1
|
McCaleb MR, Miranda AM, Khammash HA, Torres RM, Pelanda R. Regulation of Foxo1 expression is critical for central B cell tolerance and allelic exclusion. Cell Rep 2024; 43:114283. [PMID: 38796853 PMCID: PMC11246624 DOI: 10.1016/j.celrep.2024.114283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Resolving the molecular mechanisms of central B cell tolerance might unveil strategies that prevent autoimmunity. Here, using a mouse model of central B cell tolerance in which Forkhead box protein O1 (Foxo1) is either deleted or over-expressed in B cells, we show that deleting Foxo1 blocks receptor editing, curtails clonal deletion, and decreases CXCR4 expression, allowing high-avidity autoreactive B cells to emigrate to the periphery whereby they mature but remain anergic and short lived. Conversely, expression of degradation-resistant Foxo1 promotes receptor editing in the absence of self-antigen but leads to allelic inclusion. Foxo1 over-expression also restores tolerance in autoreactive B cells harboring active PI3K, revealing opposing roles of Foxo1 and PI3K in B cell selection. Overall, we show that the transcription factor Foxo1 is a major gatekeeper of central B cell tolerance and that PI3K drives positive selection of immature B cells and establishes allelic exclusion by suppressing Foxo1.
Collapse
Affiliation(s)
- Megan R McCaleb
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anjelica M Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hadeel A Khammash
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Pelanda R, Greaves SA, Alves da Costa T, Cedrone LM, Campbell ML, Torres RM. B-cell intrinsic and extrinsic signals that regulate central tolerance of mouse and human B cells. Immunol Rev 2022; 307:12-26. [PMID: 34997597 PMCID: PMC8986553 DOI: 10.1111/imr.13062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 12/20/2022]
Abstract
The random recombination of immunoglobulin V(D)J gene segments produces unique IgM antibodies that serve as the antigen receptor for each developing B cell. Hence, the newly formed B cell repertoire is comprised of a variety of specificities that display a range of reactivity with self-antigens. Newly generated IgM+ immature B cells that are non-autoreactive or that bind self-antigen with low avidity are licensed to leave the bone marrow with their intact antigen receptor and to travel via the blood to the peripheral lymphoid tissue for further selection and maturation. In contrast, clones with medium to high avidity for self-antigen remain within the marrow and undergo central tolerance, a process that revises their antigen receptor or eliminates the autoreactive B cell altogether. Thus, central B cell tolerance is critical for reducing the autoreactive capacity and avidity for self-antigen of our circulating B cell repertoire. Bone marrow cultures and mouse models have been instrumental for understanding the mechanisms that regulate the selection of bone marrow B cells. Here, we review recent studies that have shed new light on the contribution of the ERK, PI3K, and CXCR4 signaling pathways in the selection of mouse and human immature B cells that either bind or do not bind self-antigen.
Collapse
Affiliation(s)
- Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Sarah A Greaves
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Thiago Alves da Costa
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Lena M Cedrone
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Margaret L Campbell
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
3
|
Cashman KS, Jenks SA, Woodruff MC, Tomar D, Tipton CM, Scharer CD, Lee EH, Boss JM, Sanz I. Understanding and measuring human B-cell tolerance and its breakdown in autoimmune disease. Immunol Rev 2019; 292:76-89. [PMID: 31755562 PMCID: PMC6935423 DOI: 10.1111/imr.12820] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
The maintenance of immunological tolerance of B lymphocytes is a complex and critical process that must be implemented as to avoid the detrimental development of autoreactivity and possible autoimmunity. Murine models have been invaluable to elucidate many of the key components in B-cell tolerance; however, translation to human homeostatic and pathogenic immune states can be difficult to assess. Functional autoreactive, flow cytometric, and single-cell cloning assays have proven to be critical in deciphering breaks in B-cell tolerance within autoimmunity; however, newer approaches to assess human B-cell tolerance may prove to be vital in the further exploration of underlying tolerance defects. In this review, we supply a comprehensive overview of human immune tolerance checkpoints with associated mechanisms of enforcement, and highlight current and future methodologies which are likely to benefit future studies into the mechanisms that become defective in human autoimmune conditions.
Collapse
Affiliation(s)
- Kevin S. Cashman
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Scott A. Jenks
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Matthew C. Woodruff
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Deepak Tomar
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Christopher M. Tipton
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Christopher D. Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - 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
| | - Jeremy M. Boss
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Greaves SA, Peterson JN, Strauch P, Torres RM, Pelanda R. Active PI3K abrogates central tolerance in high-avidity autoreactive B cells. J Exp Med 2019; 216:1135-1153. [PMID: 30948496 PMCID: PMC6504226 DOI: 10.1084/jem.20181652] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/23/2019] [Accepted: 03/22/2019] [Indexed: 01/02/2023] Open
Abstract
High-avidity autoreactive B cells are typically removed by central tolerance mechanisms in the bone marrow. Greaves et al. demonstrate that B cell–intrinsic expression of active PI3Kα prevents central tolerance and effectively promotes differentiation and activation of high-avidity autoreactive B cells in the periphery. Autoreactive B cells that bind self-antigen with high avidity in the bone marrow undergo mechanisms of central tolerance that prevent their entry into the peripheral B cell population. These mechanisms are breached in many autoimmune patients, increasing their risk of B cell–mediated autoimmune diseases. Resolving the molecular pathways that can break central B cell tolerance could therefore provide avenues to diminish autoimmunity. Here, we show that B cell–intrinsic expression of a constitutively active form of PI3K-P110α by high-avidity autoreactive B cells of mice completely abrogates central B cell tolerance and further promotes these cells to escape from the bone marrow, differentiate in peripheral tissue, and undergo activation in response to self-antigen. Upon stimulation with T cell help factors, these B cells secrete antibodies in vitro but remain unable to secrete autoantibodies in vivo. Overall, our data demonstrate that activation of the PI3K pathway leads high-avidity autoreactive B cells to breach central, but not late, stages of peripheral tolerance.
Collapse
Affiliation(s)
- Sarah A Greaves
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Jacob N Peterson
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Pamela Strauch
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO.,Department of Biomedical Research, National Jewish Health, Denver, CO
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO .,Department of Biomedical Research, National Jewish Health, Denver, CO
| |
Collapse
|
5
|
Lindeman I, Stubbington MJT. Antigen Receptor Sequence Reconstruction and Clonality Inference from scRNA-Seq Data. Methods Mol Biol 2019; 1935:223-249. [PMID: 30758830 DOI: 10.1007/978-1-4939-9057-3_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this chapter, we describe TraCeR and BraCeR, our computational tools for reconstruction of paired full-length antigen receptor sequences and clonality inference from single-cell RNA-seq (scRNA-seq) data. In brief, TraCeR reconstructs T-cell receptor (TCR) sequences from scRNA-seq data by extracting sequencing reads derived from TCRs by aligning the reads from each cell against synthetic TCR sequences. TCR-derived reads are then assembled into full-length recombined TCR sequences. BraCeR builds on the TraCeR pipeline and accounts for somatic hypermutations (SHM) and isotype switching. Here we discuss experimental design, use of the tools, and interpretation of the results.
Collapse
Affiliation(s)
- Ida Lindeman
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- KG Jebsen Coeliac Disease Research Centre and Department of Immunology, University of Oslo, Oslo, Norway
| | | |
Collapse
|
6
|
Innate and adaptive signals enhance differentiation and expansion of dual-antibody autoreactive B cells in lupus. Nat Commun 2018; 9:3973. [PMID: 30266981 PMCID: PMC6162205 DOI: 10.1038/s41467-018-06293-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Autoreactive B cells have a major function in autoimmunity. A small subset of B cells expressing two distinct B-cell-antigen-receptors (B2R cells) is elevated in many patients with systematic lupus erythematosus (SLE) and in the MRL(/lpr) mouse model of lupus, and is often autoreactive. Here we show, using RNAseq and in vitro and in vivo analyses, signals that are required for promoting B2R cell numbers and effector function in autoimmune mice. Compared with conventional B cells, B2R cells are more responsive to Toll-like receptor 7/9 and type I/II interferon treatment, display higher levels of MHCII and co-receptors, and depend on IL-21 for their homeostasis; moreover they expand better upon T cell-dependent antigen stimulation, and mount a more robust memory response, which are characteristics essential for enhanced (auto)immune responses. Our findings thus provide insights on the stimuli for the expansion of an autoreactive B cell subset that may contribute to the etiology of SLE.
Collapse
|
7
|
Greaves SA, Peterson JN, Torres RM, Pelanda R. Activation of the MEK-ERK Pathway Is Necessary but Not Sufficient for Breaking Central B Cell Tolerance. Front Immunol 2018; 9:707. [PMID: 29686680 PMCID: PMC5900439 DOI: 10.3389/fimmu.2018.00707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/22/2018] [Indexed: 01/12/2023] Open
Abstract
Newly generated bone marrow B cells are positively selected into the peripheral lymphoid tissue only when they express a B cell receptor (BCR) that is nonautoreactive or one that binds self-antigen with only minimal avidity. This positive selection process, moreover, is critically contingent on the ligand-independent tonic signals transduced by the BCR. We have previously shown that when autoreactive B cells express an active form of the rat sarcoma (RAS) oncogene, they upregulate the receptor for the B cell activating factor (BAFFR) and undergo differentiation in vitro and positive selection into the spleen in vivo, overcoming central tolerance. Based on the in vitro use of pharmacologic inhibitors, we further showed that this cell differentiation process is critically dependent on the activation of the mitogen-activated protein kinase kinase pathway MEK (MAPKK)-extracellular signal-regulated kinase (ERK), which is downstream of RAS. Here, we next investigated if activation of ERK is not only necessary but also sufficient to break central B cell tolerance and induce differentiation of autoreactive B cells in vitro and in vivo. Our results demonstrate that activation of ERK is critical for upregulating BAFFR and overcoming suboptimal levels of tonic BCR signals or low amounts of antigen-induced BCR signals during in vitro B cell differentiation. However, direct activation of ERK does not lead high avidity autoreactive B cells to increase BAFFR levels and undergo positive selection and differentiation in vivo. B cell-specific MEK-ERK activation in mice is also unable to lead to autoantibody secretion, and this in spite of a general increase of serum immunoglobulin levels. These findings indicate that additional pathways downstream of RAS are required for high avidity autoreactive B cells to break central and/or peripheral tolerance.
Collapse
Affiliation(s)
- Sarah A Greaves
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Jacob N Peterson
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| |
Collapse
|
8
|
Clark AG, Worni-Schudel IM, Korte FM, Foster MH. A murine Ig light chain transgene reveals IGKV3 gene contributions to anti-collagen types IV and II specificities. Mol Immunol 2017; 91:49-56. [PMID: 28886586 DOI: 10.1016/j.molimm.2017.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/30/2017] [Accepted: 08/16/2017] [Indexed: 01/06/2023]
Abstract
A subset of autoimmune diseases result from autoantibodies targeting epitopes on matrix collagen. The most extensively studied are anti-glomerular basement membrane glomerulonephritis (or its systemic counterpart Goodpasture's disease) that destroys kidneys and lungs, and rheumatoid arthritis that leads to disabling arthritis. Autoantibodies in these disorders bind evolutionarily conserved conformational epitopes on the noncollagenous domain 1 (NC1) of the alpha3 chain of type IV [alpha3(IV)NC1] collagen in glomerular and alveolar basement membranes, and on native or citrullinated type II collagen (CII) in joint cartilage, respectively. The genetic origins of pathogenic anti-collagen B cells in these diseases is unknown, but observations from murine models raise the possibility that they overlap despite distinct in vivo immunopathologies. Monoclonal autoantibodies isolated from mice immunized with alpha3(IV)NC1 collagen or CII show a biased use of Ig light chains (LC) encoded by genes of the IGKV3 subgroup (previously Vk21 family), paired with diverse Ig heavy chains. To further explore this relationship and determine if a single murine IGKV3 LC independently predisposes to both anti-collagen responses, we generated a novel transgenic (Tg) C57BL/6 mouse that expresses a productively rearranged IGKV3-encoded LC, termed mLCV3-Tg, in conjunction with endogenously rearranged Ig heavy chains. Tg mice are also genetically deficient in endogenous kappa chains to permit tracking of the mLCV3 transgene. We show that mLCV3-Tg mice are susceptible to humoral autoimmunity against both collagen chains. Anti-alpha3(IV)NC1 collagen, but not anti-CII, mLCV3-encoded Ig are detected in serum of unmanipulated Tg mice, while Toll-like receptor ligands induce secretion of mLCV3-Tg autoantibodies of both collagen specificities from splenocytes ex vivo. This indicates developmental survival of mLCV3-Tg B cells reactive with each antigen, and is consistent with production of the two anti-collagen autoIg from distinct B cell populations. Reduced B cell numbers, low serum Ig kappa levels, low cell surface Ig kappa density, and abundant endogenous lambda chain expression suggest that subsets of IGKV3-encoded B cells are regulated in vivo by mechanisms that include deletion, anergy, and LC editing. These results support the notion that murine IGKV3 LCs contribute structural fitness to antigen binding sites that support diverse anti-collagen autoimmune responses, that these responses are regulated in vivo, and that these cells can nonetheless readily escape immune regulation.
Collapse
Affiliation(s)
- Amy G Clark
- Department of Medicine, Duke University Health System, Durham, NC, USA; Durham VA Medical Center, Durham, NC, USA.
| | - Inge M Worni-Schudel
- Department of Medicine, Duke University Health System, Durham, NC, USA; Durham VA Medical Center, Durham, NC, USA.
| | - Francesca M Korte
- Department of Medicine, Duke University Health System, Durham, NC, USA.
| | - Mary H Foster
- Department of Medicine, Duke University Health System, Durham, NC, USA; Durham VA Medical Center, Durham, NC, USA.
| |
Collapse
|
9
|
Spohn G, Arenas-Ramirez N, Bouchaud G, Boyman O. Endogenous polyclonal anti–IL-1 antibody responses potentiate IL-1 activity during pathogenic inflammation. J Allergy Clin Immunol 2017; 139:1957-1965.e3. [DOI: 10.1016/j.jaci.2016.09.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 08/27/2016] [Accepted: 09/06/2016] [Indexed: 10/20/2022]
|
10
|
DeKosky BJ, Lungu OI, Park D, Johnson EL, Charab W, Chrysostomou C, Kuroda D, Ellington AD, Ippolito GC, Gray JJ, Georgiou G. Large-scale sequence and structural comparisons of human naive and antigen-experienced antibody repertoires. Proc Natl Acad Sci U S A 2016; 113:E2636-45. [PMID: 27114511 PMCID: PMC4868480 DOI: 10.1073/pnas.1525510113] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Elucidating how antigen exposure and selection shape the human antibody repertoire is fundamental to our understanding of B-cell immunity. We sequenced the paired heavy- and light-chain variable regions (VH and VL, respectively) from large populations of single B cells combined with computational modeling of antibody structures to evaluate sequence and structural features of human antibody repertoires at unprecedented depth. Analysis of a dataset comprising 55,000 antibody clusters from CD19(+)CD20(+)CD27(-) IgM-naive B cells, >120,000 antibody clusters from CD19(+)CD20(+)CD27(+) antigen-experienced B cells, and >2,000 RosettaAntibody-predicted structural models across three healthy donors led to a number of key findings: (i) VH and VL gene sequences pair in a combinatorial fashion without detectable pairing restrictions at the population level; (ii) certain VH:VL gene pairs were significantly enriched or depleted in the antigen-experienced repertoire relative to the naive repertoire; (iii) antigen selection increased antibody paratope net charge and solvent-accessible surface area; and (iv) public heavy-chain third complementarity-determining region (CDR-H3) antibodies in the antigen-experienced repertoire showed signs of convergent paired light-chain genetic signatures, including shared light-chain third complementarity-determining region (CDR-L3) amino acid sequences and/or Vκ,λ-Jκ,λ genes. The data reported here address several longstanding questions regarding antibody repertoire selection and development and provide a benchmark for future repertoire-scale analyses of antibody responses to vaccination and disease.
Collapse
Affiliation(s)
- Brandon J DeKosky
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712
| | - Oana I Lungu
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712; Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Daechan Park
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712; Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Erik L Johnson
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712
| | - Wissam Charab
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712
| | | | - Daisuke Kuroda
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Andrew D Ellington
- Center for Systems and Synthetic Biology University of Texas at Austin, Austin, TX 78712
| | - Gregory C Ippolito
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712; Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712; Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712
| |
Collapse
|
11
|
Lang J, Ota T, Kelly M, Strauch P, Freed BM, Torres RM, Nemazee D, Pelanda R. Receptor editing and genetic variability in human autoreactive B cells. J Exp Med 2015; 213:93-108. [PMID: 26694971 PMCID: PMC4710202 DOI: 10.1084/jem.20151039] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/23/2015] [Indexed: 12/11/2022] Open
Abstract
Lang et al. show in a humanized mouse model that human B cells undergo central tolerance via a combination of receptor editing and clonal deletion. The mechanisms by which B cells undergo tolerance, such as receptor editing, clonal deletion, and anergy, have been established in mice. However, corroborating these mechanisms in humans remains challenging. To study how autoreactive human B cells undergo tolerance, we developed a novel humanized mouse model. Mice expressing an anti–human Igκ membrane protein to serve as a ubiquitous neo self-antigen (Ag) were transplanted with a human immune system. By following the fate of self-reactive human κ+ B cells relative to nonautoreactive λ+ cells, we show that tolerance of human B cells occurs at the first site of self-Ag encounter, the bone marrow, via a combination of receptor editing and clonal deletion. Moreover, the amount of available self-Ag and the genetics of the cord blood donor dictate the levels of central tolerance and autoreactive B cells in the periphery. Thus, this model can be useful for studying specific mechanisms of human B cell tolerance and to reveal differences in the extent of this process among human populations.
Collapse
Affiliation(s)
- Julie Lang
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Takayuki Ota
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Margot Kelly
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Pamela Strauch
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Brian M Freed
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Division of Allergy and Clinical Immunology, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - David Nemazee
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045 Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| |
Collapse
|
12
|
Bonami RH, Thomas JW. Targeting Anti-Insulin B Cell Receptors Improves Receptor Editing in Type 1 Diabetes-Prone Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:4730-41. [PMID: 26432895 DOI: 10.4049/jimmunol.1500438] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
Autoreactive B lymphocytes that commonly arise in the developing repertoire can be salvaged by receptor editing, a central tolerance mechanism that alters BCR specificity through continued L chain rearrangement. It is unknown whether autoantigens with weak cross-linking potential, such as insulin, elicit receptor editing, or whether this process is dysregulated in related autoimmunity. To resolve these issues, we developed an editing-competent model in which anti-insulin Vκ125 was targeted to the Igκ locus and paired with anti-insulin VH125Tg. Physiologic, circulating insulin increased RAG-2 expression and was associated with BCR replacement that eliminated autoantigen recognition in a proportion of developing anti-insulin B lymphocytes. The proportion of anti-insulin B cells that underwent receptor editing was reduced in the type 1 diabetes-prone NOD strain relative to a nonautoimmune strain. Resistance to editing was associated with increased surface IgM expression on immature (but not transitional or mature) anti-insulin B cells in the NOD strain. The actions of mAb123 on central tolerance were also investigated, because selective targeting of insulin-occupied BCR by mAb123 eliminates anti-insulin B lymphocytes and prevents type 1 diabetes. Autoantigen targeting by mAb123 increased RAG-2 expression and dramatically enhanced BCR replacement in newly developed B lymphocytes. Administering F(ab')2123 induced IgM downregulation and reduced the frequency of anti-insulin B lymphocytes within the polyclonal repertoire of VH125Tg/NOD mice, suggesting enhanced central tolerance by direct BCR interaction. These findings indicate that weak or faulty checkpoints for central tolerance can be overcome by autoantigen-specific immunomodulatory therapy.
Collapse
Affiliation(s)
- Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232; and
| | - James W Thomas
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232; and Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN 37232
| |
Collapse
|
13
|
Fraser LD, Zhao Y, Lutalo PMK, D'Cruz DP, Cason J, Silva JS, Dunn‐Walters DK, Nayar S, Cope AP, Spencer J. Immunoglobulin light chain allelic inclusion in systemic lupus erythematosus. Eur J Immunol 2015; 45:2409-19. [PMID: 26036683 PMCID: PMC5102633 DOI: 10.1002/eji.201545599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/23/2015] [Accepted: 05/29/2015] [Indexed: 11/23/2022]
Abstract
The principles of allelic exclusion state that each B cell expresses a single light and heavy chain pair. Here, we show that B cells with both kappa and lambda light chains (Igκ and Igλ) are enriched in some patients with the systemic autoimmune disease systemic lupus erythematosus (SLE), but not in the systemic autoimmune disease control granulomatosis with polyangiitis. Detection of dual Igκ and Igλ expression by flow cytometry could not be abolished by acid washing or by DNAse treatment to remove any bound polyclonal antibody or complexes, and was retained after two days in culture. Both surface and intracytoplasmic dual light chain expression was evident by flow cytometry and confocal microscopy. We observed reduced frequency of rearrangements of the kappa-deleting element (KDE) in SLE and an inverse correlation between the frequency of KDE rearrangement and the frequency of dual light chain expressing B cells. We propose that dual expression of Igκ and Igλ by a single B cell may occur in some patients with SLE when this may be a consequence of reduced activity of the KDE.
Collapse
Affiliation(s)
- Louise D. Fraser
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| | - Yuan Zhao
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| | | | - David P. D'Cruz
- Louise Coote Lupus Unit Guy's and St Thomas’ NHS TrustLondonUK
| | - John Cason
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| | - Joselli S. Silva
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| | | | - Saba Nayar
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| | - Andrew P. Cope
- Academic Department of RheumatologyKing's College LondonLondonUK
| | - Jo Spencer
- Programme in Infection and ImmunobiologyKing's College LondonLondonUK
| |
Collapse
|
14
|
Xing Y, Ji Q, Lin Y, Fu M, Gao J, Zhang P, Hu X, Feng L, Liu Y, Han H, Li W. Positive selection of natural poly-reactive B cells in the periphery occurs independent of heavy chain allelic inclusion. PLoS One 2015; 10:e0125747. [PMID: 25993514 PMCID: PMC4437983 DOI: 10.1371/journal.pone.0125747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/25/2015] [Indexed: 11/28/2022] Open
Abstract
Natural autoreactive B cells are important mediators of autoimmune diseases. Receptor editing is known to play an important role in both central and peripheral B cell tolerance. However, the role of allelic inclusion in the development of natural autoreactive B cells is not clear. Previously, we generated μ chain (TgVH3B4I) and μ/κ chains (TgVH/L3B4) transgenic mice using transgene derived from the 3B4 hybridoma, which produce poly-reactive natural autoantibodies. In this study, we demonstrate that a considerable population of B cells edited their B cells receptors (BCRs) via light chain or heavy chain allelic inclusion during their development in TgVH3B4I mice. Additionally, allelic inclusion occurred more frequently in the periphery and promoted the differentiation of B cells into marginal zone or B-1a cells in TgVH3B4I mice. B cells from TgVH/L3B4 mice expressing the intact transgenic 3B4 BCR without receptor editing secreted poly-reactive 3B4 antibody. Interestingly, however, B cell that underwent allelic inclusion in TgVH3B4I mice also produced poly-reactive autoantibodies in vivo and in vitro. Our findings suggest that receptor editing plays a minor role in the positive selection of B cells expressing natural poly-reactive BCRs, which can be positively selected through heavy chain allelic inclusion to retain their poly-reactivity in the periphery.
Collapse
Affiliation(s)
- Ying Xing
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Endocrinology and Metabolism Disease, Xijing Hospital, Fourth Military Medical University, Xi'an, China; State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Qiuhe Ji
- Department of Endocrinology and Metabolism Disease, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Lin
- Department of Otolaryngology Head and Neck surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jixin Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Zhang
- Department of Otolaryngology Head and Neck surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xingbin Hu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Lei Feng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Yufeng Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Wei Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Detanico T, Guo W, Wysocki LJ. Predominant role for activation-induced cytidine deaminase in generating IgG anti-nucleosomal antibodies of murine SLE. J Autoimmun 2015; 58:67-77. [PMID: 25634361 DOI: 10.1016/j.jaut.2015.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/08/2015] [Accepted: 01/12/2015] [Indexed: 12/11/2022]
Abstract
Serum IgG anti-nuclear antibodies (ANA) directed to complexes of DNA and histones are a hallmark of systemic lupus erythematosus (SLE) and reflect a failure in lymphocyte self-tolerance. A prior study utilizing spontaneously autoimmune B6.Nba2 mice deficient in terminal deoxynucleotidyl transferase (TdT) and with heterozygous deficiencies in Jh and Igk loci underscored the importance of somatic hypermutation (SHM) as a major generator of SLE-associated ANA. This interpretation had to be qualified because of severely limited opportunities for receptor editing and restricted VHCDR3 diversity. Therefore, we performed the converse study using mice that carried functional Tdt genes and wild type Jh and Igk loci but that could not undergo SHM. Analyses of ANA and ANA-producing hybridomas from B6.Nba2 Aicda(-/-) mice revealed that few animals produced high titers of the prototypical ANA directed to complexes of histones and DNA, that this response was delayed and that those cells that did produce such antibody exhibited limited clonal expansion, unusual Jk use and only infrequent dual receptor expression. This, together with the additional finding of an intrinsic propensity for SHM to generate Arg codons selectively in CDRs, reinforce the view that most IgG autoimmune clones producing prototypical anti-nucleosome antibodies in wild type mice are created by SHM.
Collapse
Affiliation(s)
- Thiago Detanico
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, CO, USA
| | - Wenzhong Guo
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, CO, USA
| | - Lawrence J Wysocki
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, CO, USA.
| |
Collapse
|
16
|
de Almeida CR, Hendriks RW, Stadhouders R. Dynamic Control of Long-Range Genomic Interactions at the Immunoglobulin κ Light-Chain Locus. Adv Immunol 2015; 128:183-271. [DOI: 10.1016/bs.ai.2015.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
In-depth determination and analysis of the human paired heavy- and light-chain antibody repertoire. Nat Med 2014; 21:86-91. [PMID: 25501908 DOI: 10.1038/nm.3743] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 10/08/2014] [Indexed: 12/19/2022]
Abstract
High-throughput immune repertoire sequencing has emerged as a critical step in the understanding of adaptive responses following infection or vaccination or in autoimmunity. However, determination of native antibody variable heavy-light pairs (VH-VL pairs) remains a major challenge, and no technologies exist to adequately interrogate the >1 × 10(6) B cells in typical specimens. We developed a low-cost, single-cell, emulsion-based technology for sequencing antibody VH-VL repertoires from >2 × 10(6) B cells per experiment with demonstrated pairing precision >97%. A simple flow-focusing apparatus was used to sequester single B cells into emulsion droplets containing lysis buffer and magnetic beads for mRNA capture; subsequent emulsion RT-PCR generated VH-VL amplicons for next-generation sequencing. Massive VH-VL repertoire analyses of three human donors provided new immunological insights including (i) the identity, frequency and pairing propensity of shared, or 'public', VL genes, (ii) the detection of allelic inclusion (an implicated autoimmune mechanism) in healthy individuals and (iii) the occurrence of antibodies with features, in terms of gene usage and CDR3 length, associated with broadly neutralizing antibodies to rapidly evolving viruses such as HIV-1 and influenza.
Collapse
|
18
|
Activation of Ras overcomes B-cell tolerance to promote differentiation of autoreactive B cells and production of autoantibodies. Proc Natl Acad Sci U S A 2014; 111:E2797-806. [PMID: 24958853 DOI: 10.1073/pnas.1402159111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Newly generated immature B cells are selected to enter the peripheral mature B-cell pool only if they do not bind (or bind limited amount of) self-antigen. We previously suggested that this selection relies on basal extracellular signal-regulated kinase (Erk) activation mediated by tonic B-cell antigen receptor (BCR) signaling and that this signal can be replaced by an active rat sarcoma (Ras), which are small GTPase proteins. In this study we compared the activity of Ras and Erk in nonautoreactive and autoreactive immature B cells and investigated whether activation of Ras can break tolerance. Our results demonstrate lower levels of active Erk and Ras in autoreactive immature B cells, although this is evident only when these cells display medium/high avidity for self-antigen. Basal activation of Erk in immature B cells is proportional to surface IgM and dependent on sarcoma family kinases, whereas it is independent of B-cell activating factor, IFN, and Toll-like receptor signaling. Ectopic expression of the constitutively active mutant Ras form N-RasD12 in autoreactive cells raises active Erk, halts receptor editing via PI3 kinase, and promotes differentiation via Erk, breaking central tolerance. Moreover, when B cells coexpress autoreactive and nonautoreactive BCRs, N-RasD12 leads also to a break in peripheral tolerance with the production of autoantibodies. Our findings indicate that in immature B cells, basal activation of Ras and Erk are controlled by tonic BCR signaling, and that positive changes in Ras activity can lead to a break in both central and peripheral B-cell tolerance.
Collapse
|
19
|
Pelanda R. Dual immunoglobulin light chain B cells: Trojan horses of autoimmunity? Curr Opin Immunol 2014; 27:53-9. [PMID: 24549093 DOI: 10.1016/j.coi.2014.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/17/2014] [Accepted: 01/23/2014] [Indexed: 10/25/2022]
Abstract
Receptor editing, a major mechanism of B cell tolerance, can also lead to allelic inclusion at the immunoglobulin light chain loci and the development of B cells that coexpress two different immunoglobulin light chains and, therefore, two antibody specificities. Most allelically included B cells express two κ chains, although rare dual-λ cells are also observed. Moreover, these cells typically coexpress an autoreactive and a nonautoreactive antibody. Thus, allelically included B cells could operate like 'Trojan horses': expression and function of the nonautoreactive antigen receptors might promote their maturation, activation, and terminal differentiation into effector cells that also express and secrete autoantibodies. Indeed, dual-κ B cells are greatly expanded into effector B cell subsets in some autoimmune mice, thus indicating they might play an important role in disease.
Collapse
Affiliation(s)
- Roberta Pelanda
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, CO 80206, USA.
| |
Collapse
|
20
|
Peng C, Eckhardt LA. Role of the Igh intronic enhancer Eμ in clonal selection at the pre-B to immature B cell transition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:4399-411. [PMID: 24058175 PMCID: PMC3810302 DOI: 10.4049/jimmunol.1301858] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We previously described a checkpoint for allelic exclusion that occurs at the pre-B cell to immature B cell transition and is dependent upon the IgH intronic enhancer, Eμ. We now provide evidence that the breach in allelic exclusion associated with Eμ deletion results from decreased Igμ levels that make it difficult for emerging BCRs to reach the signaling threshold required for positive selection into the immature B cell compartment. We show that this compartment is smaller in mice carrying an Eμ-deficient, but functional, IgH allele (VHΔ(a)). Pre-B cells in such mice produce ≈ 50% wild-type levels of Igμ (mRNA and protein), and this is associated with diminished signals, as measured by phosphorylation of pre-BCR/BCR downstream signaling proteins. Providing Eμ-deficient mice with a preassembled VL gene led not only to a larger immature B cell compartment but also to a decrease in "double-producers," suggesting that H chain/L chain combinations with superior signaling properties can overcome the signaling defect associated with low Igμ-chain and can eliminate the selective advantage of "double-producers" that achieve higher Igμ-chain levels through expression of a second IgH allele. Finally, we found that "double-producers" in Eμ-deficient mice include a subpopulation with autoreactive BCRs. We infer that BCRs with IgH chain from the Eμ-deficient allele are ignored during negative selection owing to their comparatively low density. In summary, these studies show that Eμ's effect on IgH levels at the pre-B cell to immature B cell transition strongly influences allelic exclusion, the breadth of the mature BCR repertoire, and the emergence of autoimmune B cells.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- Cell Differentiation
- Cells, Cultured
- Clonal Selection, Antigen-Mediated
- Enhancer Elements, Genetic
- Genes, Immunoglobulin Heavy Chain
- Immunoglobulin Fragments/genetics
- Immunoglobulin Fragments/immunology
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphorylation
- Precursor Cells, B-Lymphoid/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Regulatory Sequences, Nucleic Acid
Collapse
Affiliation(s)
- Cheng Peng
- Department of Biological Sciences, Hunter College and The Graduate Center of The City University of New York, 695 Park Avenue, New York, NY, 10065
| | - Laurel A. Eckhardt
- Department of Biological Sciences, Hunter College and The Graduate Center of The City University of New York, 695 Park Avenue, New York, NY, 10065
| |
Collapse
|
21
|
Antibodies that bind complex glycosaminoglycans accumulate in the Golgi. Proc Natl Acad Sci U S A 2013; 110:11958-63. [PMID: 23818632 DOI: 10.1073/pnas.1308620110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Light (L) chains that edit anti-DNA heavy (H) chains rescue B-cell development by suppressing DNA binding. However, exceptional editor L chains allow B cells to reach splenic compartments even though their B-cell receptors remain autoreactive. Such incompletely edited B cells express multireactive antibodies that accumulate in the Golgi and are released as insoluble, amyloid-like immune complexes. Here, we examine examples of incomplete editing from the analysis of variable to joining (VJ) gene junction of the variable (Vλx) editor L chain. When paired with the anti-DNA heavy chain, VH56R, the Vλx variants yield antibodies with differing specificities, including glycosaminoglycan reactivity. Our results implicate these specificities in the evasion of receptor editing through intracellular sequestration of IgM and the release of insoluble IgM complexes. Our findings can be extrapolated to human L chains and have implications for understanding a latent component of the Ig repertoire that could exert pathogenic and protective functions.
Collapse
|
22
|
Kil LP, Hendriks RW. Aberrant B cell selection and activation in systemic lupus erythematosus. Int Rev Immunol 2013; 32:445-70. [PMID: 23768157 DOI: 10.3109/08830185.2013.786712] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The detrimental role of B lymphocytes in systemic lupus erythematosus (SLE) is evident from the high levels of pathogenic antinuclear autoantibodies (ANAs) found in SLE patients. Affirming this causative role, additional antibody-independent roles of B cells in SLE were appreciated. In recent years, many defects in B cell selection and activation have been identified in murine lupus models and SLE patients that explain the increased emergence and persistence of autoreactive B cells and their lowered activation threshold. Therefore, clinical trials with B cell depletion regimens in SLE patients were initiated but disappointingly the efficacy of B cell depleting agents proved to be limited. Remarkably however, a major breakthrough in SLE therapy was accomplished by blocking B cell survival factors rather then eliminating B cells. This surprising finding indicates that although SLE is a B cell-driven disease, the amplifying crosstalk between B cells and other cells of the immune system likely evokes the observed tolerance breakdown in B cells. Moreover, this implies that intelligent interception of pro-inflammatory loops rather then selectively silencing B cells will be key to the development of new SLE therapies. In this review, we will not only highlight the intrinsic B cell defects that facilitate the persistence of autoreactive B cells and their activation, but in addition we will focus on B cell extrinsic signals derived from T cells and innate immune cells that lower the activation threshold for B cells.
Collapse
Affiliation(s)
- Laurens P Kil
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, The Netherlands
| | | |
Collapse
|
23
|
Rowland SL, Tuttle K, Torres RM, Pelanda R. Antigen and cytokine receptor signals guide the development of the naïve mature B cell repertoire. Immunol Res 2013; 55:231-40. [PMID: 22941591 DOI: 10.1007/s12026-012-8366-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Immature B cells are generated daily in the bone marrow tissue. More than half of the newly generated immature B cells are autoreactive and bind a self-antigen, while the others are nonautoreactive. A selection process has evolved on the one hand to thwart development of autoreactive immature B cells and, on the other hand, to promote further differentiation of nonautoreactive immature B cells into transitional and mature B cells. These negative and positive selection events are carefully regulated by signals that emanate from the antigen receptor, whether antigen-mediated or tonic, and are influenced by signals that are generated by receptors that bind cytokines, chemokines, and other factors produced in the bone marrow tissue. These signals, therefore, are the predominant driving forces for the generation of a B cell population that is capable of protecting the body from infections while maintaining self-tolerance. Here, we review recent findings from our group and others that describe how tonic antigen receptor signaling and bone marrow cytokines regulate the selection of immature B cells.
Collapse
Affiliation(s)
- Sarah L Rowland
- Integrated Department of Immunology, University of Colorado School of Medicine, National Jewish Health, Denver, CO, USA
| | | | | | | |
Collapse
|
24
|
Immunogenetics shows that not all MBL are equal: the larger the clone, the more similar to CLL. Blood 2013; 121:4521-8. [DOI: 10.1182/blood-2012-12-471698] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Key Points
Low-count and high-count monoclonal B-cell lymphocytosis (MBL) have distinct immunogenetic signatures, with only the latter resembling CLL. Rather than a true premalignant condition, low-count MBL may merely reflect immune senescence or result from persistent antigen stimulation.
Collapse
|
25
|
Makdasi E, Eilat D. L chain allelic inclusion does not increase autoreactivity in lupus-prone New Zealand Black/New Zealand White mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:1472-80. [PMID: 23319731 DOI: 10.4049/jimmunol.1202331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
L chain allelic inclusion has been proposed as a B cell tolerance mechanism in addition to clonal deletion, clonal anergy, and receptor editing. It is said to rescue autoreactive B cells from elimination by diluting out the self-reactive BCR through the expression of a second innocuous L chain. In autoimmune animals, such as lupus-prone mice, allelically included B cells could be activated and produce pathogenic autoantibodies. We have previously shown that anti-DNA hybridomas from diseased New Zealand Black/New Zealand White F1 mice exhibit nearly perfect allelic exclusion. In the current study, we have analyzed single B cells from these and from nonautoimmune mice. In addition, we have cloned and expressed the Ig variable regions of several L chain-included B cells in cell culture. We find that although the number of L chain-included B cells increases as a result of receptor editing, the majority of such cells do not retain an autoreactive HxL chain combination and, therefore, allelic inclusion in itself does not serve as a B cell tolerance mechanism in these autoimmune mice.
Collapse
Affiliation(s)
- Efi Makdasi
- Department of Medicine, Hadassah University Hospital, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | | |
Collapse
|
26
|
Andrews SF, Zhang Q, Lim S, Li L, Lee JH, Zheng NY, Huang M, Taylor WM, Farris AD, Ni D, Meng W, Luning Prak ET, Wilson PC. Global analysis of B cell selection using an immunoglobulin light chain-mediated model of autoreactivity. ACTA ACUST UNITED AC 2012; 210:125-42. [PMID: 23267014 PMCID: PMC3549719 DOI: 10.1084/jem.20120525] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The nature of the immunoglobulin light chain affects peripheral B cell tolerance and autoreactivity. The important subtleties of B cell tolerance are best understood in a diverse immunoglobulin (Ig) repertoire context encoding a full spectrum of autoreactivity. To achieve this, we used mice expressing Igκ transgenes that confer varying degrees of autoreactivity within a diverse heavy chain (HC) repertoire. These transgenes, coupled with a biomarker to identify receptor-edited cells and combined with expression cloning of B cell receptors, allowed us to analyze tolerance throughout B cell development. We found that both the nature of the autoantigen and the Ig HC versus light chain (LC) contribution to autoreactivity dictate the developmental stage and mechanism of tolerance. Furthermore, although selection begins in the bone marrow, over one third of primary tolerance occurs in the periphery at the late transitional developmental stage. Notably, we demonstrate that the LC has profound effects on tolerance and can lead to exacerbated autoantibody production.
Collapse
Affiliation(s)
- Sarah F Andrews
- Section of Rheumatology, Department of Medicine, Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Gatto M, Zen M, Ghirardello A, Bettio S, Bassi N, Iaccarino L, Punzi L, Doria A. Emerging and critical issues in the pathogenesis of lupus. Autoimmun Rev 2012; 12:523-36. [PMID: 23000207 DOI: 10.1016/j.autrev.2012.09.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 09/10/2012] [Indexed: 01/10/2023]
Abstract
Systemic lupus erythematosus (SLE) is a multisystemic, autoimmune disease, encompassing either mild or severe manifestations. SLE was originally labeled as being an immune complex-mediated disease, but further knowledge suggested its pathogenesis is motlier than that, involving complex interactions between predisposed individuals and their environment. People affected with SLE have their immune system skewed toward aberrant self-recognition usually after encountering a triggering agent. Defeats in early and late immune checkpoints contribute to tolerance breakdown and further generation and expansion of autoreactive cell-clones. B and T cells play a master role in SLE, however clues are emerging about other cell types and new light is being shed on SLE autoantibodies, since some of them display really harmful potential (pathogenic antibodies), while others are just connected with disease development (pathological antibodies) and may even be protective. Autoantibody generation is elicited by abnormal apoptosis and inefficient clearance of cellular debris causing intracellular autoantigens (e.g. nucleosomes) to persist in the extracellular environment, being further recognized by autoreactive cells. Here we explore the complexity of SLE pathogenesis through five core issues, i.e. genetic predisposition, B and T cell abnormalities, abnormal autoantigen availability, autoantibody generation and organ damage, relying on current knowledge and recent insights into SLE development.
Collapse
Affiliation(s)
- Mariele Gatto
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Receptor editing is the process that replaces the heavy chain or light chain variable region genes in a B cell immunoglobulin receptor that is already productively rearranged. It is a major mechanism in the bone marrow for maintaining B cell tolerance to autoantigens. We propose that a pathological autoimmune process can use receptor editing to induce the de novo creation and activation of B cells with autoreactive receptors in the peripheral immune system.
Collapse
Affiliation(s)
- Robert A Eisenberg
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA 19104-6160, United States.
| |
Collapse
|
29
|
Fournier EM, Velez MG, Leahy K, Swanson CL, Rubtsov AV, Torres RM, Pelanda R. Dual-reactive B cells are autoreactive and highly enriched in the plasmablast and memory B cell subsets of autoimmune mice. ACTA ACUST UNITED AC 2012; 209:1797-812. [PMID: 22927551 PMCID: PMC3457739 DOI: 10.1084/jem.20120332] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dual–light chain–expressing B cells in autoimmune prone mice increase with age, contribute to the memory and plasma cell compartments, and are autoreactive. Rare dual-reactive B cells expressing two types of Ig light or heavy chains have been shown to participate in immune responses and differentiate into IgG+ cells in healthy mice. These cells are generated more often in autoreactive mice, leading us to hypothesize they might be relevant in autoimmunity. Using mice bearing Igk allotypic markers and a wild-type Ig repertoire, we demonstrate that the generation of dual-κ B cells increases with age and disease progression in autoimmune-prone MRL and MRL/lpr mice. These dual-reactive cells express markers of activation and are more frequently autoreactive than single-reactive B cells. Moreover, dual-κ B cells represent up to half of plasmablasts and memory B cells in autoimmune mice, whereas they remain infrequent in healthy mice. Differentiation of dual-κ B cells into plasmablasts is driven by MRL genes, whereas the maintenance of IgG+ cells is partly dependent on Fas inactivation. Furthermore, dual-κ B cells that differentiate into plasmablasts retain the capacity to secrete autoantibodies. Overall, our study indicates that dual-reactive B cells significantly contribute to the plasmablast and memory B cell populations of autoimmune-prone mice suggesting a role in autoimmunity.
Collapse
Affiliation(s)
- Emilie M Fournier
- Integrated Department of Immunology, National Jewish Health and University of Colorado, Denver School of Medicine, Denver, CO 80206, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
A fundamental property of the immune system is its ability to mediate self-defense with a minimal amount of collateral damage to the host. The system uses several different mechanisms to achieve this goal, which is collectively referred to as the "process of immunological tolerance." This article provides an introductory historical overview to these various mechanisms, which are discussed in greater detail throughout this collection, and then briefly describes what happens when this process fails, a state referred to as "autoimmunity."
Collapse
Affiliation(s)
- Ronald H Schwartz
- Laboratory of Cellular and Molecular Immunology, NIAID, National Institutes of Health, Bethesda, Maryland 20892-0420, USA.
| |
Collapse
|
31
|
Abstract
The development of an adaptive immune system based on the random generation of antigen receptors requires a stringent selection process that sifts through receptor specificities to remove those reacting with self-antigens. In the B-cell lineage, this selection process is first applied to IgM(+) immature B cells. By using increasingly sophisticated mouse models, investigators have identified the central tolerance mechanisms that negatively select autoreactive immature B cells and prevent inclusion of their antigen receptors into the peripheral B-cell pool. Additional studies have uncovered mechanisms that promote the differentiation of nonautoreactive immature B cells and their positive selection into the peripheral B-cell population. These mechanisms of central selection are fundamental to the generation of a naïve B-cell repertoire that is largely devoid of self-reactivity while capable of reacting with any foreign insult.
Collapse
Affiliation(s)
- Roberta Pelanda
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver School of Medicine, Denver, Colorado 80206, USA.
| | | |
Collapse
|
32
|
Khan SN, Cox JV, Nishimoto SK, Chen C, Fritzler MJ, Hendershot LM, Weigert M, Radic M. Intra-Golgi formation of IgM-glycosaminoglycan complexes promotes Ig deposition. THE JOURNAL OF IMMUNOLOGY 2011; 187:3198-207. [PMID: 21841132 DOI: 10.4049/jimmunol.1101336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immune complexes arise from interactions between secreted Ab and Ags in the surrounding milieu. However, it is not known whether intracellular Ag-Ab interactions also contribute to the formation of extracellular immune complexes. In this study, we report that certain murine B cell hybridomas accumulate intracellular IgM and release large, spherical IgM complexes. The complexes (termed "spherons") reach 2 μm in diameter, detach from the cell surface, and settle out of solution. The spherons contain IgM multimers that incorporate the J chain and resist degradation by endoglycosidase H, arguing for IgM passage through the Golgi. Treatment of cells with inhibitors of proteoglycan synthesis, or incubation of spherons with chondroitinase ABC, degrades spherons, indicating that spheron formation and growth depend on interactions between IgM and glycosaminoglycans. This inference is supported by direct binding of IgM to heparin and hyaluronic acid. We conclude that, as a consequence of IgM binding to glycosaminoglycans, multivalent IgM-glycan complexes form in transit of IgM to the cell surface. Intra-Golgi formation of immune complexes could represent a new pathogenic mechanism for immune complex deposition disorders.
Collapse
Affiliation(s)
- Salar N Khan
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Alternative mechanisms of receptor editing in autoreactive B cells. Proc Natl Acad Sci U S A 2011; 108:7125-30. [PMID: 21471456 DOI: 10.1073/pnas.1019389108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pathogenic anti-DNA antibodies expressed in systemic lupus erythematosis bind DNA mainly through electrostatic interactions between the positively charged Arg residues of the antibody complementarity determining region (CDR) and the negatively charged phosphate groups of DNA. The importance of Arg in CDR3 for DNA binding has been shown in mice with transgenes coding for anti-DNA V(H) regions; there is also a close correlation between arginines in CDR3 of antibodies and DNA binding. Codons for Arg can readily be formed by V(D)J rearrangement; thereby, antibodies that bind DNA are part of the preimmune repertoire. Anti-DNAs in healthy mice are regulated by receptor editing, a mechanism that replaces κ light (L) chains compatible with DNA binding with κ L chains that harbor aspartic residues. This negatively charged amino acid is thought to neutralize Arg sites in the V(H). Editing by replacement is allowed at the κ locus, because the rearranged VJ is nested between unrearranged Vs and Js. However, neither λ nor heavy (H) chain loci are organized so as to allow such second rearrangements. In this study, we analyze regulation of anti-DNA H chains in mice that lack the κ locus, κ-/κ- mice. These mice show that the endogenous preimmune repertoire does indeed include a high frequency of antibodies with Arg in their CDR3s (putative anti-DNAs) and they are associated mainly with the editor L chain λx. The editing mechanisms in the case of λ-expressing B cells include L chain allelic inclusion and V(H) replacement.
Collapse
|
34
|
Luning Prak ET, Monestier M, Eisenberg RA. B cell receptor editing in tolerance and autoimmunity. Ann N Y Acad Sci 2011; 1217:96-121. [PMID: 21251012 DOI: 10.1111/j.1749-6632.2010.05877.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Receptor editing is the process of ongoing antibody gene rearrangement in a lymphocyte that already has a functional antigen receptor. The expression of a functional antigen receptor will normally terminate further rearrangement (allelic exclusion). However, lymphocytes with autoreactive receptors have a chance at escaping negative regulation by "editing" the specificities of their receptors with additional antibody gene rearrangements. As such, editing complicates the Clonal Selection Hypothesis because edited cells are not simply endowed for life with a single, invariant antigen receptor. Furthermore, if the initial immunoglobulin gene is not inactivated during the editing process, allelic exclusion is violated and the B cell can exhibit two specificities. Here, we describe the discovery of editing, the pathways of receptor editing at the heavy (H) and light (L) chain loci, and current evidence regarding how and where editing happens and what effects it has on the antibody repertoire.
Collapse
Affiliation(s)
- Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | |
Collapse
|
35
|
Almqvist N, Winkler TH, Mårtensson IL. Autoantibodies: Focus on anti-DNA antibodies. SELF NONSELF 2011; 2:11-18. [PMID: 21776330 DOI: 10.4161/self.2.1.15087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 11/19/2022]
Abstract
Ever since the days of Ehrlich and the birth of humoral immunity, self-reactivity or 'horror autotoxicus' as referred to by Paul Ehrlich, has been of great concern. For instance, in patients with the autoimmune disease systemic lupus erythematosus (SLE), anti-nuclear and anti-DNA antibodies have been recognized for many years. Despite this, the exact mechanism as to how the immune system fails to protect the individual and allows these autoantibodies to develop in this and other systemic autoimmune diseases remains uncertain. So how can we explain their presence? Evidence suggests that B cells expressing autoreactive antibodies do not normally arise but rather undergo negative selection as they develop. In light of this, it might seem contradictory that not all autoreactive B cell clones are eliminated, although this may not even be the intention since autoantibodies are also found in healthy individuals and may even protect from autoimmunity. Here, we will discuss autoantibodies, in particular those recognizing DNA, with regard to their reactivity and their potentially pathogenic or protective properties.
Collapse
Affiliation(s)
- Nina Almqvist
- Department of Rheumatology and Inflammation Research; the Sahlgrenska Academy; University of Gothenburg; Gothenburg, Sweden
| | | | | |
Collapse
|
36
|
Link A, Bachmann MF. Immunodrugs: breaking B- but not T-cell tolerance with therapeutic anticytokine vaccines. Immunotherapy 2010; 2:561-74. [PMID: 20636009 DOI: 10.2217/imt.10.30] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathology in most chronic inflammatory diseases is characterized by an imbalance in cytokine expression. Targeting cytokines with monoclonal antibodies has proven to be a highly effective treatment. However, monoclonal antibody therapy has disadvantages such as high production costs, generation of antimonoclonal antibodies and the inconvenience of frequent injections. Therapeutic vaccines have the potential to overcome these limitations. The aim of active vaccination is to induce B-cell responses and obtain autoantibodies capable of neutralizing the interaction of the targeted cytokine with its receptor. In order to achieve this, therapeutic vaccines need to circumvent the potent tolerance mechanisms that exist to prevent immune responses against self-molecules. This article focuses on the tolerance mechanisms of the B- and T-cell compartments and how these may be manipulated to obtain high-affinity autoantibodies without inducing potentially dangerous autoreactive T-cell responses.
Collapse
Affiliation(s)
- Alexander Link
- Cytos Biotechnology AG, CH-8952 Zurich-Schlieren, Switzerland
| | | |
Collapse
|
37
|
Rowland SL, Leahy KF, Halverson R, Torres RM, Pelanda R. BAFF receptor signaling aids the differentiation of immature B cells into transitional B cells following tonic BCR signaling. THE JOURNAL OF IMMUNOLOGY 2010; 185:4570-81. [PMID: 20861359 DOI: 10.4049/jimmunol.1001708] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BAFF is an important prosurvival cytokine for mature B cells. However, previous studies have shown that BAFFR is already expressed at the immature B cell stage, and that the prosurvival protein Bcl-2 does not completely complement the B cell defects resulting from the absence of BAFFR or BAFF. Thus, we hypothesized that BAFF also functions to aid the differentiation of nonautoreactive immature B cells into transitional B cells and to promote their positive selection. We found that BAFFR is expressed at higher levels on nonautoreactive than on autoreactive immature B cells and that its expression correlates with that of surface IgM and with tonic BCR signaling. Our data indicate that BAFFR signaling enhances the generation of transitional CD23(-) B cells in vitro by increasing cell survival. In vivo, however, BAFFR signaling is dispensable for the generation of CD23(-) transitional B cells in the bone marrow, but it is important for the development of transitional CD23(-) T1 B cells in the spleen. Additionally, we show that BAFF is essential for the differentiation of CD23(-) into CD23(+) transitional B cells both in vitro and in vivo through a mechanism distinct from that mediating cell survival, but requiring tonic BCR signaling. In summary, our data indicate that BAFFR and tonic BCR signals cooperate to enable nonautoreactive immature B cells to differentiate into transitional B cells and to be positively selected into the naive B cell repertoire.
Collapse
Affiliation(s)
- Sarah L Rowland
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, CO 80206, USA
| | | | | | | | | |
Collapse
|
38
|
Basten A, Silveira PA. B-cell tolerance: mechanisms and implications. Curr Opin Immunol 2010; 22:566-74. [PMID: 20829011 DOI: 10.1016/j.coi.2010.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 08/07/2010] [Indexed: 11/26/2022]
Abstract
Advances in our knowledge of the spectrum of B-cell activities combined with the remarkable clinical efficacy of B-cell inhibitors in autoimmunity and transplantation settings serve to re-emphasise the importance of tolerance to self and foreign antigens in the B-cell repertoire. In particular, new information is emerging about the molecular mechanisms involved in B-cell tolerance induction and identification of B-cell selective defects that contribute to the pathogenesis of autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
- Antony Basten
- Immunology Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| | | |
Collapse
|
39
|
Abstract
The allelic exclusion of immunoglobulin (Ig) genes is one of the most evolutionarily conserved features of the adaptive immune system and underlies the monospecificity of B cells. While much has been learned about how Ig allelic exclusion is established during B-cell development, the relevance of monospecificity to B-cell function remains enigmatic. Here, we review the theoretical models that have been proposed to explain the establishment of Ig allelic exclusion and focus on the molecular mechanisms utilized by developing B cells to ensure the monoallelic expression of Ig kappa and Ig lambda light chain genes. We also discuss the physiological consequences of Ig allelic exclusion and speculate on the importance of monospecificity of B cells for immune recognition.
Collapse
Affiliation(s)
- Christian Vettermann
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | |
Collapse
|
40
|
Abstract
For several decades, allograft rejection was believed to be mediated almost exclusively by cellular immune responses, but it is now realized that humoral responses also play a major role. Although directed typically against donor human leukocyte antigen, it is becoming increasingly evident that the antibody response can also target autoantigens that are shared between donor and recipient and that this autoantibody may contribute to graft rejection. Many aspects of transplant-induced humoral autoimmunity remain poorly understood and key questions persist; not least what triggers the response and how autoantibody causes graft damage. Here, we collate results from recent clinical and experimental studies in transplantation and autoimmune diseases to propose answers to these questions.
Collapse
|
41
|
Tussiwand R, Bosco N, Ceredig R, Rolink AG. Tolerance checkpoints in B-cell development: Johnny B good. Eur J Immunol 2009; 39:2317-24. [PMID: 19714572 DOI: 10.1002/eji.200939633] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
B-cell development up to the immature B-cell stage takes place in the bone marrow, while final maturation into mature B cells occurs in the spleen. During differentiation, the precursor and immature B cells have to pass several checkpoints, including those in which they are censored for being auto-reactive, and therefore being potentially dangerous. Numerous studies have shown that the immature B-cell stage in the bone marrow and the transitional B-cell stages in the spleen comprise distinct checkpoints at which auto-reactivity is censored. Recently, evidence has been provided that the large pre-BII stage in the bone marrow, at which the pre-BCR is expressed, is yet another B-cell tolerance checkpoint. Here, we review these findings and speculate on directions for possible further experimentation.
Collapse
Affiliation(s)
- Roxane Tussiwand
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Mattenstrasse, Basel, Switzerland
| | | | | | | |
Collapse
|
42
|
Burioni R, Canducci F, Saita D, Perotti M, Mancini N, De Marco D, Clementi N, Chieffo A, Denaro M, Cianflone D, Manfredi AA, Colombo A, Maseri A, Clementi M. Antigen-driven evolution of B lymphocytes in coronary atherosclerotic plaques. THE JOURNAL OF IMMUNOLOGY 2009; 183:2537-44. [PMID: 19635916 DOI: 10.4049/jimmunol.0901076] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent data indicated that adaptive immunity is involved in the process of atherogenesis. Oligoclonal recruitment of T lymphocytes has been described in coronary plaques of patients with acute coronary syndrome. However, the nature of immune response remains to be determined. In the present study, we examined the Ab response in six coronary plaques obtained by endoluminal directional atherectomy. The IgG1/kappa-coding gene repertoires of B lymphocytes present in circulating blood and in coronary plaques were cloned and analyzed. In all of the six plaques, we observed 1) a skewed usage of heavy and light IgG1/kappa Ab-coding genes, 2) an oligoclonal distribution of V(K), J(K), and V(H), D(H), and J(H) genes with overrepresentation of some rarely used IgG genes, and 3) the unequivocal signs of Ag-driven clonal expansion and evolution of B cells. The data document for the first time the presence of a local Ag-driven clonal evolution of B cells in human atherosclerotic plaques.
Collapse
|
43
|
Makdasi E, Fischel R, Kat I, Eilat D. Autoreactive Anti-DNA Transgenic B Cells in Lupus-Prone New Zealand Black/New Zealand White Mice Show Near Perfect L Chain Allelic Exclusion. THE JOURNAL OF IMMUNOLOGY 2009; 182:6143-8. [DOI: 10.4049/jimmunol.0803610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
44
|
Abstract
Work from multiple groups continues to provide additional evidence for the powerful and highly diverse roles, both protective and pathogenic, that B cells play in autoimmune diseases. Similarly, it has become abundantly clear that antibody-independent functions may account for the opposing influences that B cells exercise over other arms of the immune response and ultimately over autoimmunity itself. Finally, it is becoming apparent that the clinical impact of B-cell depletion therapy may be, to a large extent, determined by the functional balance between different B-cell subsets that may be generated by this therapeutic intervention. In this review, we postulate that our perspective of B-cell tolerance and our experimental approach to its understanding are fundamentally changed by this view of B cells. Accordingly, we first discuss current knowledge of B-cell tolerance conventionally defined as the censoring of autoantibody-producing B cells (with an emphasis on human B cells). Therefore, we discuss a different model that contemplates B cells not only as targets of tolerance but also as mediators of tolerance. This model is based on the notion that the onset of clinical autoimmune disease may require a B-cell gain-of-pathogenic function (or a B-cell loss-of-regulatory-function) and that accordingly, disease remission may depend on the restoration of the physiological balance between B-cell pathogenic and protective functions.
Collapse
Affiliation(s)
- Nataly Manjarrez-Orduño
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | |
Collapse
|
45
|
Zhang Y, Su SC, Hecox DB, Brady GF, Mackin KM, Clark AG, Foster MH. Central tolerance regulates B cells reactive with Goodpasture antigen alpha3(IV)NC1 collagen. THE JOURNAL OF IMMUNOLOGY 2009; 181:6092-100. [PMID: 18941198 DOI: 10.4049/jimmunol.181.9.6092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Patients and rodents with Goodpasture's syndrome (GPS) develop severe autoimmune crescentic glomerulonephritis, kidney failure, and lung hemorrhage due to binding of pathogenic autoantibodies to the NC1 domain of the alpha3 chain of type IV collagen. Target epitopes are cryptic, normally hidden from circulating Abs by protein-protein interactions and the highly tissue-restricted expression of the alpha3(IV) collagen chain. Based on this limited Ag exposure, it has been suggested that target epitopes are not available as B cell tolerogens. To determine how pathogenic anti-GPS autoantibody responses are regulated, we generated an Ig transgenic (Tg) mouse model that expresses an Ig that binds alpha3(IV)NC1 collagen epitopes recognized by serum IgG of patients with GPS. Phenotypic analysis reveals B cell depletion and L chain editing in Tg mice. To determine the default tolerance phenotype in the absence of receptor editing and endogenous lymphocyte populations, we crossed Tg mice two generations with mice deficient in Rag. Resulting Tg Rag-deficient mice have central B cell deletion. Thus, development of Tg anti-alpha3(IV)NC1 collagen B cells is halted in the bone marrow, at which point the cells are deleted unless rescued by a Rag enzyme-dependent process, such as editing. The central tolerance phenotype implies that tolerizing self-Ag is expressed in bone marrow.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Li F, Eckhardt LA. A role for the IgH intronic enhancer E mu in enforcing allelic exclusion. ACTA ACUST UNITED AC 2008; 206:153-67. [PMID: 19114667 PMCID: PMC2626684 DOI: 10.1084/jem.20081202] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intronic enhancer (Eμ) of the immunoglobulin heavy chain (IgH) locus is critical for V region gene assembly. To determine Eμ's subsequent functions, we created an Igh allele with assembled VH gene but with Eμ removed. In mice homozygous for this Eμ-deficient allele, B cell development was normal and indistinguishable from that of mice with the same VH knockin and Eμ intact. In mice heterozygous for the Eμ-deficient allele, however, allelic exclusion was severely compromised. Surprisingly, this was not a result of reduced suppression of V-DJ assembly on the second allele. Rather, the striking breakdown in allelic exclusion took place at the pre-B to immature B cell transition. These findings reveal both an important role for Eμ in influencing the fate of newly arising B cells and a second checkpoint for allelic exclusion.
Collapse
Affiliation(s)
- Fubin Li
- Hunter College and Graduate Center of the City University of New York, New York, NY 10065, USA
| | | |
Collapse
|
47
|
Panigrahi AK, Goodman NG, Eisenberg RA, Rickels MR, Naji A, Luning Prak ET. RS rearrangement frequency as a marker of receptor editing in lupus and type 1 diabetes. ACTA ACUST UNITED AC 2008; 205:2985-94. [PMID: 19075293 PMCID: PMC2605238 DOI: 10.1084/jem.20082053] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Continued antibody gene rearrangement, termed receptor editing, is an important mechanism of central B cell tolerance that may be defective in some autoimmune individuals. We describe a quantitative assay for recombining sequence (RS) rearrangement that we use to estimate levels of antibody light chain receptor editing in various B cell populations. RS rearrangement is a recombination of a noncoding gene segment in the κ antibody light chain locus. RS rearrangement levels are highest in the most highly edited B cells, and are inappropriately low in autoimmune mouse models of systemic lupus erythematosus (SLE) and type 1 diabetes (T1D), including those without overt disease. Low RS rearrangement levels are also observed in human subjects with SLE or T1D.
Collapse
Affiliation(s)
- Anil K Panigrahi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
48
|
Mietzner B, Tsuiji M, Scheid J, Velinzon K, Tiller T, Abraham K, Gonzalez JB, Pascual V, Stichweh D, Wardemann H, Nussenzweig MC. Autoreactive IgG memory antibodies in patients with systemic lupus erythematosus arise from nonreactive and polyreactive precursors. Proc Natl Acad Sci U S A 2008; 105:9727-32. [PMID: 18621685 PMCID: PMC2474524 DOI: 10.1073/pnas.0803644105] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Indexed: 12/30/2022] Open
Abstract
Persistent autoantibody production in patients with systemic lupus erythematosus (SLE) suggests the existence of autoreactive humoral memory, but the frequency of self-reactive memory B cells in SLE has not been determined. Here, we report on the reactivity of 200 monoclonal antibodies from single IgG+ memory B cells of four SLE patients. The overall frequency of polyreactive and HEp-2 self-reactive antibodies in this compartment was similar to controls. We found 15% of IgG memory B cell antibodies highly reactive and specific for SLE-associated extractable nuclear antigens (ENA) Ro52 and La in one patient with serum autoantibody titers of the same specificity but not in the other three patients or healthy individuals. The germ-line forms of the ENA antibodies were non-self-reactive or polyreactive with low binding to Ro52, supporting the idea that somatic mutations contributed to autoantibody specificity and reactivity. Heterogeneity in the frequency of memory B cells expressing SLE-associated autoantibodies suggests that this variable may be important in the outcome of therapies that ablate this compartment.
Collapse
Affiliation(s)
- Brun Mietzner
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | | | | | | | - Thomas Tiller
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Klaus Abraham
- Central Institute for Laboratory Medicine and Pathobiochemistry, Charité Hospital, 13353 Berlin, Germany
| | - Jose B. Gonzalez
- Institute for Clinical Chemistry and Pathobiochemistry, Charité Hospital, 12200 Berlin, Germany; and
| | - Virginia Pascual
- Baylor Institute for Immunology Research, Baylor University, Dallas, TX 75204
| | - Dorothee Stichweh
- Baylor Institute for Immunology Research, Baylor University, Dallas, TX 75204
| | - Hedda Wardemann
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | | |
Collapse
|
49
|
Abstract
B cells are essential for the development and pathogenesis of both systemic and organ-specific autoimmune diseases. Autoreactive B cells are typically thought of as sources of autoantibody, but their most important pathogenetic roles may be to present autoantigens to T cells and to secrete proinflammatory cytokines. A rate-limiting step in the genesis of autoimmunity then is the activation of autoreactive B cells. Here, mechanisms are discussed that normally prevent such activation and how they break down during disease. Integrating classic work with recent insights, emphasis is placed on efforts to pinpoint the precursor cells for autoantibody-secreting cells and the unique stimuli and pathways by which they are activated.
Collapse
|
50
|
Witsch EJ, Bettelheim E. Allelic and Isotypic Light Chain Inclusion in Peripheral B Cells from Anti-DNA Antibody Transgenic C57BL/6 and BALB/c Mice. THE JOURNAL OF IMMUNOLOGY 2008; 180:3708-18. [DOI: 10.4049/jimmunol.180.6.3708] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|