1
|
Hillion S, Miranda A, Le Dantec C, Boudigou M, Le Pottier L, Cornec D, Torres RM, Pelanda R. Maf expression in B cells restricts reactive plasmablast and germinal center B cell expansion. Nat Commun 2024; 15:7982. [PMID: 39266537 PMCID: PMC11393457 DOI: 10.1038/s41467-024-52224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 08/29/2024] [Indexed: 09/14/2024] Open
Abstract
Precise regulation of B cell differentiation is essential for an effective adaptive immune response. Here, we show that B cell development in mice with B cell-specific Maf deletion is unaffected, but marginal zone B cells, germinal centre B cells, and plasmablasts are significantly more frequent in the spleen of naive Maf-deficient mice compared to wild type controls. In the context of a T cell-dependent immunization, Maf deletion causes increased proliferation of germinal centre B cells and extrafollicular plasmablasts. This is accompanied by higher production of antigen-specific IgG1 antibodies with minimal modification of early memory B cells, but a reduction in plasma cell numbers. Single-cell RNA sequencing shows upregulation of genes associated with DNA replication and cell cycle progression, confirming the role of Maf in cell proliferation. Subsequent pathway analysis reveals that Maf influences cellular metabolism, transporter activity, and mitochondrial proteins, which have been implicated in controlling the germinal centre reaction. In summary, our findings demonstrate that Maf acts intrinsically in B cells as a negative regulator of late B cell differentiation, plasmablast proliferation and germinal centre B cell formation.
Collapse
Affiliation(s)
- Sophie Hillion
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Anjelica Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | | | | | | | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France
| | - 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
|
Hartmeier PR, Kosanovich JL, Velankar KY, Ostrowski SM, Busch EE, Lipp MA, Empey KM, Meng WS. Modeling the kinetics of lymph node retention and exposure of a cargo protein delivered by biotin-functionalized nanoparticles. Acta Biomater 2023; 170:453-463. [PMID: 37652212 PMCID: PMC10592217 DOI: 10.1016/j.actbio.2023.08.048] [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: 04/04/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Generation of protective immunity through vaccination arises from the adaptive immune response developed primarily in the lymph nodes drained from the immunization site. Relative to the intramuscular route, subcutaneous administration allows for direct and rapid access to the lymphatics, but accumulation of soluble protein antigens within the lymph nodes is limited. Subunit vaccines also require immune stimulating adjuvants which may not accumulate in the same lymph nodes simultaneously with antigen. Herein we report the use of biotinylated poly (lactic-co-glycolic acid) nanoparticles (bNPs) to enhance delivery of a model protein antigen to the lymphatics. bNPs provide dual functionality as adjuvant and vehicle to localize antigens with stimulated immune cells in the same draining lymph node. Using streptavidin as a model antigen, which can be loaded directly onto the bNP surface, we evaluated the kinetics of lymph node occupancy and adaptive immune responses in wildtype C57BL/6 mice. Antigen exposure in vivo was significantly improved through surface loading onto bNPs, and we developed a working kinetic model to account for the retention of both particles and antigen in draining lymph nodes. We observed enhanced T cell responses and antigen-specific B cell response in vivo when antigen was delivered on the particle surface. This work highlights the advantage of combining intrinsic adjuvant and antigen loading in a single entity, and the utility of kinetic modeling in the understanding of particle-based vaccines. STATEMENT OF SIGNIFICANCE: Development of safe and effective subunit vaccines depends on effective formulations that render optimized exposure and colocalization of antigens and adjuvants. In this work, we utilize a nanoparticle system which features self-adjuvanting properties and allows for surface loading of recombinant protein antigens. Using in vivo imaging, we demonstrated prolonged co-localization of the antigen and adjuvant particles in draining lymph nodes and provided evidence of B cell activation for up to 21 days following subcutaneous injection. A pharmacokinetic model was developed as a step towards bridging the translational gap between particulate-based vaccines and observed outcomes. The results have implications for the rational design of particle-based vaccines.
Collapse
Affiliation(s)
- Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Jessica L Kosanovich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15219, USA
| | - Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Sarah M Ostrowski
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15219, USA
| | - Emelia E Busch
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Madeline A Lipp
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15219, USA
| | - Kerry M Empey
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15219, USA; Department of Immunology, School of Medicine, University of Pittsburgh, PA 15219, USA.
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15219, USA.
| |
Collapse
|
3
|
McNitt DH, Joosse BA, Thomas JW, Bonami RH. Productive Germinal Center Responses Depend on the Nature of Stimuli Received by Anti-Insulin B Cells in Type 1 Diabetes-Prone Mice. Immunohorizons 2023; 7:384-397. [PMID: 37261716 PMCID: PMC10448785 DOI: 10.4049/immunohorizons.2300036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Islet autoantibodies, including those directed at insulin, predict type 1 diabetes (T1D) in mice and humans and signal immune tolerance breach by B lymphocytes. High-affinity insulin autoantibodies and T follicular helper cell involvement implicate germinal centers (GCs) in T1D. The VH125SD BCR transgenic model, in which 1-2% of peripheral B lymphocytes recognize insulin, enables direct study of insulin-binding B cells. Our prior studies showed that anti-insulin B cell receptor transgene site-directed to H chain locus mice fail to generate insulin Ab following T-dependent immunization, but it was unclear whether anti-insulin B cells were blocked for GC initiation, survival, or differentiation into Ab-secreting cells. Here, we show that insulin-binding B cells in T1D-prone anti-insulin B cell receptor transgene site-directed to H chain locus mice can spontaneously adopt a GC phenotype and undergo class switching to the IgG1 isotype, with little if any switching to IgG2b. T-dependent immunizations with insulin SRBC or insulin CFA drove anti-insulin B lymphocytes to adopt a GC phenotype, despite blunted insulin Ab production. Dual immunization against self (insulin) and foreign (4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin) Ags showed an anti-insulin (but not anti-4-hydroxy-3-nitrophenylacetyl) Ab block that tracked with increased expression of the apoptosis marker, activated caspase 3, in self-reactive GC B cells. Finally, T-independent immunization with insulin conjugated to Brucella abortus ring test Ag released immune tolerance to allow robust expansion of anti-insulin GC B cells and IgG-switched insulin Ab production. Overall, these data pinpoint GC survival and Ab-secreting cell differentiation as immune tolerance blocks that limit T-dependent, but not T-independent, stimulation of anti-insulin B cell responses.
Collapse
Affiliation(s)
- Dudley H. McNitt
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Bryan A. Joosse
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - James W. Thomas
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
4
|
Kuraoka M, Yeh CH, Bajic G, Kotaki R, Song S, Windsor I, Harrison SC, Kelsoe G. Recall of B cell memory depends on relative locations of prime and boost immunization. Sci Immunol 2022; 7:eabn5311. [PMID: 35522723 PMCID: PMC9169233 DOI: 10.1126/sciimmunol.abn5311] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immunization or microbial infection can establish long-term B cell memory not only systemically but also locally. Evidence has suggested that local B cell memory contributes to early local plasmacytic responses after secondary challenge. However, it is unclear whether locality of immunization plays any role in memory B cell participation in recall germinal centers (GCs), which is essential for updating their B cell antigen receptors (BCRs). Using single B cell culture and fate mapping, we have characterized BCR repertoires in recall GCs after boost immunizations at sites local or distal to the priming. Local boosts with homologous antigen recruit the progeny of primary GC B cells to recall GCs more efficiently than do distal boosts. Recall GCs elicited by local boosts contain significantly more B cells with elevated levels of immunoglobulin (Ig) mutation and higher avidity BCRs. This local preference is unaffected by blocking CD40:CD154 interaction to terminate active, GC responses. Local boosts with heterologous antigens elicit secondary GCs with B cell populations enriched for cross-reactivity to the prime and boost antigens; in contrast, cross-reactive GC B cells are rare after distal boosts. Our results suggest that local B cell memory is retained in the form of memory B cells, GC B cells, and GC phenotype B cells that are independent of organized GC structures and that these persistent "primed B cells" contribute to recall GC responses at local sites. Our findings indicate the importance of locality in humoral immunity and inform serial vaccination strategies for evolving viruses.
Collapse
Affiliation(s)
| | - Chen-Hao Yeh
- Department of Immunology, Duke University, Durham, NC, USA
| | - Goran Bajic
- Laboratory of Molecular Medicine, Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryutaro Kotaki
- Department of Immunology, Duke University, Durham, NC, USA
| | - Shengli Song
- Department of Immunology, Duke University, Durham, NC, USA
| | - Ian Windsor
- Laboratory of Molecular Medicine, Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen C. Harrison
- Laboratory of Molecular Medicine, Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Garnett Kelsoe
- Department of Immunology, Duke University, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Wakui M, Uwamino Y, Kurafuji T, Noguchi M, Ohno A, Yokota H, Kikuchi H, Hasegawa N, Saya H, Murata M. Assessment of humoral responses in COVID-19 using various quantitative antibody tests. Ann Clin Biochem 2021; 58:368-376. [PMID: 33730868 PMCID: PMC8685744 DOI: 10.1177/00045632211006740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Quantitative antibody tests are expected to be useful in diagnostics of COVID-19 and investigation of herd immunity against SARS-CoV-2. To make it proper to perform them, understanding of the immunological aspects is critically important. The present study aimed to assess humoral responses in COVID-19 using various quantitative antibody tests. Methods Four quantitative antibody tests that are different in targeted antigens, detectable immunoglobulin classes and avidity were used. Diagnosis was confirmed by RT-PCR for SARS-CoV-2 detection. Antibody titres of 117 samples collected from 24 COVID-19 patients and 23 non-COVID-19 patients were measured to evaluate correlations between different tests. For 24 COVID-19 patients, antibody titres measured at various time points after the onset or the RT-PCR diagnosis were subjected to assessment of humoral responses. Results Correlations between tests were observed to some degree, although there were discrepancies putatively due to differences in measurement principle. Seronegative COVID-19 was diagnosed for some patients, in whom antibody titres were less than the cut-off value in each test throughout the time courses. IgG seroconversion without prior IgM seroconversion most frequently occurred, while predominance of IgM responses over IgG responses was observed in some severe cases. Viral burdens estimated according to threshold cycle values at the RT-PCR seemed to impact antibody responses. Conclusions The results provide insights into the nature of humoral responses to SARS-CoV-2 and diagnostic performance of antibody tests.
Collapse
Affiliation(s)
- Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Uwamino
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | | | - Masayo Noguchi
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | - Akemi Ohno
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | | | - Haruhito Kikuchi
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuru Murata
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Nakagawa R, Calado DP. Positive Selection in the Light Zone of Germinal Centers. Front Immunol 2021; 12:661678. [PMID: 33868314 PMCID: PMC8044421 DOI: 10.3389/fimmu.2021.661678] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/16/2021] [Indexed: 12/29/2022] Open
Abstract
Germinal centers (GCs) are essential sites for the production of high-affinity antibody secreting plasma cells (PCs) and memory-B cells (MBCs), which form the framework of vaccination. Affinity maturation and permissive selection in GCs are key for the production of PCs and MBCs, respectively. For these purposes, GCs positively select “fit” cells in the light zone of the GC and instructs them for one of three known B cell fates: PCs, MBCs and persistent GC-B cells as dark zone entrants. In this review, we provide an overview of the positive selection process and discuss its mechanisms and how B cell fates are instructed.
Collapse
Affiliation(s)
- Rinako Nakagawa
- Immunity and Cancer Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Dinis Pedro Calado
- Immunity and Cancer Laboratory, The Francis Crick Institute, London, United Kingdom.,Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
7
|
Bigas A, Zanoni I, Hepworth MR, Eisenbarth SC, Masters SL, Kipnis J, Vinuesa CG, Good-Jacobson KL, Tangye SG, Yamazaki S, Hivroz C, Tait Wojno E, Shulman Z, Colonna M. JEM career launchpad. J Exp Med 2021; 218:e20202509. [PMID: 33464291 PMCID: PMC7814352 DOI: 10.1084/jem.20202509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
JEM has been a launching pad for scientific careers since its inception. Here is a collection of testimonials attesting to the diversity of the scientific community it serves.
Collapse
|
8
|
Kuraoka M, Adachi Y, Takahashi Y. Hide and seek: interplay between influenza viruses and B cells. Int Immunol 2020; 32:605-611. [PMID: 32304215 PMCID: PMC7478158 DOI: 10.1093/intimm/dxaa028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Influenza virus constantly acquires genetic mutations/reassortment in the major surface protein, hemagglutinin (HA), resulting in the generation of strains with antigenic variations. There are, however, HA epitopes that are conserved across influenza viruses and are targeted by broadly protective antibodies. A goal for the next-generation influenza vaccines is to stimulate B-cell responses against such conserved epitopes in order to provide broad protection against divergent influenza viruses. Broadly protective B cells, however, are not easily activated by HA antigens with native structure, because the virus has multiple strategies to escape from the humoral immune responses directed to the conserved epitopes. One such strategy is to hide the conserved epitopes from the B-cell surveillance by steric hindrance. Technical advancement in the analysis of the human B-cell antigen receptor (BCR) repertoire has dissected the BCRs to HA epitopes that are hidden in the native structure but are targeted by broadly protective antibodies. We describe here the characterization and function of broadly protective antibodies and strategies that enable B cells to seek these hidden epitopes, with potential implications for the development of universal influenza vaccines.
Collapse
Affiliation(s)
| | - Yu Adachi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
9
|
Brooks JF, Murphy PR, Barber JEM, Wells JW, Steptoe RJ. Peripheral Tolerance Checkpoints Imposed by Ubiquitous Antigen Expression Limit Antigen-Specific B Cell Responses under Strongly Immunogenic Conditions. THE JOURNAL OF IMMUNOLOGY 2020; 205:1239-1247. [DOI: 10.4049/jimmunol.2000377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/06/2020] [Indexed: 01/17/2023]
|
10
|
Molari M, Eyer K, Baudry J, Cocco S, Monasson R. Quantitative modeling of the effect of antigen dosage on B-cell affinity distributions in maturating germinal centers. eLife 2020; 9:e55678. [PMID: 32538783 PMCID: PMC7360369 DOI: 10.7554/elife.55678] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Affinity maturation is a complex dynamical process allowing the immune system to generate antibodies capable of recognizing antigens. We introduce a model for the evolution of the distribution of affinities across the antibody population in germinal centers. The model is amenable to detailed mathematical analysis and gives insight on the mechanisms through which antigen availability controls the rate of maturation and the expansion of the antibody population. It is also capable, upon maximum-likelihood inference of the parameters, to reproduce accurately the distributions of affinities of IgG-secreting cells we measure in mice immunized against Tetanus Toxoid under largely varying conditions (antigen dosage, delay between injections). Both model and experiments show that the average population affinity depends non-monotonically on the antigen dosage. We show that combining quantitative modeling and statistical inference is a concrete way to investigate biological processes underlying affinity maturation (such as selection permissiveness), hardly accessible through measurements.
Collapse
Affiliation(s)
- Marco Molari
- Laboratoire de Physique de l’École Normale Supérieure, ENS, PSL University, CNRS UMR8023, Sorbonne Université, Université Paris-Diderot, Sorbonne Paris CitéParisFrance
| | - Klaus Eyer
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Jean Baudry
- Laboratoire Colloides et Materiaux Divises (LCMD), Chemistry, Biology and Innovation (CBI), ESPCI, PSL Research and CNRSParisFrance
| | - Simona Cocco
- Laboratoire de Physique de l’École Normale Supérieure, ENS, PSL University, CNRS UMR8023, Sorbonne Université, Université Paris-Diderot, Sorbonne Paris CitéParisFrance
| | - Rémi Monasson
- Laboratoire de Physique de l’École Normale Supérieure, ENS, PSL University, CNRS UMR8023, Sorbonne Université, Université Paris-Diderot, Sorbonne Paris CitéParisFrance
| |
Collapse
|
11
|
Lettau M, Wiedemann A, Schrezenmeier EV, Giesecke-Thiel C, Dörner T. Human CD27+ memory B cells colonize a superficial follicular zone in the palatine tonsils with similarities to the spleen. A multicolor immunofluorescence study of lymphoid tissue. PLoS One 2020; 15:e0229778. [PMID: 32187186 PMCID: PMC7080255 DOI: 10.1371/journal.pone.0229778] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Memory B cell (mBC) induction and maintenance is one of the keys to long-term protective humoral immunity. MBCs are fundamental to successful medical interventions such as vaccinations and therapy in autoimmunity. However, their lifestyle and anatomic residence remain enigmatic in humans. Extrapolation from animal studies serves as a conceptual basis but might be misleading due to major anatomical distinctions between species. METHODS AND FINDINGS Multicolor immunofluorescence stainings on fixed and unfixed frozen tissue sections were established using primary antibodies coupled to haptens and secondary signal amplification. The simultaneous detection of five different fluorescence signals enabled the localization and characterization of human CD27+CD20+Ki67- mBCs for the first time within one section using laser scanning microscopy. As a result, human tonsillar mBCs were initially identified within their complex microenvironment and their relative location to naïve B cells, plasma cells and T cells could be directly studied and compared to the human splenic mBC niche. In all investigated tonsils (n = 15), mBCs appeared to be not only located in a so far subepithelial defined area but were also follicle associated with a previous undescribed gradual decline towards the follicular mantle comparable to human spleen. However, mBC areas around secondary follicles with large germinal centers (GCs) in tonsils showed interruptions and a general widening towards the epithelium while in spleen the mBC-containing marginal zones (MZ) around smaller GCs were relatively broad and symmetrical. Considerably fewer IgM+IgD+/- pre-switch compared to IgA+ or IgG+ post-switch mBCs were detected in tonsils in contrast to spleen. CONCLUSIONS This study extends existing insights into the anatomic residence of human mBCs showing structural similarities of the superficial follicular area in human spleen and tonsil. Our data support the debate of renaming the human splenic MZ to 'superficial zone' in order to be aware of the differences in rodents and, moreover, to consider this term equally for the human palatine tonsil.
Collapse
Affiliation(s)
- Marie Lettau
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Annika Wiedemann
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Eva Vanessa Schrezenmeier
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| | - Claudia Giesecke-Thiel
- Department of Rheumatology and Clinical Immunology, Formerly at the Charité University Medicine Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- * E-mail:
| |
Collapse
|
12
|
Wholey WY, Mueller JL, Tan C, Brooks JF, Zikherman J, Cheng W. Synthetic Liposomal Mimics of Biological Viruses for the Study of Immune Responses to Infection and Vaccination. Bioconjug Chem 2020; 31:685-697. [PMID: 31940172 DOI: 10.1021/acs.bioconjchem.9b00825] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human viruses possess very complex supramolecular structures. Both icosahedral and enveloped viruses typically display an array of viral-encoded protein antigens at varied spatial densities on the viral particle surface. The viral nucleic acid genome, on the other hand, is encapsulated inside the viral particle. Although both the surface antigen and the interior nucleic acids could independently produce immunological responses, how B cells integrate these two types of signals and respond to a typical virus particle to initiate activation is not well understood at a molecular level. The study of these fundamental biological processes would benefit from the development of viral structural mimics that are well constructed to incorporate both quantitative and qualitative viral features for presentation to B cells. These novel tools would enable researchers to systematically dissect the underlying processes. Here we report the development of such particulate antigens based on liposomes engineered to display a model protein antigen, hen egg lysozyme (HEL). We developed methods to overexpress and purify various affinity mutants of HEL from E. coli. We conjugated the purified recombinant HEL proteins onto the surface of a virion-sized liposome in an orientation-specific manner at defined spatial densities and also encapsulated nucleic acid molecules into the interior of the liposome. Both the chemical conjugation of the HEL antigen on liposome surfaces and the encapsulation of nucleic acids were stable under physiologically relevant conditions. These liposomes elicited antigen-specific B-cell responses in vitro, which validate these supramolecular structures as a novel and effective approach to mimic and systematically isolate the role of essential viral features in directing the B-cell response to particulate antigens.
Collapse
Affiliation(s)
- Wei-Yun Wholey
- Department of Pharmaceutical Sciences, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
| | - James L Mueller
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, California 94143, United States
| | - Corey Tan
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, California 94143, United States
| | - Jeremy F Brooks
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, California 94143, United States
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, California 94143, United States
| | - Wei Cheng
- Department of Pharmaceutical Sciences, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States.,Department of Biological Chemistry, University of Michigan Medical School, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
13
|
Shlomchik MJ, Luo W, Weisel F. Linking signaling and selection in the germinal center. Immunol Rev 2019; 288:49-63. [PMID: 30874353 DOI: 10.1111/imr.12744] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 12/24/2022]
Abstract
Germinal centers (GC) are sites of rapid B-cell proliferation in response to certain types of immunization. They arise in about 1 week and can persist for several months. In GCs, B cells differentiate in a unique way and begin to undergo somatic mutation of the Ig V regions at a high rate. GC B cells (GCBC) thus undergo clonal diversification that can affect the affinity of the newly mutant B-cell receptor (BCR) for its driving antigen. Through processes that are still poorly understood, GCBC with higher affinity are selectively expanded while those with mutations that inactivate the BCR are lost. In addition, at various times during the extended GC reaction, some GCBC undergo differentiation into either long-lived memory B cells (MBC) or plasma cells. The cellular and molecular signals that govern these fate decisions are not well-understood, but are an active area of research in multiple laboratories. In this review, we cover both the history of this field and focus on recent work that has helped to elucidate the signals and molecules, such as key transcription factors, that coordinate both positive selection as well as differentiation of GCBC.
Collapse
Affiliation(s)
- Mark J Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wei Luo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Florian Weisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Haberman AM, Gonzalez DG, Wong P, Zhang TT, Kerfoot SM. Germinal center B cell initiation, GC maturation, and the coevolution of its stromal cell niches. Immunol Rev 2019; 288:10-27. [PMID: 30874342 DOI: 10.1111/imr.12731] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022]
Abstract
Throughout the developing GC response, B cell survival and fate choices made at the single cell level are dependent on signals received largely through interactions with other cells, often with cognate T cells. The type of signals that a given B cell can encounter is dictated by its location within tissue microarchitecture. The focus of this review is on the initiation and evolution of the GC response at the earliest time points. Here, we review the key factors influencing the progression of GC B cell differentiation that are both stage and context dependent. Finally, we describe the coevolution of niches within and surrounding the GC that influence the outcome of the GC response.
Collapse
Affiliation(s)
- Ann M Haberman
- Department of Immunobiology, Yale University, New Haven, Connecticut.,Department of Laboratory Medicine, Yale University, New Haven, Connecticut
| | - David G Gonzalez
- Department of Immunobiology, Yale University, New Haven, Connecticut.,Department of Genetics, Yale University, New Haven, Connecticut
| | - Patrick Wong
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | - Ting-Ting Zhang
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | - Steven M Kerfoot
- Department of Microbiology and Immunology, Western University, London, ON, Canada
| |
Collapse
|
15
|
Finney J, Watanabe A, Kelsoe G, Kuraoka M. Minding the gap: The impact of B-cell tolerance on the microbial antibody repertoire. Immunol Rev 2019; 292:24-36. [PMID: 31559648 PMCID: PMC6935408 DOI: 10.1111/imr.12805] [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: 06/15/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022]
Abstract
B lymphocytes must respond to vast numbers of foreign antigens, including those of microbial pathogens. To do so, developing B cells use combinatorial joining of V-, D-, and J-gene segments to generate an extraordinarily diverse repertoire of B-cell antigen receptors (BCRs). Unsurprisingly, a large fraction of this initial BCR repertoire reacts to self-antigens, and these "forbidden" B cells are culled by immunological tolerance from mature B-cell populations. While culling of autoreactive BCRs mitigates the risk of autoimmunity, it also opens gaps in the BCR repertoire, which are exploited by pathogens that mimic the forbidden self-epitopes. Consequently, immunological tolerance, necessary for averting autoimmune disease, also acts to limit effective microbial immunity. In this brief review, we recount the evidence for the linkage of tolerance and impaired microbial immunity, consider the implications of this linkage for vaccine development, and discuss modulating tolerance as a potential strategy for strengthening humoral immune responses.
Collapse
Affiliation(s)
- Joel Finney
- Department of Immunology, Duke University, Durham, NC, USA
| | - Akiko Watanabe
- Department of Immunology, Duke University, Durham, NC, USA
| | - Garnett Kelsoe
- Department of Immunology, Duke University, Durham, NC, USA
- Duke University Human Vaccine Institute, Duke University, Durham, NC, USA
| | | |
Collapse
|
16
|
Tan C, Noviski M, Huizar J, Zikherman J. Self-reactivity on a spectrum: A sliding scale of peripheral B cell tolerance. Immunol Rev 2019; 292:37-60. [PMID: 31631352 DOI: 10.1111/imr.12818] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/02/2019] [Indexed: 12/16/2022]
Abstract
Efficient mechanisms of central tolerance, including receptor editing and deletion, prevent highly self-reactive B cell receptors (BCRs) from populating the periphery. Despite this, modest self-reactivity persists in (and may even be actively selected into) the mature B cell repertoire. In this review, we discuss new insights into mechanisms of peripheral B cell tolerance that restrain mature B cells from mounting inappropriate responses to endogenous antigens, and place recent work into historical context. In particular, we discuss new findings that have arisen from application of a novel in vivo reporter of BCR signaling, Nur77-eGFP, expression of which scales with the degree of self-reactivity in both monoclonal and polyclonal B cell repertoires. We discuss new and historical evidence that self-reactivity is not just tolerated, but actively selected into the peripheral repertoire. We review recent progress in understanding how dual expression of the IgM and IgD BCR isotypes on mature naive follicular B cells tunes responsiveness to endogenous antigen recognition, and discuss how this may be integrated with other features of clonal anergy. Finally, we discuss how expression of Nur77 itself couples chronic antigen stimulation with B cell tolerance.
Collapse
Affiliation(s)
- Corey Tan
- Biomedical Sciences (BMS) Graduate Program, University of California, San Francisco, CA, USA
| | - Mark Noviski
- Biomedical Sciences (BMS) Graduate Program, University of California, San Francisco, CA, USA.,Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, University of California, San Francisco, CA, USA
| | - John Huizar
- School of Medicine, HHMI Medical Fellows Program, University of California, San Francisco, CA, USA
| | - Julie Zikherman
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, University of California, San Francisco, CA, USA
| |
Collapse
|
17
|
Kwak K, Akkaya M, Pierce SK. B cell signaling in context. Nat Immunol 2019; 20:963-969. [PMID: 31285625 DOI: 10.1038/s41590-019-0427-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Over the past several decades, B cell antigen receptor (BCR)-induced signaling pathways have been described in extraordinary molecular detail, mainly from studies of B cell responses to antigens in vitro. BCR signaling has been shown to govern the initiation of transcriptional programs associated with B cell activation and fate decisions, as well as the BCR-dependent processing of antigen and presentation of antigen to T cells. However, although the potential of the BCR to orchestrate B cell behavior was known, there was no clear appreciation of the context in which B cells signal in secondary lymphoid organs in vivo or how that context influences signaling. In this Review, we describe the current view of the cellular consequences of BCR signaling and advances in the understanding of B cell signaling in context in vivo.
Collapse
Affiliation(s)
- Kihyuck Kwak
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
18
|
Silva M, Nguyen TH, Philbrook P, Chu M, Sears O, Hatfield S, Abbott RK, Kelsoe G, Sitkovsky MV. Targeted Elimination of Immunodominant B Cells Drives the Germinal Center Reaction toward Subdominant Epitopes. Cell Rep 2019; 21:3672-3680. [PMID: 29281817 PMCID: PMC5771444 DOI: 10.1016/j.celrep.2017.12.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/14/2017] [Accepted: 12/01/2017] [Indexed: 01/12/2023] Open
Abstract
Rapidly evolving pathogens such as HIV or influenza can quickly mutate their antigenic profiles, reducing the efficacy of conventional vaccines. Despite this challenge, functionally required epitopes are highly conserved among heterologous viral strains and represent a key vulnerability that could be targeted during vaccine development. As the antigenicity of these conserved epitopes is frequently subdominant, there is a critical need for innovative vaccination strategies designed to target these neutralizing epitopes. Here, we immunized mice with antigens containing discrete immunodominant and subdominant moieties and show that treatment with soluble heterologous antigen bearing only the immunodominant epitope selectively suppresses these germinal center (GC) B cells. By exploiting this intrinsic tolerance mechanism, we promote the expansion of subdominant B cells in the GC and the subsequent long-lived components of the humoral response. We propose that this strategy may be applied to elicit preferential expansion of subdominant B cells that recognize weakly immunogenic epitopes on microbial pathogens.
Collapse
Affiliation(s)
- Murillo Silva
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA.
| | - Thao H Nguyen
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Phaethon Philbrook
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Matthew Chu
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Olivia Sears
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Stephen Hatfield
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Robert K Abbott
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | - Garnett Kelsoe
- Department of Immunology and Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Michail V Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Finney J, Kelsoe G. Poly- and autoreactivity of HIV-1 bNAbs: implications for vaccine design. Retrovirology 2018; 15:53. [PMID: 30055635 PMCID: PMC6064052 DOI: 10.1186/s12977-018-0435-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/23/2018] [Indexed: 01/17/2023] Open
Abstract
A central puzzle in HIV-1 research is the inability of vaccination or even infection to reliably elicit humoral responses against broadly neutralizing epitopes in the HIV-1 envelope protein. In infected individuals, broadly neutralizing antibodies (bNAbs) do arise in a substantial minority, but only after 2 or more years of chronic infection. All known bNAbs possess at least one of three traits: a high frequency of somatic hypermutation, a long third complementarity determining region in the antibody heavy chain (HCDR3), or significant poly- or autoreactivity. Collectively, these observations suggest a plausible explanation for the rarity of many types of bNAbs: namely, that their generation is blocked by immunological tolerance or immune response checkpoints, thereby mandating that B cells take a tortuous path of somatic evolution over several years to achieve broadly neutralizing activity. In this brief review, we discuss the evidence for this tolerance hypothesis, its implications for HIV-1 vaccine design, and potential ways to access normally forbidden compartments of the antibody repertoire by modulating or circumventing tolerance controls.
Collapse
Affiliation(s)
- Joel Finney
- Department of Immunology, Duke University, DUMC 3010, Durham, NC, 27710, USA
| | - Garnett Kelsoe
- Department of Immunology, Duke University, DUMC 3010, Durham, NC, 27710, USA. .,Human Vaccine Institute, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
20
|
Roders N, Herr F, Ambroise G, Thaunat O, Portier A, Vazquez A, Durrbach A. SYK Inhibition Induces Apoptosis in Germinal Center-Like B Cells by Modulating the Antiapoptotic Protein Myeloid Cell Leukemia-1, Affecting B-Cell Activation and Antibody Production. Front Immunol 2018; 9:787. [PMID: 29740433 PMCID: PMC5928208 DOI: 10.3389/fimmu.2018.00787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/29/2018] [Indexed: 12/24/2022] Open
Abstract
B cells play a major role in the antibody-mediated rejection (AMR) of solid organ transplants, a major public health concern. The germinal center (GC) is involved in the generation of donor-specific antibody-producing plasma cells and memory B cells, which are often poorly controlled by current treatments. Myeloid cell leukemia-1 (Mcl-1), an antiapoptotic member of the B-cell lymphoma-2 family, is essential for maintenance of the GC reaction and B-cell differentiation. During chronic AMR (cAMR), tertiary lymphoid structures resembling GCs appear in the rejected organ, suggesting local lymphoid neogenesis. We report the infiltration of the kidneys with B cells expressing Mcl-1 in patients with cAMR. We modulated GC viability by impairing B-cell receptor signaling, by spleen tyrosine kinase (SYK) inhibition. SYK inhibition lowers viability and Mcl-1 protein levels in Burkitt's lymphoma cell lines. This downregulation of Mcl-1 is coordinated at the transcriptional level, possibly by signal transducer and activator of transcription 3 (STAT3), as shown by (1) the impaired translocation of STAT3 to the nucleus following SYK inhibition, and (2) the lower levels of Mcl-1 transcription upon STAT3 inhibition. Mcl-1 overproduction prevented cells from entering apoptosis following SYK inhibition. In vitro studies with primary tonsillar B cells confirmed that SYK inhibition impaired cell survival and decreased Mcl-1 protein levels. It also impaired B-cell activation and immunoglobulin G secretion by tonsillar B cells. These findings suggest that the SYK-Mcl-1 pathway could be targeted, to improve graft survival by manipulating the humoral immune response.
Collapse
Affiliation(s)
- Nathalie Roders
- Institut Francilien de Recherche en Nephrologie et Transplantation (IFRNT), Service de Néphrologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,INSERM UMRS-MD 1197, Villejuif, France.,Université Paris Sud, Orsay, France
| | - Florence Herr
- Institut Francilien de Recherche en Nephrologie et Transplantation (IFRNT), Service de Néphrologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,INSERM UMRS-MD 1197, Villejuif, France.,Université Paris Sud, Orsay, France
| | | | - Olivier Thaunat
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot University Hospital, Lyon, France.,Claude Bernard University Lyon 1, Lyon, France
| | - Alain Portier
- INSERM UMRS-MD 1197, Villejuif, France.,Université Paris Sud, Orsay, France
| | - Aimé Vazquez
- INSERM UMRS-MD 1197, Villejuif, France.,Université Paris Sud, Orsay, France
| | - Antoine Durrbach
- Institut Francilien de Recherche en Nephrologie et Transplantation (IFRNT), Service de Néphrologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,INSERM UMRS-MD 1197, Villejuif, France.,Université Paris Sud, Orsay, France
| |
Collapse
|
21
|
Critical influences on the pathogenesis of follicular lymphoma. Blood 2018; 131:2297-2306. [PMID: 29666116 DOI: 10.1182/blood-2017-11-764365] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/28/2017] [Indexed: 12/15/2022] Open
Abstract
The development of follicular lymphoma (FL) from a founder B cell with an upregulation of B-cell lymphoma 2 (BCL2), via the t(14;18) translocation, to a proliferating clone, poised to undergo further transformation to an aggressive lymphoma, illustrates the opportunistic Darwinian process of tumorigenesis. Protection against apoptosis allows an innocent cell to persist and divide, with dangerous accumulation of further mutational changes, commonly involving inactivation of chromatin-modifying genes. But this is not all. FL cells reflect normal B cells in relying on expression of surface immunoglobulin. In doing so, they add another supportive mechanism by exploiting the natural process of somatic hypermutation of the IGV genes. Positive selection of motifs for addition of glycan into the antigen-binding sites of virtually all cases, and the placement of unusual mannoses in those sites, reveals a posttranslational strategy to engage the microenvironment. A bridge between mannosylated surface immunoglobulin of FL cells and macrophage-expressed dendritic cell-specific ICAM-3-grabbing nonintegrin produces a persistent low-level signal that appears essential for life in the hostile germinal center. Early-stage FL therefore requires a triad of changes: protection from apoptosis, mutations in chromatin modifiers, and an ability to interact with lectin-expressing macrophages. These changes are common and persistent. Genetic/epigenetic analysis is providing important data but investigation of the posttranslational landscape is the next challenge. We have one glimpse of its operation via the influence of added glycan on the B-cell receptor of FL. The consequential interaction with environmental lectins illustrates how posttranslational modifications can be exploited by tumor cells, and could lead to new approaches to therapy.
Collapse
|
22
|
Abstract
Maintenance of immunological self-tolerance requires lymphocytes carrying self-reactive antigen receptors to be selectively prevented from mounting destructive or inflammatory effector responses. Classically, self-tolerance is viewed in terms of the removal, editing, or silencing of B and T cells that have formed self-reactive antigen receptors during their early development. However, B cells activated by foreign antigen can enter germinal centers (GCs), where they further modify their antigen receptor by somatic hypermutation (SHM) of their immunoglobulin genes. The inevitable emergence of activated, self-reactive GC B cells presents a unique challenge to the maintenance of self-tolerance that must be rapidly countered to avoid autoantibody production. Here we discuss current knowledge of the mechanisms that enforce B cell self-tolerance, with particular focus on the control of self-reactive GC B cells. We also consider how self-reactive GC B cells can escape self-tolerance to initiate autoantibody production or instead be redeemed via SHM and used in productive antibody responses.
Collapse
Affiliation(s)
- Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia; , .,Faculty of Medicine, UNSW Sydney, New South Wales 2052, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia; , .,Faculty of Medicine, UNSW Sydney, New South Wales 2052, Australia
| |
Collapse
|
23
|
Tan Q, Tai N, Li Y, Pearson J, Pennetti S, Zhou Z, Wong FS, Wen L. Activation-induced cytidine deaminase deficiency accelerates autoimmune diabetes in NOD mice. JCI Insight 2018; 3:95882. [PMID: 29321370 DOI: 10.1172/jci.insight.95882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/05/2017] [Indexed: 01/15/2023] Open
Abstract
B cells play an important role in type 1 diabetes (T1D) development. However, the role of B cell activation-induced cytidine deaminase (AID) in diabetes development is not clear. We hypothesized that AID is important in the immunopathogenesis of T1D. To test this hypothesis, we generated AID-deficient (AID-/-) NOD mice. We found that AID-/-NOD mice developed accelerated T1D, with worse insulitis and high levels of anti-insulin autoantibody in the circulation. Interestingly, neither maternal IgG transferred through placenta, nor IgA transferred through milk affected the accelerated diabetes development. AID-/-NOD mice showed increased activation and proliferation of B and T cells. We found enhanced T-B cell interactions in AID-/-NOD mice, with increased T-bet and IFN-γ expression in CD4+ T cells in the presence of AID-/- B cells. Moreover, excessive lymphoid expansion was observed in AID-/-NOD mice. Importantly, antigen-specific BDC2.5 CD4+ T cells caused more rapid onset of diabetes when cotransferred with AID-/- B cells than when cotransferred with AID+/+ B cells. Thus, our study provides insights into the role of AID in T1D. Our data also suggest that AID is a negative regulator of immune tolerance and ablation of AID can lead to exacerbated islet autoimmunity and accelerated T1D development.
Collapse
Affiliation(s)
- Qiyuan Tan
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China.,Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yangyang Li
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Endocrinology, Jilin University, Changchun, China
| | - James Pearson
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sean Pennetti
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,School of Medicine, Quinnipiac University, North Haven, Connecticut, USA
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Li Wen
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China.,Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Lycke NY, Bemark M. The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol 2017; 10:1361-1374. [PMID: 28745325 DOI: 10.1038/mi.2017.62] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/20/2017] [Indexed: 02/04/2023]
Abstract
The majority of activated B cells differentiate into IgA plasma cells, with the gut being the largest producer of immunoglobulin in the body. Secretory IgA antibodies have numerous critical functions of which protection against infections and the role for establishing a healthy microbiota appear most important. Expanding our knowledge of the regulation of IgA B-cell responses and how effective mucosal vaccines can be designed are of critical importance. Here we discuss recent developments in the field that shed light on the uniqueness and complexity of mucosal IgA responses and the control of protective IgA responses in the gut, specifically.
Collapse
Affiliation(s)
- N Y Lycke
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - M Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Mayer CT, Gazumyan A, Kara EE, Gitlin AD, Golijanin J, Viant C, Pai J, Oliveira TY, Wang Q, Escolano A, Medina-Ramirez M, Sanders RW, Nussenzweig MC. The microanatomic segregation of selection by apoptosis in the germinal center. Science 2017; 358:science.aao2602. [PMID: 28935768 DOI: 10.1126/science.aao2602] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/29/2017] [Indexed: 01/04/2023]
Abstract
B cells undergo rapid cell division and affinity maturation in anatomically distinct sites in lymphoid organs called germinal centers (GCs). Homeostasis is maintained in part by B cell apoptosis. However, the precise contribution of apoptosis to GC biology and selection is not well defined. We developed apoptosis-indicator mice and used them to visualize, purify, and characterize dying GC B cells. Apoptosis is prevalent in the GC, with up to half of all GC B cells dying every 6 hours. Moreover, programmed cell death is differentially regulated in the light zone and the dark zone: Light-zone B cells die by default if they are not positively selected, whereas dark-zone cells die when their antigen receptors are damaged by activation-induced cytidine deaminase.
Collapse
Affiliation(s)
- Christian T Mayer
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Ervin E Kara
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Alexander D Gitlin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jovana Golijanin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Charlotte Viant
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Joy Pai
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Qiao Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Amelia Escolano
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Max Medina-Ramirez
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, Netherlands.,Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA. .,Howard Hughes Medical Institute (HHMI), The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
26
|
Degn SE, van der Poel CE, Firl DJ, Ayoglu B, Al Qureshah FA, Bajic G, Mesin L, Reynaud CA, Weill JC, Utz PJ, Victora GD, Carroll MC. Clonal Evolution of Autoreactive Germinal Centers. Cell 2017; 170:913-926.e19. [PMID: 28841417 DOI: 10.1016/j.cell.2017.07.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/17/2017] [Accepted: 07/14/2017] [Indexed: 01/08/2023]
Abstract
Germinal centers (GCs) are the primary sites of clonal B cell expansion and affinity maturation, directing the production of high-affinity antibodies. This response is a central driver of pathogenesis in autoimmune diseases, such as systemic lupus erythematosus (SLE), but the natural history of autoreactive GCs remains unclear. Here, we present a novel mouse model where the presence of a single autoreactive B cell clone drives the TLR7-dependent activation, expansion, and differentiation of other autoreactive B cells in spontaneous GCs. Once tolerance was broken for one self-antigen, autoreactive GCs generated B cells targeting other self-antigens. GCs became independent of the initial clone and evolved toward dominance of individual clonal lineages, indicating affinity maturation. This process produced serum autoantibodies to a breadth of self-antigens, leading to antibody deposition in the kidneys. Our data provide insight into the maturation of the self-reactive B cell response, contextualizing the epitope spreading observed in autoimmune disease.
Collapse
Affiliation(s)
- Søren E Degn
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Cees E van der Poel
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J Firl
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Burcu Ayoglu
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Fahd A Al Qureshah
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Goran Bajic
- Laboratory of Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Luka Mesin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Claude-Agnès Reynaud
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMS 8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Jean-Claude Weill
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMS 8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Paul J Utz
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Gabriel D Victora
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Abstract
Induction of broadly neutralizing antibodies (bNAbs) is a major goal of HIV vaccine development. BNAbs are made during HIV infection by a subset of individuals but currently cannot be induced in the setting of vaccination. Considerable progress has been made recently in understanding host immunologic controls of bNAb induction and maturation in the setting of HIV infection, and point to key roles for both central and peripheral immunologic tolerance mechanisms in limiting bnAb development. Immune tolerance checkpoint inhibition has been transformative in promotion of anti-tumor CD8 T-cell responses in the treatment of certain malignancies. Here, we review the evidence for host controls of bNAb responses, and discuss strategies for the transient modulation of immune responses with vaccines toward the goal of enhancing germinal center B-cell responses to favor bNAb B-cell lineages and to foster their maturation to full neutralization potency.
Collapse
Affiliation(s)
- Garnett Kelsoe
- Departments of Immunology and Medicine, Duke University School of Medicine, Duke Human Vaccine Institute, Durham, NC, 27710, USA
| | - Barton F Haynes
- Departments of Immunology and Medicine, Duke University School of Medicine, Duke Human Vaccine Institute, Durham, NC, 27710, USA
| |
Collapse
|
28
|
Hannestad K, Scott H. A Nonadjuvanted IgG2a Monoclonal Antibody against Nucleosomes Elicits Potent T Cell-Dependent, Idiotype-Specific IgG1 Responses and Glomerular IgG1/IgG2a Deposits in Normal Mice. THE JOURNAL OF IMMUNOLOGY 2017; 199:489-500. [PMID: 28592426 DOI: 10.4049/jimmunol.1600718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/04/2017] [Indexed: 11/19/2022]
Abstract
Idiotypes (Ids) are unique epitopes of Ab V regions and can trigger anti-Id immune responses, but immunization with several nonadjuvanted isologous IgG mAbs has induced tolerance to their Ids. We immunized non-lupus-prone mice with 11 allotype "a" of IgG2a (IgG2aa) and 4 IgG2c nonadjuvanted, isologous mAbs purified from serum-free medium. Of five IgG2aa mAbs with specificity for nucleosomes, the repeating histone-DNA subunit of chromatin, four elicited an IgG1 anti-mAb response and one mAb was nonimmunogenic. In contrast, none of six IgG2aa mAbs with unknown specificity triggered anti-mAb responses. The data suggested a link between immunogenicity and specificity for nucleosomes. One anti-nucleosome IgG2aa mAb, termed 3F7.A10, copurified with self-histones and was a potent immunogen for BALB/c mice. The response against IgG2aa 3F7.A10 was CD4+ Th cell-dependent, dominated by the IgG1 subclass, and Id specific. Ultracentrifugation converted the purified 3F7.A10 mAb into a weak immunogen, suggesting that the mAb had formed immunogenicity-enhancing immune complexes (ICs) with nucleosomal Ags during cell culture. BALB/c mice injected with viable MHC-incompatible 3F7.A10 hybridoma cells grown in serum-free medium mounted strong anti-Id responses. TLR9-deficient mice responded significantly weaker to Id-3F7.A10 than did TLR9-sufficient mice, suggesting that the cognate BCR efficiently internalizes the Id in an IC with nucleosomes. Passive transfer of IgG2aa 3F7.A10 to BALB/c mice with high titers of IgG1 anti-3F7.A10 led to glomerular deposits of IgG1/IgG2a complexes. The immunogenicity of Id-3F7.A10 raises the possibility that diverse Ids of nucleosome-specific Abs form ICs with nucleosomes released from dying cells and elicit spontaneous formation of anti-Id Abs in vivo.
Collapse
Affiliation(s)
- Kristian Hannestad
- Department of Immunology, Oslo University Hospital, University of Oslo, 0372 Oslo, Norway; and
| | - Helge Scott
- Institute of Pathology, Oslo University Hospital, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
29
|
Verkoczy L. Humanized Immunoglobulin Mice: Models for HIV Vaccine Testing and Studying the Broadly Neutralizing Antibody Problem. Adv Immunol 2017; 134:235-352. [PMID: 28413022 PMCID: PMC5914178 DOI: 10.1016/bs.ai.2017.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A vaccine that can effectively prevent HIV-1 transmission remains paramount to ending the HIV pandemic, but to do so, will likely need to induce broadly neutralizing antibody (bnAb) responses. A major technical hurdle toward achieving this goal has been a shortage of animal models with the ability to systematically pinpoint roadblocks to bnAb induction and to rank vaccine strategies based on their ability to stimulate bnAb development. Over the past 6 years, immunoglobulin (Ig) knock-in (KI) technology has been leveraged to express bnAbs in mice, an approach that has enabled elucidation of various B-cell tolerance mechanisms limiting bnAb production and evaluation of strategies to circumvent such processes. From these studies, in conjunction with the wealth of information recently obtained regarding the evolutionary pathways and paratopes/epitopes of multiple bnAbs, it has become clear that the very features of bnAbs desired for their function will be problematic to elicit by traditional vaccine paradigms, necessitating more iterative testing of new vaccine concepts. To meet this need, novel bnAb KI models have now been engineered to express either inferred prerearranged V(D)J exons (or unrearranged germline V, D, or J segments that can be assembled into functional rearranged V(D)J exons) encoding predecessors of mature bnAbs. One encouraging approach that has materialized from studies using such newer models is sequential administration of immunogens designed to bind progressively more mature bnAb predecessors. In this review, insights into the regulation and induction of bnAbs based on the use of KI models will be discussed, as will new Ig KI approaches for higher-throughput production and/or altering expression of bnAbs in vivo, so as to further enable vaccine-guided bnAb induction studies.
Collapse
Affiliation(s)
- Laurent Verkoczy
- Duke University Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
30
|
Wiehe K, Nicely NI, Lockwood B, Kuraoka M, Anasti K, Arora S, Bowman CM, Stolarchuk C, Parks R, Lloyd KE, Xia SM, Duffy R, Shen X, Kyratsous CA, Macdonald LE, Murphy AJ, Scearce RM, Moody MA, Alam SM, Verkoczy L, Tomaras GD, Kelsoe G, Haynes BF. Immunodominance of Antibody Recognition of the HIV Envelope V2 Region in Ig-Humanized Mice. THE JOURNAL OF IMMUNOLOGY 2016; 198:1047-1055. [PMID: 28011932 DOI: 10.4049/jimmunol.1601640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/18/2016] [Indexed: 12/13/2022]
Abstract
In the RV144 gp120 HIV vaccine trial, decreased transmission risk was correlated with Abs that reacted with a linear epitope at a lysine residue at position 169 (K169) in the HIV-1 envelope (Env) V2 region. The K169 V2 response was restricted to Abs bearing Vλ rearrangements that expressed aspartic acid/glutamic acid in CDR L2. The AE.A244 gp120 in AIDSVAX B/E also bound to the unmutated ancestor of a V2-glycan broadly neutralizing Ab, but this Ab type was not induced in the RV144 trial. In this study, we sought to determine whether immunodominance of the V2 linear epitope could be overcome in the absence of human Vλ rearrangements. We immunized IgH- and Igκ-humanized mice with the AE.A244 gp120 Env. In these mice, the V2 Ab response was focused on a linear epitope that did not include K169. V2 Abs were isolated that used the same human VH gene segment as an RV144 V2 Ab but paired with a mouse λ L chain. Structural characterization of one of these V2 Abs revealed how the linear V2 epitope could be engaged, despite the lack of aspartic acid/glutamic acid encoded in the mouse repertoire. Thus, despite the absence of the human Vλ locus in these humanized mice, the dominance of Vλ pairing with human VH for HIV-1 Env V2 recognition resulted in human VH pairing with mouse λ L chains instead of allowing otherwise subdominant V2-glycan broadly neutralizing Abs to develop.
Collapse
Affiliation(s)
- Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710; .,Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - Nathan I Nicely
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Bradley Lockwood
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | | | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Sabrina Arora
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Cindy M Bowman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Christina Stolarchuk
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Krissey E Lloyd
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Shi-Mao Xia
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Ryan Duffy
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | | | | | | | - Richard M Scearce
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Immunology, Duke University, Durham, NC 27710.,Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Pathology, Duke University School of Medicine, Durham, NC 27710; and
| | - Laurent Verkoczy
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Pathology, Duke University School of Medicine, Durham, NC 27710; and
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Garnett Kelsoe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Immunology, Duke University, Durham, NC 27710
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710.,Department of Immunology, Duke University, Durham, NC 27710
| |
Collapse
|
31
|
Mesin L, Ersching J, Victora GD. Germinal Center B Cell Dynamics. Immunity 2016; 45:471-482. [PMID: 27653600 PMCID: PMC5123673 DOI: 10.1016/j.immuni.2016.09.001] [Citation(s) in RCA: 663] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/01/2023]
Abstract
Germinal centers (GCs) are the site of antibody diversification and affinity maturation and as such are vitally important for humoral immunity. The study of GC biology has undergone a renaissance in the past 10 years, with a succession of findings that have transformed our understanding of the cellular dynamics of affinity maturation. In this review, we discuss recent developments in the field, with special emphasis on how GC cellular and clonal dynamics shape antibody affinity and diversity during the immune response.
Collapse
Affiliation(s)
- Luka Mesin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Jonatan Ersching
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Gabriel D Victora
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| |
Collapse
|
32
|
DeFranco AL. Germinal centers and autoimmune disease in humans and mice. Immunol Cell Biol 2016; 94:918-924. [PMID: 27562062 DOI: 10.1038/icb.2016.78] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Antibodies are involved in the pathogenesis of many autoimmune diseases. Although the mechanisms underlying the antibody response to infection or vaccination are reasonably well understood, we still have a poor understanding of the nature of autoimmune antibody responses. The most well studied are the anti-nuclear antibody responses characteristic of systemic lupus erythematosus and studies over the past decade or so have demonstrated a critical role for signaling by TLR7 and/or TLR9 in B cells to promote these responses. These Toll-like receptors (TLRs) can promote T-cell-independent extrafollicular antibody responses with a heavy-chain class switch and a low degree of somatic mutation, but they can also strongly boost the germinal center response that gives rise to high-affinity antibodies and long-lived plasma cells. TLRs have been shown to enhance affinity maturation in germinal center responses to produce high-affinity neutralizing antibodies in several virus infection models of mice. Although more data are needed, it appears that anti-nuclear antibodies in mouse models of lupus and in lupus patients can be generated by either pathway, provided there are genetic susceptibility alleles that compromise B-cell tolerance at one or another stage. Limited data in other autoimmune diseases suggest that the germinal center response may be the predominant pathway leading to autoantibodies in those diseases. A better understanding of the mechanisms of autoantibody production may ultimately be helpful in the development of targeted therapeutics for lupus or other autoimmune diseases.
Collapse
Affiliation(s)
- Anthony L DeFranco
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| |
Collapse
|
33
|
Autonomous membrane IgE signaling prevents IgE-memory formation. Nat Immunol 2016; 17:1109-17. [PMID: 27428827 DOI: 10.1038/ni.3508] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/02/2016] [Indexed: 12/14/2022]
Abstract
Aberrant production of IgE antibodies can lead to allergic diseases. Normally, IgE(+) B cells rarely differentiate into memory B cells (Bmem) or long-lived plasma cells (LLPCs), as they only transiently participate in the germinal center (GC), but the mechanism behind this remains elusive. We found that membrane IgE (mIgE) autonomously triggered rapid plasma-cell differentiation and apoptosis independently of antigen or cellular context, predominantly through the mutually independent CD19-PI3K-Akt-IRF4 and BLNK-Jnk/p38 pathways, respectively, and we identified the ectodomains of mIgE as being responsible. Accordingly, deregulated GC IgE(+) B cell proliferation and prolonged IgE production with exaggerated anaphylaxis were observed in CD19- and BLNK-deficient mice. Our findings reveal an autonomous mIgE signaling mechanism that normally prevents IgE(+) Bmem and LLPC formation, providing insights into the molecular pathogenesis of allergic diseases.
Collapse
|
34
|
EAF2 mediates germinal centre B-cell apoptosis to suppress excessive immune responses and prevent autoimmunity. Nat Commun 2016; 7:10836. [PMID: 26935903 PMCID: PMC4782062 DOI: 10.1038/ncomms10836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/25/2016] [Indexed: 01/03/2023] Open
Abstract
Regulated apoptosis of germinal centre (GC) B cells is critical for normal humoral immune responses. ELL-associated factor 2 (EAF2) regulates transcription elongation and has been shown to be an androgen-responsive potential tumour suppressor in prostate by inducing apoptosis. Here we show that EAF2 is selectively upregulated in GC B cells among various immune cell types and promotes apoptosis of GC B cells both in vitro and in vivo. EAF2 deficiency results in enlarged GCs and elevated antibody production during a T-dependent immune response. After immunization with type II collagen, mice lacking EAF2 produce high levels of collagen-specific autoantibodies and rapidly develop severe arthritis. Moreover, the mutant mice spontaneously produce anti-dsDNA, rheumatoid factor and anti-nuclear antibodies as they age. These results demonstrate that EAF2-mediated apoptosis in GC B cells limits excessive humoral immune responses and is important for maintaining self-tolerance.
Collapse
|
35
|
Haynes BF, Shaw GM, Korber B, Kelsoe G, Sodroski J, Hahn BH, Borrow P, McMichael AJ. HIV-Host Interactions: Implications for Vaccine Design. Cell Host Microbe 2016; 19:292-303. [PMID: 26922989 DOI: 10.1016/j.chom.2016.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Development of an effective AIDS vaccine is a global priority. However, the extreme diversity of HIV type 1 (HIV-1), which is a consequence of its propensity to mutate to escape immune responses, along with host factors that prevent the elicitation of protective immune responses, continue to hinder vaccine development. Breakthroughs in understanding of the biology of the transmitted virus, the structure and nature of its envelope trimer, vaccine-induced CD8 T cell control in primates, and host control of broadly neutralizing antibody elicitation have given rise to new vaccine strategies. Despite this promise, emerging data from preclinical trials reinforce the need for additional insight into virus-host biology in order to facilitate the development of a successful vaccine.
Collapse
Affiliation(s)
- Barton F Haynes
- Department of Medicine, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke University Human Vaccine Institute, Duke University, Durham, NC 27710, USA.
| | - George M Shaw
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bette Korber
- Los Alamos National Laboratory, Los Alamos, NM 87544, USA
| | - Garnett Kelsoe
- Department of Immunology, Duke University, Durham, NC 27710, USA; Duke University Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Joseph Sodroski
- Dana Farber-Cancer Institute, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Beatrice H Hahn
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Persephone Borrow
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Andrew J McMichael
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| |
Collapse
|
36
|
Butt D, Chan TD, Bourne K, Hermes JR, Nguyen A, Statham A, O'Reilly LA, Strasser A, Price S, Schofield P, Christ D, Basten A, Ma CS, Tangye SG, Phan TG, Rao VK, Brink R. FAS Inactivation Releases Unconventional Germinal Center B Cells that Escape Antigen Control and Drive IgE and Autoantibody Production. Immunity 2015; 42:890-902. [PMID: 25979420 DOI: 10.1016/j.immuni.2015.04.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/09/2015] [Accepted: 03/25/2015] [Indexed: 12/01/2022]
Abstract
The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.
Collapse
Affiliation(s)
- Danyal Butt
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Tyani D Chan
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Katherine Bourne
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Jana R Hermes
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Akira Nguyen
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Aaron Statham
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Lorraine A O'Reilly
- Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andreas Strasser
- Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Susan Price
- Molecular Development Section, Laboratory of Immunology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Peter Schofield
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Daniel Christ
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Antony Basten
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Cindy S Ma
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - V Koneti Rao
- Molecular Development Section, Laboratory of Immunology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
37
|
Yusuf I, Stern J, McCaughtry TM, Gallagher S, Sun H, Gao C, Tedder T, Carlesso G, Carter L, Herbst R, Wang Y. Germinal center B cell depletion diminishes CD4+ follicular T helper cells in autoimmune mice. PLoS One 2014; 9:e102791. [PMID: 25101629 PMCID: PMC4125140 DOI: 10.1371/journal.pone.0102791] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 06/24/2014] [Indexed: 01/25/2023] Open
Abstract
Background Continuous support from follicular CD4+ T helper (Tfh) cells drives germinal center (GC) responses, which last for several weeks to produce high affinity memory B cells and plasma cells. In autoimmune Sle1 and NZB/W F1 mice, elevated numbers of Tfh cells persist, promoting the expansion of self-reactive B cells. Expansion of circulating Tfh like cells have also been described in several autoimmune diseases. Although, the signals required for Tfh differentiation have now been well described, the mechanisms that sustain the maintenance of fully differentiated Tfh are less understood. Recent data demonstrate a role for GC B cells for Tfh maintenance after protein immunization. Methods and Finding Given the pathogenic role Tfh play in autoimmune disease, we explored whether B cells are required for maintenance of autoreactive Tfh. Our data suggest that the number of mature autoreactive Tfh cells is controlled by GC B cells. Depletion of B cells in Sle1 autoimmune mice leads to a dramatic reduction in Tfh cells. In NZB/W F1 autoimmune mice, similar to the SRBC immunization model, GC B cells support the maintenance of mature Tfh, which is dependent mainly on ICOS. The CD28-associated pathway is dispensable for Tfh maintenance in SRBC immunized mice, but is required in the spontaneous NZB/W F1 model. Conclusion These data suggest that mature Tfh cells require signals from GC B cells to sustain their optimal numbers and function in both autoimmune and immunization models. Thus, immunotherapies targeting B cells in autoimmune disease may affect pathogenic Tfh cells.
Collapse
Affiliation(s)
- Isharat Yusuf
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Jessica Stern
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Tom M McCaughtry
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Sandra Gallagher
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Hong Sun
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Changshou Gao
- Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Thomas Tedder
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Gianluca Carlesso
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Laura Carter
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Ronald Herbst
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| | - Yue Wang
- Department of Respiratory, Inflammation and Autoimmunity Research, MedImmune LLC, Gaithersburg, Maryland, United States of America
| |
Collapse
|
38
|
|
39
|
Or-Guil M, Faro J. A major hindrance in antibody affinity maturation investigation: we never succeeded in falsifying the hypothesis of single-step selection. Front Immunol 2014; 5:237. [PMID: 24904585 PMCID: PMC4033600 DOI: 10.3389/fimmu.2014.00237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 05/07/2014] [Indexed: 01/17/2023] Open
Affiliation(s)
- Michal Or-Guil
- Systems Immunology Laboratory, Department of Biology, Humboldt-Universität zu Berlin , Berlin , Germany ; Research Center ImmunoSciences, Charité-Universitätsmedizin Berlin , Berlin , Germany
| | - Jose Faro
- Area of Immunology, Faculty of Biology, Biomedical Research Center (CINBIO), Universidade de Vigo , Vigo , Spain ; Instituto Biomédico de Vigo , Vigo , Spain ; Instituto Gulbenkian de Ciência , Oeiras , Portugal
| |
Collapse
|
40
|
Holl TM, Yang G, Kuraoka M, Verkoczy L, Alam SM, Moody MA, Haynes BF, Kelsoe G. Enhanced antibody responses to an HIV-1 membrane-proximal external region antigen in mice reconstituted with cultured lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:3269-79. [PMID: 24591365 PMCID: PMC4003504 DOI: 10.4049/jimmunol.1302829] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We have shown that the protective HIV-1 Ab, 2F5, avidly reacts with a conserved mammalian self-Ag, kynureninase, and that the development of B cells specific for the 2F5 epitope is constrained by immunological tolerance. These observations suggest that the capacity to mount Ab responses to the 2F5 epitope is mitigated by tolerance, but such capacity may be latent in the pretolerance and/or anergic B cell pools. In this study, we use B cell tetramer reagents to track the frequencies of B cells that recognize the HIV-1 2F5 epitope (SP62): in C57BL/6 mice, SP62-binding transitional B cells are readily identified in bone marrow but are lost during subsequent development. Unsurprisingly then, immunization with SP62 immunogen does not elicit significant humoral responses in normal C57BL/6 mice. Reconstitution of Rag1(null) mice with normal congenic B cells that have matured in vitro restores the capacity to mount significant serum Ab and germinal center responses to this HIV-1 epitope. These B cell cultures are permissive for the development of autoreactive B cells and support the development of SP62-specific B cell compartments normally lost in 2F5 Ab knockin mice. The recovery of humoral responses to the 2F5/SP62 epitope of HIV-1 by reconstitution with B cells containing forbidden, autoreactive clones provides direct evidence that normal C57BL/6 mice latently possess the capacity to generate humoral responses to a conserved, neutralizing HIV-1 epitope.
Collapse
Affiliation(s)
- T. Matt Holl
- Department of Immunology, Duke University, Durham, North Carolina 27710, USA
| | - Guang Yang
- Department of Immunology, Duke University, Durham, North Carolina 27710, USA
| | - Masayuki Kuraoka
- Department of Immunology, Duke University, Durham, North Carolina 27710, USA
| | - Laurent Verkoczy
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
- Department of Pathology, Duke University, Durham, North Carolina 27710, USA
| | - S. Munir Alam
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
- Department of Pathology, Duke University, Durham, North Carolina 27710, USA
- Department of Medicine Duke University, Durham, North Carolina 27710, USA
| | - M. Anthony Moody
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
- Department of Pathology, Duke University, Durham, North Carolina 27710, USA
| | - Barton F. Haynes
- Department of Immunology, Duke University, Durham, North Carolina 27710, USA
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
- Department of Medicine Duke University, Durham, North Carolina 27710, USA
| | - Garnett Kelsoe
- Department of Immunology, Duke University, Durham, North Carolina 27710, USA
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
| |
Collapse
|
41
|
Brink R. The imperfect control of self-reactive germinal center B cells. Curr Opin Immunol 2014; 28:97-101. [PMID: 24686094 DOI: 10.1016/j.coi.2014.03.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 03/06/2014] [Accepted: 03/06/2014] [Indexed: 12/01/2022]
Abstract
Unlike T cells, B cells diversify their antigen receptor (BCR) binding specificities at two distinct stages of differentiation. Thus, in addition to initial variable region gene rearrangements, B cells recruited into T-dependent immune responses further modify their BCR specificity via iterative rounds of somatic hypermutation (SHM) within germinal centers (GCs). Although critical for providing the high-affinity antibody specificities required for long-term immune protection, SHM can also generate self-reactive B cells capable of differentiating into autoantibody-producing plasma cells. Recent data confirm that self-reactive GC B cells can be effectively removed from the secondary repertoire so as to maintain self-tolerance. However, they can also escape deletion under certain circumstances and so contribute to autoimmune disease via production of somatically mutated, pathogenic autoantibodies.
Collapse
Affiliation(s)
- Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
42
|
Clonal and cellular dynamics in germinal centers. Curr Opin Immunol 2014; 28:90-6. [PMID: 24681449 DOI: 10.1016/j.coi.2014.02.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/23/2014] [Accepted: 02/24/2014] [Indexed: 01/16/2023]
Abstract
Germinal centers (GCs) are the site of antibody affinity maturation, a process that involves complex clonal and cellular dynamics. Selection of B cells bearing higher-affinity immunoglobulins proceeds via a stereotyped pattern whereby B cells migrate cyclically between the GC's two anatomical compartments. This process occurs in a timeframe that is well suited to analysis by intravital microscopy, and much has been learned in recent years by use of these techniques. On a longer time scale, the diversity of B cell clones and variants within individual GCs is also thought to change as affinity maturation progresses; however, our understanding of clonal dynamics in individual GCs is limited. We discuss recent progress in the elucidation of clonal and cellular dynamics patterns.
Collapse
|
43
|
Weinstein JS, Bertino SA, Hernandez SG, Poholek AC, Teplitzky TB, Nowyhed HN, Craft J. B cells in T follicular helper cell development and function: separable roles in delivery of ICOS ligand and antigen. THE JOURNAL OF IMMUNOLOGY 2014; 192:3166-79. [PMID: 24610013 DOI: 10.4049/jimmunol.1302617] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
B cells are required for follicular Th (Tfh) cell development, as is the ICOS ligand (ICOS-L); however, the separable contributions of Ag and ICOS-L delivery by cognate B cells to Tfh cell development and function are unknown. We find that Tfh cell and germinal center differentiation are dependent on cognate B cell display of ICOS-L, but only when Ag presentation by the latter is limiting, with the requirement for B cell expression of ICOS-L overcome by robust Ag delivery. These findings demonstrate that Ag-specific B cells provide different, yet compensatory, signals for Tfh cell differentiation, while reconciling conflicting data indicating a requirement for ICOS-L expression on cognate B cells for Tfh cell development with those demonstrating that the latter requirement could be bypassed in lieu of that tendered by noncognate B cells. Our findings clarify the separable roles of delivery of Ag and ICOS-L by cognate B cells for Tfh cell maturation and function, and have implications for using therapeutic ICOS blockade in settings of abundantly available Ag, such as in systemic autoimmunity.
Collapse
Affiliation(s)
- Jason S Weinstein
- Section of Rheumatology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520
| | | | | | | | | | | | | |
Collapse
|
44
|
Giltiay NV, Lu Y, Cullen JL, Jørgensen TN, Shlomchik MJ, Li X. Spontaneous loss of tolerance of autoreactive B cells in Act1-deficient rheumatoid factor transgenic mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:2155-63. [PMID: 23904159 DOI: 10.4049/jimmunol.1300152] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Self-reactive B cells in BALB/c AM14 transgenic (Tg) rheumatoid factor mice are not subject to central or peripheral tolerization. Instead, they remain at a stage of "clonal ignorance"; that is, they do not proliferate and differentiate into Ab-forming cells. However, the immunoregulatory mechanisms that prevent autoantibody production in these mice remain unclear. In this study, we show that crossing AM14 Tg mice to a mouse strain deficient in Act1, a molecule involved in the regulation of BAFF-R and CD40-signaling in B cells, results in spontaneous activation of AM14 Tg B cells and production of AM14-specific Abs. Three- to 5-mo-old AM14 Tg Act1(-/-) mice showed significant expansion of AM14 Tg B cells, including a 2- to 3-fold increase in the spleen and cervical lymph nodes compared with AM14 Tg Act1(+/+) mice. Furthermore, in the presence of endogenous self-Ag (IgH(a) congenic background), AM14 Tg Act1(-/-) B cells were spontaneously activated and differentiated into Ab-forming cells. In contrast with previous studies using AM14 Tg MLR.Fas(lpr) mice, we found that a significant number of AM14 Tg cells AM14 Tg Act1(-/-) mice displayed phenotypic characteristics of germinal center B cells. Anti-CD40L treatment significantly limited the expansion and activation of AM14 Tg Act1(-/-) B cells, suggesting that CD40L-mediated signals are required for the retention of these cells. Our results support the important role of Act1 in the regulation of self-reactive B cells and reveal how Act1 functions to prevent the production of autoantibodies.
Collapse
Affiliation(s)
- Natalia V Giltiay
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
45
|
Faro J, Or-Guil M. How oligoclonal are germinal centers? A new method for estimating clonal diversity from immunohistological sections. BMC Bioinformatics 2013; 14 Suppl 6:S8. [PMID: 23734629 PMCID: PMC3633029 DOI: 10.1186/1471-2105-14-s6-s8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background The germinal center (GC) reaction leads to antibody affinity maturation and generation of memory B cells, but its underlying mechanisms are poorly understood. To assemble this puzzle, several key pieces of information are needed, one in particular being the number of participating B cell clones. Since this clonal diversity cannot be observed directly, earlier studies resorted to interpreting two types of available experimental data: Immunohistology of GCs containing two phenotypically distinct B-cell populations, and antibody gene sequences of small B-cell samples from GCs. Based on a simple model, investigators concluded that a typical GC was seeded by 2-8 B cells, endorsing the current notion that GCs are oligoclonal from the onset. Results A re-evaluation of these data showed that the used simple model is not statistically consistent with the original data. From an analysis of the experimental system, we propose a new model for estimating GC clonal diversity, including the initially neglected sampling and measurement errors, and making more general assumptions. Consistency analysis with the new model yielded an estimation of sampling and measurement errors in the experimental data of 10-11% for one B-cell population and 62-64% for the other population, and an average number of 19-23 seeder B cells. An independent analysis of antibody gene sequences of small B-cell samples from GCs, using an adapted Yule estimator of diversity, yielded a minimum estimation of 20-30 GC founder B cells, confirming the previous results. Conclusions Our new experimental-based model provides a highly improved method to estimate the clonal diversity of GCs from inmunohistochemistry data of chimeric animals. Calculations based on this model, and validated by an independent approach, indicate that GCs most likely contain broadly varying numbers of different B cell clones, averaging 5- to 10-fold more clones than previously estimated. These findings, in line with recent results showing that GC sizes and life times are also subject to high variability, dramatically change the picture of GC dynamics.
Collapse
Affiliation(s)
- Jose Faro
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain.
| | | |
Collapse
|
46
|
Affiliation(s)
- Roybel R. Ramiscal
- Department of Pathogens and Immunity, John Curtin School of Medical Research; Australian National University; Canberra; ACT; Australia
| | - Carola G. Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research; Australian National University; Canberra; ACT; Australia
| |
Collapse
|
47
|
Yang G, Holl TM, Liu Y, Li Y, Lu X, Nicely NI, Kepler TB, Alam SM, Liao HX, Cain DW, Spicer L, VandeBerg JL, Haynes BF, Kelsoe G. Identification of autoantigens recognized by the 2F5 and 4E10 broadly neutralizing HIV-1 antibodies. ACTA ACUST UNITED AC 2013; 210:241-56. [PMID: 23359068 PMCID: PMC3570098 DOI: 10.1084/jem.20121977] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Many human monoclonal antibodies that neutralize multiple clades of HIV-1 are polyreactive and bind avidly to mammalian autoantigens. Indeed, the generation of neutralizing antibodies to the 2F5 and 4E10 epitopes of HIV-1 gp41 in man may be proscribed by immune tolerance because mice expressing the V(H) and V(L) regions of 2F5 have a block in B cell development that is characteristic of central tolerance. This developmental blockade implies the presence of tolerizing autoantigens that are mimicked by the membrane-proximal external region of HIV-1 gp41. We identify human kynureninase (KYNU) and splicing factor 3b subunit 3 (SF3B3) as the primary conserved, vertebrate self-antigens recognized by the 2F5 and 4E10 antibodies, respectively. 2F5 binds the H4 domain of KYNU which contains the complete 2F5 linear epitope (ELDKWA). 4E10 recognizes an epitope of SF3B3 that is strongly dependent on hydrophobic interactions. Opossums carry a rare KYNU H4 domain that abolishes 2F5 binding, but they retain the SF3B3 4E10 epitope. Immunization of opossums with HIV-1 gp140 induced extraordinary titers of serum antibody to the 2F5 ELDKWA epitope but little or nothing to the 4E10 determinant. Identification of structural motifs shared by vertebrates and HIV-1 provides direct evidence that immunological tolerance can impair humoral responses to HIV-1.
Collapse
Affiliation(s)
- Guang Yang
- Department of Immunology, Duke University, Durham, NC 27705, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Elimination of Germinal-Center-Derived Self-Reactive B Cells Is Governed by the Location and Concentration of Self-Antigen. Immunity 2012; 37:893-904. [DOI: 10.1016/j.immuni.2012.07.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 07/12/2012] [Indexed: 11/29/2022]
|
49
|
B-cell-lineage immunogen design in vaccine development with HIV-1 as a case study. Nat Biotechnol 2012; 30:423-33. [PMID: 22565972 DOI: 10.1038/nbt.2197] [Citation(s) in RCA: 380] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Failure of immunization with the HIV-1 envelope to induce broadly neutralizing antibodies against conserved epitopes is a major barrier to producing a preventive HIV-1 vaccine. Broadly neutralizing monoclonal antibodies (BnAbs) from those subjects who do produce them after years of chronic HIV-1 infection have one or more unusual characteristics, including polyreactivity for host antigens, extensive somatic hypermutation and long, variable heavy-chain third complementarity-determining regions, factors that may limit their expression by host immunoregulatory mechanisms. The isolation of BnAbs from HIV-1-infected subjects and the use of computationally derived clonal lineages as templates provide a new path for HIV-1 vaccine immunogen design. This approach, which should be applicable to many infectious agents, holds promise for the construction of vaccines that can drive B cells along rare but desirable maturation pathways.
Collapse
|
50
|
Affiliation(s)
- Gabriel D. Victora
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142;
| | - Michel C. Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| |
Collapse
|