: PMC7027322 DOI: 10.1016/j.clim.2019.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/26/2018] [Accepted: 01/08/2019] [Indexed: 12/25/2022]
Abstract
Ataxia-Telangiectasia (AT) is an immunodeficiency most often associated with T cell abnormalities. We describe a patient with a hyper-IgM phenotype and immune cell abnormalities that suggest a distinct clinical phenotype. Significant B cell abnormalities with increased unswitched memory B cells, decreased naive transitional B cells, and an elevated frequency of CD19+CD38loCD27-CD10-CD21-/low B cells expressing high levels of T-bet and Fas were demonstrated. The B cells were hyporesponsive to in vitro stimulation through the B cell receptor, Toll like receptors (TLR) 7 and 9, and CD40. T cell homeostasis was also disturbed with a significant increase in γδ T cells, circulating T follicular helper cells (Tfh), and decreased numbers of T regulatory cells. The ATM mutations in this patient are posited to have resulted in the perturbations in the frequencies and distributions of B and T cell subsets, resulting in the phenotype in this patient. KEY MESSAGES: A novel mutation creating a premature stop codon and a nonsense mutation in the ATM gene are postulated to have resulted in the unique clinical picture characterized by abnormal B and T cell populations, lymphocyte subset dysfunction, granuloma formation, and a hyper-IgM phenotype. CAPSULE SUMMARY: A patient presented with ataxia-telangiectasia, cutaneous granulomas, and a hyper-IgM phenotype; a novel combination of mutations in the ATM gene was associated with abnormal distributions, frequencies, and function of T and B lymphocyte subsets.
Collapse
Affiliation(s)
- Heather Minto
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, CO 80206, United States
| | - Kofi A Mensah
- Department of Immunobiology and Division of Rheumatology, Yale University School of Medicine, New Haven, CT 06511, United States
| | - Paul R Reynolds
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, CO 80206, United States
| | - Eric Meffre
- Department of Immunobiology and Division of Rheumatology, Yale University School of Medicine, New Haven, CT 06511, United States
| | - Kira Rubtsova
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, United States
| | - Erwin W Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, CO 80206, United States.
| |
Collapse
|
420
|
Ma S, Wang C, Mao X, Hao Y. B Cell Dysfunction Associated With Aging and Autoimmune Diseases. Front Immunol 2019; 10:318. [PMID: 30873171 PMCID: PMC6400972 DOI: 10.3389/fimmu.2019.00318] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/06/2019] [Indexed: 01/08/2023] Open
Abstract
Impaired humoral responses, as well as an increased propensity for autoimmunity, play an important role in the development of immune system dysfunction associated with aging. Accumulation of a subset of atypical B cells, termed age-associated B cells (ABCs), is one of the key age-related changes in B cell compartments. ABCs are characterized by their distinct phenotypes, gene expression profiles, special survival requirements, variations in B cell receptor repertoires, and unique functions. Here, we summarize recent progress in the knowledge base related to the features of ABCs, their potential role in immune senescence, and their relationship with autoimmune diseases.
Collapse
Affiliation(s)
- Shiliang Ma
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengwei Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinru Mao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Hao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
421
|
Shaw TN, Inkson CA, Villegas-Mendez A, Pattinson DJ, Strangward P, Else KJ, Draper SJ, Zeef LAH, Couper KN. Infection-Induced Resistance to Experimental Cerebral Malaria Is Dependent Upon Secreted Antibody-Mediated Inhibition of Pathogenic CD8 + T Cell Responses. Front Immunol 2019; 10:248. [PMID: 30846985 PMCID: PMC6394254 DOI: 10.3389/fimmu.2019.00248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.
Collapse
Affiliation(s)
- Tovah N. Shaw
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Colette A. Inkson
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Ana Villegas-Mendez
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | | | - Patrick Strangward
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Kathryn J. Else
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Leo A. H. Zeef
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
422
|
Myles A, Sanz I, Cancro MP. T-bet + B cells: A common denominator in protective and autoreactive antibody responses? Curr Opin Immunol 2019; 57:40-45. [PMID: 30784957 DOI: 10.1016/j.coi.2019.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
T-bet+ B cells have emerged as a key component of the humoral immune response in both infections and autoimmune disorders, with many of their phenotypic and functional attributes conserved between mice and humans. They are protective (infections) and pathogenic (autoimmunity), although the associated commonalities and differences remain unclear. Heterogeneity within this pool, in terms of origin, fate and function may underlie these divergent roles. Their significance is context-dependent- they may constitute a persistent effector memory cell pool, or products of recent primary responses. In both cases however, T-bet+ cells likely represent antigen-experienced progenitors of antibody-secreting cells with multipotent properties. Given their key contributions to both immunity and disease, T-bet+ B cells are an attractive target for vaccination and therapeutic strategies.
Collapse
Affiliation(s)
- Arpita Myles
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ignacio Sanz
- Lowance Center for Human Immunology, Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
423
|
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
|
424
|
Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol 2019; 9:3176. [PMID: 30719026 PMCID: PMC6348254 DOI: 10.3389/fimmu.2018.03176] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells, uniquely able to induce naïve T cell activation and effector differentiation. They are, likewise, involved in the induction and maintenance of immune tolerance in homeostatic conditions. Their phenotypic and functional heterogeneity points to their great plasticity and ability to modulate, according to their microenvironment, the acquired immune response and, at the same time, makes their precise classification complex and frequently subject to reviews and improvement. This review will present general aspects of the DC physiology and classification and will address their potential and actual uses in the management of human disease, more specifically cancer, as therapeutic and monitoring tools. New combination treatments with the participation of DC will be also discussed.
Collapse
Affiliation(s)
- Thiago A Patente
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana P Pinho
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline A Oliveira
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela C M Evangelista
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia C Bergami-Santos
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José A M Barbuto
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Discipline of Molecular Medicine, Department of Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
425
|
Caielli S, Veiga DT, Balasubramanian P, Athale S, Domic B, Murat E, Banchereau R, Xu Z, Chandra M, Chung CH, Walters L, Baisch J, Wright T, Punaro M, Nassi L, Stewart K, Fuller J, Ucar D, Ueno H, Zhou J, Banchereau J, Pascual V. A CD4 + T cell population expanded in lupus blood provides B cell help through interleukin-10 and succinate. Nat Med 2019; 25:75-81. [PMID: 30478422 PMCID: PMC6325012 DOI: 10.1038/s41591-018-0254-9] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 10/15/2018] [Indexed: 01/15/2023]
Abstract
Understanding the mechanisms underlying autoantibody development will accelerate therapeutic target identification in autoimmune diseases such as systemic lupus erythematosus (SLE)1. Follicular helper T cells (TFH cells) have long been implicated in SLE pathogenesis. Yet a fraction of autoantibodies in individuals with SLE are unmutated, supporting that autoreactive B cells also differentiate outside germinal centers2. Here, we describe a CXCR5-CXCR3+ programmed death 1 (PD1)hiCD4+ helper T cell population distinct from TFH cells and expanded in both SLE blood and the tubulointerstitial areas of individuals with proliferative lupus nephritis. These cells produce interleukin-10 (IL-10) and accumulate mitochondrial reactive oxygen species as the result of reverse electron transport fueled by succinate. Furthermore, they provide B cell help, independently of IL-21, through IL-10 and succinate. Similar cells are generated in vitro upon priming naive CD4+ T cells with plasmacytoid dendritic cells activated with oxidized mitochondrial DNA, a distinct class of interferogenic toll-like receptor 9 ligand3. Targeting this pathway might blunt the initiation and/or perpetuation of extrafollicular humoral responses in SLE.
Collapse
Affiliation(s)
- Simone Caielli
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | | - Preetha Balasubramanian
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Shruti Athale
- Baylor Institute for Immunology Research, Dallas, TX, USA
| | - Bojana Domic
- Baylor Institute for Immunology Research, Dallas, TX, USA
| | - Elise Murat
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | | - Zhaohui Xu
- Baylor Institute for Immunology Research, Dallas, TX, USA
| | | | - Cheng-Han Chung
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Lynnette Walters
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Jeanine Baisch
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Tracey Wright
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marilynn Punaro
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lorien Nassi
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Katie Stewart
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Julie Fuller
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hideki Ueno
- Baylor Institute for Immunology Research, Dallas, TX, USA
- Mount Sinai School of Medicine, New York, NY, USA
| | - Joseph Zhou
- Pathologists Bio-Medical Laboratories, Lewisville, TX, USA
| | | | - Virginia Pascual
- Baylor Institute for Immunology Research, Dallas, TX, USA.
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA.
| |
Collapse
|
426
|
Villa A, Notarangelo LD. RAG gene defects at the verge of immunodeficiency and immune dysregulation. Immunol Rev 2019; 287:73-90. [PMID: 30565244 PMCID: PMC6309314 DOI: 10.1111/imr.12713] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
Abstract
Mutations of the recombinase activating genes (RAG) in humans underlie a broad spectrum of clinical and immunological phenotypes that reflect different degrees of impairment of T- and B-cell development and alterations of mechanisms of central and peripheral tolerance. Recent studies have shown that this phenotypic heterogeneity correlates, albeit imperfectly, with different levels of recombination activity of the mutant RAG proteins. Furthermore, studies in patients and in newly developed animal models carrying hypomorphic RAG mutations have disclosed various mechanisms underlying immune dysregulation in this condition. Careful annotation of clinical outcome and immune reconstitution in RAG-deficient patients who have received hematopoietic stem cell transplantation has shown that progress has been made in the treatment of this disease, but new approaches remain to be tested to improve stem cell engraftment and durable immune reconstitution. Finally, initial attempts have been made to treat RAG deficiency with gene therapy.
Collapse
Affiliation(s)
- Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
427
|
B cell alterations during BAFF inhibition with belimumab in SLE. EBioMedicine 2018; 40:517-527. [PMID: 30593436 PMCID: PMC6412067 DOI: 10.1016/j.ebiom.2018.12.035] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/10/2018] [Accepted: 12/17/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a systemic autoimmune disease, which exhibits multiple B cell abnormalities including expanded populations of memory B cells and elevated levels of autoantibodies. Belimumab is a monoclonal antibody targeting the B cell cytokine BAFF (a.k.a. BLyS), approved for the treatment of SLE. METHODS In this prospective cohort study, B cells from peripheral blood of 23 SLE patients initiating belimumab treatment and followed longitudinally for up to three years, were assessed using mass cytometry. FINDINGS B cells decreased during the study period, with a rapid decrease of both naïve and CD11c+CD21- B cells at the first follow-up visit, followed by a continuous reduction at subsequent follow-ups. In contrast, plasma cells and switched memory B cells remained stable throughout the study. The observed immunological changes correlated with early, but not late, clinical improvements. Moreover, high baseline B cell counts were predictive of failure to attain low disease activity. In summary, our data unveiled both rapid and gradual later therapy-associated alterations of both known and unforeseen B cell phenotypes. INTERPRETATION Our results suggest that evaluation of B cell counts might prove useful prior to initiation of belimumab treatment and that early treatment evaluation and discontinuation might underestimate delayed clinical improvements resultant of late B cell changes.
Collapse
|
428
|
Fukushima Y, Minato N, Hattori M. The impact of senescence-associated T cells on immunosenescence and age-related disorders. Inflamm Regen 2018; 38:24. [PMID: 30603051 PMCID: PMC6304761 DOI: 10.1186/s41232-018-0082-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/30/2018] [Indexed: 12/18/2022] Open
Abstract
Immunosenescence is age-associated changes in the immunological functions, including diminished acquired immunity against infection, pro-inflammatory traits, and increased risk of autoimmunity. The proportions of memory-phenotype T cells in the peripheral T cell population steadily increase with age, but the relationship between this change and immunosenescent phenotypes remains elusive. Recently, we identified a minor memory-phenotype CD4+ T cell subpopulation that constitutively expressed PD-1 and CD153 as a bona fide age-dependent T cell population; we termed these cells senescence-associated T (SA-T) cells. SA-T cells exhibit characteristic features of cellular senescence, with defective T cell receptor-mediated proliferation and T cell cytokine production. However, upon T cell receptor stimulation, SA-T cells secrete abundant atypical pro-inflammatory cytokines such as osteopontin and chemokines, reminiscent of the SA-secretory phenotype. In addition to aging, SA-T cells accumulate and cause persistent inflammation in tissues following a wide range of insults including immune complex deposition, metabolic stresses, vascular damages, and tumors. In this review, we summarize the recent understanding of immunosenescence with particular focus on SA-T cells and their role in various age-related disorders.
Collapse
Affiliation(s)
- Yuji Fukushima
- 1Department of Immunosenescence, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Nagahiro Minato
- 2DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Masakazu Hattori
- 1Department of Immunosenescence, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| |
Collapse
|
429
|
Age (autoimmunity) associated B cells (ABCs) and their relatives. Curr Opin Immunol 2018; 55:75-80. [DOI: 10.1016/j.coi.2018.09.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
|
430
|
Romero-Ramírez S, Navarro-Hernandez IC, Cervantes-Díaz R, Sosa-Hernández VA, Acevedo-Ochoa E, Kleinberg-Bild A, Valle-Rios R, Meza-Sánchez DE, Hernández-Hernández JM, Maravillas-Montero JL. Innate-like B cell subsets during immune responses: Beyond antibody production. J Leukoc Biol 2018; 105:843-856. [PMID: 30457676 DOI: 10.1002/jlb.mr0618-227r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022] Open
Abstract
B lymphocytes are recognized for their crucial role in the adaptive immunity since they represent the only leukocyte lineage capable of differentiating into Ab-secreting cells. However, it has been demonstrated that these lymphocytes can exert several Ab-independent functions, including engulfing and processing Ags for presentation to T cells, secreting soluble mediators, providing co-stimulatory signals, and even participating in lymphoid tissues development. Beyond that, several reports claiming the existence of multiple B cell subsets contributing directly to innate immune responses have appeared. These "innate-like" B lymphocytes, whose phenotype, development pathways, tissue distribution, and functions are in most cases notoriously different from those of conventional B cells, are crucial to early protective responses against pathogens by exerting "crossover" defensive strategies that blur the established boundaries of innate and adaptive branches of immunity. Examples of these mechanisms include the rapid secretion of the polyspecific natural Abs, increased susceptibility to innate receptors-mediated activation, cytokine secretion, downstream priming of other innate cells, usage of specific variable immunoglobulin gene-segments, and other features. As these new insights emerge, it is becoming preponderant to redefine the functionality of B cells beyond their classical adaptive-immune tasks.
Collapse
Affiliation(s)
- Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Itze C Navarro-Hernandez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Víctor A Sosa-Hernández
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ernesto Acevedo-Ochoa
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Ari Kleinberg-Bild
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Ricardo Valle-Rios
- División de Investigación de la Facultad de Medicina, Universidad Nacional Autónoma de México y Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - David E Meza-Sánchez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José M Hernández-Hernández
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - José L Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
431
|
Biological sex affects vaccine efficacy and protection against influenza in mice. Proc Natl Acad Sci U S A 2018; 115:12477-12482. [PMID: 30455317 DOI: 10.1073/pnas.1805268115] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Biological sex affects adaptive immune responses, which could impact influenza infection and vaccine efficacy. Infection of mice with 2009 H1N1 induced antibody responses, CD4+ T cell and CD8+ T cell memory responses that were greater in females than males; both sexes, however, were equally protected against secondary challenge with an H1N1 drift variant virus. To test whether greater antibody in females is sufficient for protection against influenza, males and females were immunized with an inactivated H1N1 vaccine that induced predominantly antibody-mediated immunity. Following vaccination, females had greater antibody responses and protection against challenge with an H1N1 drift variant virus than males. Antibody derived from vaccinated females was better at protecting both naïve males and females than antibody from males, and this protection was associated with increased antibody specificity and avidity to the H1N1 virus. The expression of Tlr7 was greater in B cells from vaccinated females than males and was associated with reduced DNA methylation in the Tlr7 promoter region, higher neutralizing antibody, class switch recombination, and antibody avidity in females. Deletion of Tlr7 reduced sex differences in vaccine-induced antibody responses and protection following challenge and had a greater impact on responses in females than males. Taken together, these data illustrate that greater TLR7 activation and antibody production in females improves the efficacy of vaccination against influenza.
Collapse
|
432
|
Du SW, Arkatkar T, Jacobs HM, Rawlings DJ, Jackson SW. Generation of functional murine CD11c + age-associated B cells in the absence of B cell T-bet expression. Eur J Immunol 2018; 49:170-178. [PMID: 30353919 DOI: 10.1002/eji.201847641] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/21/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
Age-associated B cells (ABC), a novel subset of activated B cells defined by CD11b and CD11c expression, have been linked with both protective anti-viral responses and the pathogenesis of systemic autoimmunity. Expression of the TH 1 lineage transcription factor T-bet has been identified as a defining feature of ABC biology, with B cell-intrinsic expression of this transcription factor proposed to be required for ABC formation. In contrast to this model, we report that Tbx21 (encoding T-bet)-deficient B cells upregulate CD11b and CD11c surface expression in vitro in response to integrated TLR and cytokine signals. Moreover, B cell-intrinsic T-bet deletion in a murine lupus model exerted no impact of ABC generation in vivo, with Tbx21-/- ABCs exhibiting an identical surface phenotype to wild-type (WT) ABCs. Importantly, WT and Tbx21-/- ABCs sorted from autoimmune mice produced equivalent amounts of IgM and IgG ex vivo following TLR stimulation, indicating that T-bet-deficient ABCs are likely functional in vivo. In summary, our data contradict the established literature by demonstrating that T-bet expression is not uniformly required for ABC generation.
Collapse
Affiliation(s)
- Samuel W Du
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Tanvi Arkatkar
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Holly M Jacobs
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.,Departments of Immunology and Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Shaun W Jackson
- Center for Immunity and Immunotherapy, Seattle Children's Research Institute, Seattle, WA, USA.,Departments of Immunology and Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
433
|
Sawai CM, Serpas L, Neto AG, Jang G, Rashidfarrokhi A, Kolbeck R, Sanjuan MA, Reizis B, Sisirak V. Plasmacytoid Dendritic Cells Are Largely Dispensable for the Pathogenesis of Experimental Inflammatory Bowel Disease. Front Immunol 2018; 9:2475. [PMID: 30410494 PMCID: PMC6209677 DOI: 10.3389/fimmu.2018.02475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition caused by an aberrant immune response to microbial components of the gastrointestinal tract. Plasmacytoid dendritic cells (pDCs) are innate immune cells specialized in the production of type I interferons and were recently implicated in the pathogenesis of autoimmune disorders such as lupus and scleroderma. While pDCs were shown to infiltrate intestinal mucosa of IBD patients and proposed to participate in intestinal inflammation, their net contribution to the disease remains unclear. We addressed this question by targeting the pDC-specific transcription factor TCF4 (E2-2) in experimental IBD caused by deficiency of Wiskott-Aldrich syndrome protein (WASP) or of interleukin-10 (IL-10). Monoallelic Tcf4 deletion, which was previously shown to abrogate experimental lupus, did not affect autoimmunity manifestations or colitis in WASP-deficient animals. Furthermore, conditional biallelic Tcf4 targeting resulted in a near-complete pDC ablation, yet had no effect on the development of colitis in IL-10-deficient mice. Our results suggest that, in contrast to other inflammatory and autoimmune diseases, pDCs do not play a major role in the pathogenesis of intestinal inflammation during IBD.
Collapse
Affiliation(s)
- Catherine M Sawai
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,INSERM, ACTION Laboratory, University of Bordeaux, Bordeaux, France
| | - Lee Serpas
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Antonio Galvao Neto
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Geunhyo Jang
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Miguel A Sanjuan
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Vanja Sisirak
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,CNRS-UMR, Immunoconcept, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
434
|
Jenks SA, Cashman KS, Zumaquero E, Marigorta UM, Patel AV, Wang X, Tomar D, Woodruff MC, Simon Z, Bugrovsky R, Blalock EL, Scharer CD, Tipton CM, Wei C, Lim SS, Petri M, Niewold TB, Anolik JH, Gibson G, Lee FEH, Boss JM, Lund FE, Sanz I. Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus. Immunity 2018; 49:725-739.e6. [PMID: 30314758 PMCID: PMC6217820 DOI: 10.1016/j.immuni.2018.08.015] [Citation(s) in RCA: 676] [Impact Index Per Article: 96.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/13/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022]
Abstract
Systemic Lupus Erythematosus (SLE) is characterized by B cells lacking IgD and CD27 (double negative; DN). We show that DN cell expansions reflected a subset of CXCR5- CD11c+ cells (DN2) representing pre-plasma cells (PC). DN2 cells predominated in African-American patients with active disease and nephritis, anti-Smith and anti-RNA autoantibodies. They expressed a T-bet transcriptional network; increased Toll-like receptor-7 (TLR7); lacked the negative TLR regulator TRAF5; and were hyper-responsive to TLR7. DN2 cells shared with activated naive cells (aNAV), phenotypic and functional features, and similar transcriptomes. Their PC differentiation and autoantibody production was driven by TLR7 in an interleukin-21 (IL-21)-mediated fashion. An in vivo developmental link between aNAV, DN2 cells, and PC was demonstrated by clonal sharing. This study defines a distinct differentiation fate of autoreactive naive B cells into PC precursors with hyper-responsiveness to innate stimuli, as well as establishes prominence of extra-follicular B cell activation in SLE, and identifies therapeutic targets.
Collapse
Affiliation(s)
- Scott A Jenks
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Kevin S Cashman
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Esther Zumaquero
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Urko M Marigorta
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Aakash V Patel
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Xiaoqian Wang
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Deepak Tomar
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Zoe Simon
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Regina Bugrovsky
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Emily L Blalock
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | | | - Christopher M Tipton
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Chungwen Wei
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - S Sam Lim
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Michelle Petri
- Hopkins Lupus Center, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Timothy B Niewold
- Colton Center for Autoimmunity, NYU School of Medicine, New York, NY, USA
| | - Jennifer H Anolik
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy and Critical Care, Emory University, Atlanta, GA, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Frances E Lund
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
435
|
Jaime-Pérez JC, Aguilar-Calderón PE, Salazar-Cavazos L, Gómez-Almaguer D. Evans syndrome: clinical perspectives, biological insights and treatment modalities. J Blood Med 2018; 9:171-184. [PMID: 30349415 PMCID: PMC6190623 DOI: 10.2147/jbm.s176144] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Evans syndrome (ES) is a rare and chronic autoimmune disease characterized by autoimmune hemolytic anemia and immune thrombocytopenic purpura with a positive direct anti-human globulin test. It is classified as primary and secondary, with the frequency in patients with autoimmune hemolytic anemia being 37%–73%. It predominates in children, mainly due to primary immunodeficiencies or autoimmune lymphoproliferative syndrome. ES during pregnancy is associated with high fetal morbidity, including severe hemolysis and intracranial bleeding with neurological sequelae and death. The clinical presentation can include fatigue, pallor, jaundice and mucosal bleeding, with remissions and exacerbations during the person’s lifetime, and acute manifestations as catastrophic bleeding and massive hemolysis. Recent molecular theories explaining the physiopathology of ES include deficiencies of CTLA-4, LRBA, TPP2 and a decreased CD4/CD8 ratio. As in other autoimmune cytopenias, there is no established evidence-based treatment and steroids are the first-line therapy, with intravenous immunoglobulin administered as a life-saving resource in cases of severe immune thrombocytopenic purpura manifestations. Second-line treatment for refractory ES includes rituximab, mofetil mycophenolate, cyclosporine, vincristine, azathioprine, sirolimus and thrombopoietin receptor agonists. In cases unresponsive to immunosuppressive agents, hematopoietic stem cell transplantation has been successful, although it is necessary to consider its potential serious adverse effects. In conclusion, ES is a disease with a heterogeneous course that remains challenging to patients and physicians, with prospective clinical trials needed to explore potential targeted therapy to achieve an improved long-term response or even a cure.
Collapse
Affiliation(s)
- José Carlos Jaime-Pérez
- Department of Hematology, Internal Medicine Division, Dr José E González University Hospital, School of Medicine of the Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México,
| | - Patrizia Elva Aguilar-Calderón
- Department of Hematology, Internal Medicine Division, Dr José E González University Hospital, School of Medicine of the Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México,
| | - Lorena Salazar-Cavazos
- Department of Hematology, Internal Medicine Division, Dr José E González University Hospital, School of Medicine of the Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México,
| | - David Gómez-Almaguer
- Department of Hematology, Internal Medicine Division, Dr José E González University Hospital, School of Medicine of the Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México,
| |
Collapse
|
436
|
Gonzalez-Quintial R, Nguyen A, Kono DH, Oldstone MBA, Theofilopoulos AN, Baccala R. Lupus acceleration by a MAVS-activating RNA virus requires endosomal TLR signaling and host genetic predisposition. PLoS One 2018; 13:e0203118. [PMID: 30199535 PMCID: PMC6130858 DOI: 10.1371/journal.pone.0203118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022] Open
Abstract
Viruses have long been implicated in the pathogenesis of autoimmunity, yet their contribution remains circumstantial partly due to the lack of well-documented information on infections prior to autoimmune disease onset. Here, we used the lymphocytic choriomeningitis virus (LCMV) as a model to mechanistically dissect the impact of viral infection on lupus-like autoimmunity. Virus persistence strongly enhanced disease in mice with otherwise weak genetic predisposition but not in highly predisposed or non-autoimmune mice, indicating a synergistic interplay between genetic susceptibility and virus infection. Moreover, endosomal Toll-like receptors (TLRs) and plasmacytoid dendritic cells (pDCs) were both strictly required for disease acceleration, even though LCMV also induces strong TLR-independent type I interferon (IFN-I) production via RNA helicases and MAVS in conventional DCs. These results suggest that LCMV enhances systemic autoimmunity primarily by providing stimulatory nucleic acids for endosomal TLR engagement, whereas overstimulation of the MAVS-dependent cytosolic pathway in the absence of endosomal TLR signaling is insufficient for disease induction.
Collapse
Affiliation(s)
- Rosana Gonzalez-Quintial
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Anthony Nguyen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Dwight H. Kono
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael B. A. Oldstone
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Argyrios N. Theofilopoulos
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Roberto Baccala
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
437
|
Hartwell BL, Pickens CJ, Leon M, Northrup L, Christopher MA, Griffin JD, Martinez-Becerra F, Berkland C. Soluble antigen arrays disarm antigen-specific B cells to promote lasting immune tolerance in experimental autoimmune encephalomyelitis. J Autoimmun 2018; 93:76-88. [PMID: 30007842 PMCID: PMC6117839 DOI: 10.1016/j.jaut.2018.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
Autoreactive lymphocytes that escape central immune tolerance may be silenced via an endogenous peripheral tolerance mechanism known as anergy. Antigen-specific therapies capable of inducing anergy may restore patients with autoimmune diseases to a healthy phenotype while avoiding deleterious side effects associated with global immunosuppression. Inducing anergy in B cells may be a particularly potent intervention, as B cells can contribute to autoimmune diseases through multiple mechanisms and offer the potential for direct antigen-specific targeting through the B cell receptor (BCR). Our previous results suggested autoreactive B cells may be silenced by multivalent 'soluble antigen arrays' (SAgAs), which are polymer conjugates displaying multiple copies of autoantigen with or without a secondary peptide that blocks intracellular cell-adhesion molecule-1 (ICAM-1). Here, key therapeutic molecular properties of SAgAs were identified and linked to the immunological mechanism through comprehensive cellular and in vivo analyses. We determined non-hydrolyzable 'cSAgAs' displaying multivalent 'click'-conjugated antigen more potently suppressed experimental autoimmune encephalomyelitis (EAE) compared to hydrolyzable SAgAs capable of releasing conjugated antigen. cSAgAs restored a healthy phenotype in disease-specific antigen presenting cells (APCs) by inducing an anergic response in B cells and a subset of B cells called autoimmune-associated B cells (ABCs) that act as potent APCs in autoimmune disease. Accompanied by a cytokine response skewed towards a Th2/regulatory phenotype, this generated an environment of autoantigenic tolerance. By identifying key therapeutic molecular properties and an immunological mechanism that drives SAgA efficacy, this work guides the design of antigen-specific immunotherapies capable of inducing anergy.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/immunology
- B-Lymphocyte Subsets/drug effects
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/pathology
- Click Chemistry
- Clonal Anergy/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Hydrolysis
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacology
- Immunotherapy/methods
- Injections, Subcutaneous
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Mice
- Myelin Proteolipid Protein/administration & dosage
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Protein Array Analysis
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Spleen/immunology
- Spleen/pathology
- Th2 Cells/immunology
- Th2 Cells/pathology
Collapse
Affiliation(s)
- Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Chad J Pickens
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Martin Leon
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| | - Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Francisco Martinez-Becerra
- Immunology Core Laboratory of the Kansas Vaccine Institute, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, USA
| | - Cory Berkland
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA; Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, 1530 West 15th Street, Lawrence, KS 66045, USA.
| |
Collapse
|
438
|
Sadighi Akha AA. Aging and the immune system: An overview. J Immunol Methods 2018; 463:21-26. [PMID: 30114401 DOI: 10.1016/j.jim.2018.08.005] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 07/17/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022]
Abstract
The world is witnessing a rapid demographic shift towards an older population, a trend with major medical, social, economic and political implications. Aging is a multifaceted process, involving numerous molecular and cellular mechanisms in the context of different organ systems. A crucial component of aging is a set of functional and structural alterations in the immune system that can manifest as a decreased ability to fight infection, diminished response to vaccination, increased incidence of cancer, higher prevalence of autoimmunity and constitutive low-grade inflammation, among others. In addition to cell-intrinsic changes in both innate and adaptive immune cells, alterations in the stromal microenvironment in primary and secondary lymphoid organs play an important role in age-associated immune dysfunction. This article will provide a broad overview of these phenomena and point out some of their clinical and therapeutic implications.
Collapse
Affiliation(s)
- Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States.
| |
Collapse
|
439
|
IgG3 regulates tissue-like memory B cells in HIV-infected individuals. Nat Immunol 2018; 19:1001-1012. [PMID: 30104633 DOI: 10.1038/s41590-018-0180-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 06/28/2018] [Indexed: 01/10/2023]
Abstract
Immunoglobulin G3 (IgG3) has an uncertain role in the response to infection with and vaccination against human immunodeficiency virus (HIV). Here we describe a regulatory role for IgG3 in dampening the immune system-activating effects of chronic HIV viremia on B cells. Secreted IgG3 was bound to IgM-expressing B cells in vivo in HIV-infected chronically viremic individuals but not in early-viremic or aviremic individuals. Tissue-like memory (TLM) B cells, a population expanded by persistent HIV viremia, bound large amounts of IgG3. IgG3 induced clustering of B cell antigen receptors (BCRs) on the IgM+ B cells, which was mediated by direct interactions between soluble IgG3 and membrane IgM of the BCR (IgM-BCR). The inhibitory IgG receptor CD32b (FcγRIIb), complement component C1q and inflammatory biomarker CRP contributed to the binding of secreted IgG3 onto IgM-expressing B cells of HIV-infected individuals. Notably, IgG3-bound TLM B cells were refractory to IgM-BCR stimulation, thus demonstrating that IgG3 can regulate B cells during chronic activation of the immune system.
Collapse
|
440
|
Goltsev Y, Samusik N, Kennedy-Darling J, Bhate S, Hale M, Vazquez G, Black S, Nolan GP. Deep Profiling of Mouse Splenic Architecture with CODEX Multiplexed Imaging. Cell 2018; 174:968-981.e15. [PMID: 30078711 PMCID: PMC6086938 DOI: 10.1016/j.cell.2018.07.010] [Citation(s) in RCA: 905] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 02/05/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022]
Abstract
A highly multiplexed cytometric imaging approach, termed co-detection by indexing (CODEX), is used here to create multiplexed datasets of normal and lupus (MRL/lpr) murine spleens. CODEX iteratively visualizes antibody binding events using DNA barcodes, fluorescent dNTP analogs, and an in situ polymerization-based indexing procedure. An algorithmic pipeline for single-cell antigen quantification in tightly packed tissues was developed and used to overlay well-known morphological features with de novo characterization of lymphoid tissue architecture at a single-cell and cellular neighborhood levels. We observed an unexpected, profound impact of the cellular neighborhood on the expression of protein receptors on immune cells. By comparing normal murine spleen to spleens from animals with systemic autoimmune disease (MRL/lpr), extensive and previously uncharacterized splenic cell-interaction dynamics in the healthy versus diseased state was observed. The fidelity of multiplexed spatial cytometry demonstrated here allows for quantitative systemic characterization of tissue architecture in normal and clinically aberrant samples. Autoimmunity analyzed by multiplexed DNA-tagged antibody staining (CODEX) CODEX data reveal pairwise interactions and niches changing with disease First tier of neighbors significantly impacts marker expression in the index cells Changes in splenic morphology correlate with shifts in cell frequencies
Collapse
Affiliation(s)
- Yury Goltsev
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nikolay Samusik
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia Kennedy-Darling
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Salil Bhate
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew Hale
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gustavo Vazquez
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah Black
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
441
|
Nipper AJ, Smithey MJ, Shah RC, Canaday DH, Landay AL. Diminished antibody response to influenza vaccination is characterized by expansion of an age-associated B-cell population with low PAX5. Clin Immunol 2018; 193:80-87. [PMID: 29425852 PMCID: PMC9934856 DOI: 10.1016/j.clim.2018.02.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/19/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Individuals over the age of 65 comprise a substantial portion of the world population and become more susceptible to vaccine-preventable infections with age as vaccination response diminishes. The underlying reason for this impaired vaccine response in older individuals is not entirely clear. We evaluated potential differences in phenotypic and functional responses of B cells from healthy younger (22-45years) and older (64-95years) individuals that may associate with a diminished antibody response to influenza vaccination. We report that age is associated with expansion of atypical memory B cells (CD10-CD20+CD21-CD27-) and an age-associated B cell (ABC, CD21-T-bet+CD11c+) phenotype. Reduced expression of PAX5 was also seen in older individuals. Poor influenza-specific antibody production following vaccination was associated with low PAX5 expression and a distinct composition of the ABC compartment. Collectively, these findings demonstrate that the characteristics of the ABC populations of older individuals are associated with antibody production following influenza vaccination.
Collapse
Affiliation(s)
- Allison J. Nipper
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Megan J. Smithey
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Raj C. Shah
- Department of Family Medicine, Rush University Medical Center, Chicago, IL, USA,Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David H. Canaday
- Geriatric Research, Education, and Clinical Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA,Department of Internal Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alan L. Landay
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA,Corresponding author. Rush University Medical Center, 1735 West Harrison, Chicago, IL 60612, USA., (A.L. Landay)
| |
Collapse
|
442
|
Le Gallou S, Zhou Z, Thai LH, Fritzen R, de Los Aires AV, Mégret J, Yu P, Kitamura D, Bille E, Tros F, Nassif X, Charbit A, Weller S, Weill JC, Reynaud CA. A splenic IgM memory subset with antibacterial specificities is sustained from persistent mucosal responses. J Exp Med 2018; 215:2035-2053. [PMID: 29959173 PMCID: PMC6080908 DOI: 10.1084/jem.20180977] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
Le Gallou et al. use an AID fate-mapping model to identify an IgM memory population in the spleen of unimmunized mice, originating from persistent gut immune responses and endowed with cross-reactivity against bacteria. To what extent immune responses against the gut flora are compartmentalized within mucosal tissues in homeostatic conditions remains a much-debated issue. We describe here, based on an inducible AID fate-mapping mouse model, that systemic memory B cell subsets, including mainly IgM+ B cells in spleen, together with IgA+ plasma cells in spleen and bone marrow, are generated in mice in the absence of deliberate immunization. While the IgA component appears dependent on the gut flora, IgM memory B cells are still generated in germ-free mice, albeit to a reduced extent. Clonal relationships and renewal kinetics after anti-CD20 treatment reveal that this long-lasting splenic population is mainly sustained by output of B cell clones persisting in mucosal germinal centers. IgM-secreting hybridomas established from splenic IgM memory B cells showed reactivity against various bacterial isolates and endogenous retroviruses. Ongoing activation of B cells in gut-associated lymphoid tissues thus generates a diversified systemic compartment showing long-lasting clonal persistence and protective capacity against systemic bacterial infections.
Collapse
Affiliation(s)
- Simon Le Gallou
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Zhicheng Zhou
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lan-Huong Thai
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Remi Fritzen
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alba Verge de Los Aires
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérôme Mégret
- Flow Cytometry Core Facility, Structure Fédérative de Recherche Necker, Institut National de la Santé et de la Recherche Médicale US24-Centre National de la Recherche Scientifique UMS 3633, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Philipp Yu
- Institute of Immunology, Philipps-Universität Marburg, Marburg, Germany
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Emmanuelle Bille
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fabiola Tros
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xavier Nassif
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Charbit
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sandra Weller
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Claude Weill
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claude-Agnès Reynaud
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
443
|
Good-Jacobson KL. Strength in diversity: Phenotypic, functional, and molecular heterogeneity within the memory B cell repertoire. Immunol Rev 2018; 284:67-78. [DOI: 10.1111/imr.12663] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kim L. Good-Jacobson
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology; Biomedicine Discovery Institute, Monash University; Clayton Vic. Australia
| |
Collapse
|
444
|
Gubbels Bupp MR, Potluri T, Fink AL, Klein SL. The Confluence of Sex Hormones and Aging on Immunity. Front Immunol 2018; 9:1269. [PMID: 29915601 PMCID: PMC5994698 DOI: 10.3389/fimmu.2018.01269] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/22/2018] [Indexed: 12/17/2022] Open
Abstract
The immune systems of post-pubescent males and females differ significantly with profound consequences to health and disease. In many cases, sex-specific differences in the immune responses of young adults are also apparent in aged men and women. Moreover, as in young adults, aged women develop several late-adult onset autoimmune conditions more frequently than do men, while aged men continue to develop many cancers to a greater extent than aged women. However, sex differences in the immune systems of aged individuals have not been extensively investigated and data addressing the effectiveness of vaccinations and immunotherapies in aged men and women are scarce. In this review, we evaluate age- and sex hormone-related changes to innate and adaptive immunity, with consideration about how this impacts age- and sex-associated changes in the incidence and pathogenesis of autoimmunity and cancer as well as the efficacy of vaccination and cancer immunotherapy. We conclude that future preclinical and clinical studies should consider age and sex to better understand the ways in which these characteristics intersect with immune function and the resulting consequences for autoimmunity, cancer, and therapeutic interventions.
Collapse
Affiliation(s)
| | - Tanvi Potluri
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Ashley L Fink
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
445
|
Frasca D, Diaz A, Romero M, Thaller S, Blomberg BB. Secretion of autoimmune antibodies in the human subcutaneous adipose tissue. PLoS One 2018; 13:e0197472. [PMID: 29768501 PMCID: PMC5955545 DOI: 10.1371/journal.pone.0197472] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/02/2018] [Indexed: 11/30/2022] Open
Abstract
The adipose tissue (AT) contributes to systemic and B cell intrinsic inflammation, reduced B cell responses and secretion of autoimmune antibodies. In this study we show that adipocytes in the human obese subcutaneous AT (SAT) secrete several pro-inflammatory cytokines and chemokines, which contribute to the establishment and maintenance of local and systemic inflammation, and consequent suboptimal immune responses in obese individuals, as we have previously shown. We also show that pro-inflammatory chemokines recruit immune cells expressing the corresponding receptors to the SAT, where they also contribute to local and systemic inflammation, secreting additional pro-inflammatory mediators. Moreover, we show that the SAT generates autoimmune antibodies. During the development of obesity, reduced oxygen and consequent hypoxia and cell death lead to further release of pro-inflammatory cytokines, “self” protein antigens, cell-free DNA and lipids. All these stimulate class switch and the production of autoimmune IgG antibodies which have been described to be pathogenic. In addition to hypoxia, we have measured cell cytotoxicity and DNA damage mechanisms, which may also contribute to the release of “self” antigens in the SAT. All these processes are significantly elevated in the SAT as compared to the blood. We definitively found that fat-specific IgG antibodies are secreted by B cells in the SAT and that B cells express mRNA for the transcription factor T-bet and the membrane marker CD11c, both involved in the production of autoimmune IgG antibodies. Finally, the SAT also expresses RNA for cytokines known to promote Germinal Center formation, isotype class switch, and plasma cell differentiation. Our results show novel mechanisms for the generation of autoimmune antibody responses in the human SAT and allow the identification of new pathways to possibly manipulate in order to reduce systemic inflammation and autoantibody production in obese individuals.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- * E-mail:
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Seth Thaller
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Bonnie B. Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States of America
| |
Collapse
|
446
|
Tsay GJ, Zouali M. The Interplay Between Innate-Like B Cells and Other Cell Types in Autoimmunity. Front Immunol 2018; 9:1064. [PMID: 29868023 PMCID: PMC5964140 DOI: 10.3389/fimmu.2018.01064] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/27/2018] [Indexed: 12/12/2022] Open
Abstract
Studies performed in animal models and in humans indicate that the innate arm of the immune system provides an essential role in the initial protection against potential insults and in maintaining tolerance to self-antigens. In the B cell compartment, several subsets engage in both adaptive and innate functions. Whereas B cell subsets are recognized to play important roles in autoimmune diseases, understanding the intricacies of their effector functions remains challenging. In addition to B-1a cells and marginal zone B cells, the B cell compartment comprises other B cells with innate-like functions, including innate response activator B cells, T-bet positive B cells, natural killer-like B cells, IL-17-producing B cells, and human self-reactive VH4-34-expressing B cells. Herein, we summarize the functions of recently described B cell populations that can exert innate-like roles in both animal models and humans. We also highlight the importance of the cross talk between innate-like B cells and other adaptive and innate branches of the immune system in various autoimmune and inflammatory diseases. In as much as innate immunity seems to be important in resolving inflammation, it is possible that targeting certain innate-like B cell subsets could represent a novel therapeutic approach for inducing resolution of inflammation of autoimmune and inflammatory responses.
Collapse
Affiliation(s)
- Gregory J Tsay
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Moncef Zouali
- INSERM, U1132, Paris, France.,Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France
| |
Collapse
|
447
|
Yao Y, Huang W, Li X, Li X, Qian J, Han H, Sun H, An X, Lu L, Zhao H. Tespa1 Deficiency Dampens Thymus-Dependent B-Cell Activation and Attenuates Collagen-Induced Arthritis in Mice. Front Immunol 2018; 9:965. [PMID: 29867947 PMCID: PMC5960706 DOI: 10.3389/fimmu.2018.00965] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022] Open
Abstract
Thymocyte-expressed, positive selection-associated 1 (Tespa1) plays an important role in both T cell receptor (TCR)-driven thymocyte development and in the FcεRI-mediated activation of mast cells. Herein, we show that lack of Tespa1 does not impair B cell development but dampens the in vitro activation and proliferation of B cells induced by T cell-dependent (TD) antigens, significantly reduces serum antibody concentrations in vivo, and impairs germinal center formation in both aged and TD antigen-immunized mice. We also provide evidence that dysregulated signaling in Tespa1-deficient B cells may be linked to CD40-induced TRAF6 degradation, and subsequent effects on 1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2 (PLCγ2) phosphorylation, MAPK activation, and calcium influx. Furthermore, we demonstrate that Tespa1 plays a critical role in pathogenic B cells, since Tespa1-deficient chimeric mice showed a lower incidence and clinical disease severity of collagen-induced arthritis. Overall, our study demonstrates that Tespa1 is essential for TD B cell responses, and suggests an important role for Tespa1 during the development of autoimmune arthritis.
Collapse
Affiliation(s)
- Yunliang Yao
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Wei Huang
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Li
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Xiawei Li
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Qian
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Hui Han
- First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Hui Sun
- First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Xiangli An
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Linrong Lu
- School of Medicine, Institute of Immunology, Zhejiang University, Hangzhou, China
| | - Hongxing Zhao
- First Affiliated Hospital, Huzhou University, Huzhou, China
| |
Collapse
|
448
|
Loughran EA, Leonard AK, Hilliard TS, Phan RC, Yemc MG, Harper E, Sheedy E, Klymenko Y, Asem M, Liu Y, Yang J, Johnson J, Tarwater L, Shi Z, Leevy M, Ravosa MJ, Stack MS. Aging Increases Susceptibility to Ovarian Cancer Metastasis in Murine Allograft Models and Alters Immune Composition of Peritoneal Adipose Tissue. Neoplasia 2018; 20:621-631. [PMID: 29754071 PMCID: PMC5994778 DOI: 10.1016/j.neo.2018.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/20/2018] [Accepted: 03/26/2018] [Indexed: 11/30/2022] Open
Abstract
Ovarian cancer, the most deadly gynecological malignancy in U.S. women, metastasizes uniquely, spreading through the peritoneal cavity and often generating widespread metastatic sites before diagnosis. The vast majority of ovarian cancer cases occur in women over 40 and the median age at diagnosis is 63. Additionally, elderly women receive poorer prognoses when diagnosed with ovarian cancer. Despite age being a significant risk factor for the development of this cancer, there are little published data which address the impact of aging on ovarian cancer metastasis. Here we report that the aged host is more susceptible to metastatic success using two murine syngeneic allograft models of ovarian cancer metastasis. This age-related increase in metastatic tumor burden corresponds with an increase in tumor infiltrating lymphocytes (TILs) in tumor-bearing mice and alteration of B cell-related pathways in gonadal adipose tissue. Based on this work, further studies elucidating the status of B cell TILs in mouse models of metastasis and human tumors in the context of aging are warranted.
Collapse
Affiliation(s)
- Elizabeth A Loughran
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Annemarie K Leonard
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Tyvette S Hilliard
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Ryan C Phan
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Madeleine G Yemc
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Elizabeth Harper
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Emma Sheedy
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Yuliya Klymenko
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
| | - Marwa Asem
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Yueying Liu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Jing Yang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Jeff Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Laura Tarwater
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Zonggao Shi
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Matthew Leevy
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
| | - Matthew J Ravosa
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN; Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN; Department of Anthropology, University of Notre Dame, Notre Dame, IN
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN.
| |
Collapse
|
449
|
Hypomorphic Rag1 mutations alter the preimmune repertoire at early stages of lymphoid development. Blood 2018; 132:281-292. [PMID: 29743177 DOI: 10.1182/blood-2017-12-820985] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Hypomorphic RAG1 mutations allowing residual T- and B-cell development have been found in patients presenting with delayed-onset combined immune deficiency with granulomas and/or autoimmunity (CID-G/AI) and abnormalities of the peripheral T- and B-cell repertoire. To examine how hypomorphic Rag1 mutations affect the earliest stages of lymphocyte development, we used CRISPR/Cas9 to generate mouse models with mutations equivalent to those found in patients with CID-G/AI. Immunological characterization showed partial development of T and B lymphocytes, with persistence of naïve cells and preserved serum immunoglobulin but impaired antibody responses and presence of autoantibodies, thereby recapitulating the phenotype seen in patients with CID-G/AI. By using high-throughput sequencing, we identified marked skewing of Igh V and Trb V gene usage in early progenitors, with a bias for productive Igh and Trb rearrangements after selection occurred and increased apoptosis of B-cell progenitors. Rearrangement at the Igk locus was impaired, and polyreactive immunoglobulin M antibodies were detected. This study provides novel insights into how hypomorphic Rag1 mutations alter the primary repertoire of T and B cells, setting the stage for immune dysregulation frequently seen in patients.
Collapse
|
450
|
CD11c-Specific Deletion Reveals CREB as a Critical Regulator of DC Function during the Germinal Center Response. J Immunol Res 2018; 2018:8947230. [PMID: 29854847 PMCID: PMC5964551 DOI: 10.1155/2018/8947230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/24/2018] [Accepted: 03/11/2018] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells (DCs) are crucial for the balance between immune response and tolerance, but the molecular mechanism regulating development, differentiation, and homeostasis are poorly understood. The transcriptional activator CREB is involved in regulating different cells of the innate and adaptive immune system and is a transcriptional regulator of development, survival, activation, or proliferation in macrophages, dendritic cells, B cells, and T cells. To directly examine the role of CREB in the regulation of DCs, the CREB gene was targeted for deletion with a CD11c-cre transgene. The deletion of CREB in CD11c+ cells did not involve any developmental or systemic defects within DC populations. However, CREB deficiency in CD11c+ cells reduced germinal center (GC) B cells in steady state, and immunization with NP-CGG resulted in a reduced formation of GCs, paralleled by the reduced production of IgGs in sera of immunized mice. In conclusion, we demonstrate that CREB expression in CD11c+ cells enhances germinal center responses, most likely by altering DC function, which might have implications for autoimmune diseases that are associated with dysregulated GC responses.
Collapse
|