Elsevier

Seminars in Immunology

Volume 23, Issue 5, October 2011, Pages 341-349
Seminars in Immunology

Review
The genetic network controlling plasma cell differentiation

https://doi.org/10.1016/j.smim.2011.08.010Get rights and content

Abstract

Upon activation by antigen, mature B cells undergo immunoglobulin class switch recombination and differentiate into antibody-secreting plasma cells, the endpoint of the B cell developmental lineage. Careful quantitation of these processes, which are stochastic, independent and strongly linked to the division history of the cell, has revealed that populations of B cells behave in a highly predictable manner. Considerable progress has also been made in the last few years in understanding the gene regulatory network that controls the B cell to plasma cell transition. The mutually exclusive transcriptomes of B cells and plasma cells are maintained by the antagonistic influences of two groups of transcription factors, those that maintain the B cell program, including Pax5, Bach2 and Bcl6, and those that promote and facilitate plasma cell differentiation, notably Irf4, Blimp1 and Xbp1. In this review, we discuss progress in the definition of both the transcriptional and cellular events occurring during late B cell differentiation, as integrating these two approaches is crucial to defining a regulatory network that faithfully reflects the stochastic features and complexity of the humoral immune response.

Highlights

► Terminal differentiation of B cells to plasma cells occurs in a stochastic manner. ► A gene regulatory network controls plasma cell differentiation. ► Gene expression in B cells and plasma cells is maintained by distinct transcription factors. ► Blimp1 and Irf4 are dose-dependent regulators of plasma cells.

Introduction

Terminal differentiation is a generally irreversible process that, in the haematopoietic system, leads to the acquisition of specialized effector functions and exit from the cell cycle. The terminal differentiation process is also associated with profound alterations in the morphology, lifespan and gene expression profiles of the differentiated cells compared to their predecessors. The transition of B cells into antibody-secreting cells (ASCs) represents one such terminal differentiation process that is essential for the adaptive immune response.

There are three main subsets of mature B cells; follicular B cells, which represent the majority of naïve B cells, marginal zone (MZ) and B1 B cells. MZ B cells reside in the marginal sinus of the spleen, whereas B1 B cells occur predominantly in the peritoneal cavity and at mucosal sites [1]. In contrast, conventional or follicular B cells are almost exclusively found in the lymphoid follicles of the spleen and lymph nodes. The relative propensity of B cells to undergo terminal differentiation varies between the subsets, with MZ and B1 B cells being specialized, both geographically and genetically, for rapid response to so-called T-independent antigens (TI), such as bacterial components [2]. Follicular B cells may also respond to TI antigens, but appear specialized for responding to antigens that also elicit CD4+ helper T cell responses. Upon antigen encounter and receipt of T cell help, follicular B cells undergo multiple rounds of division and have the unique ability to differentiate into both ASCs (plasmablasts and plasma cells) and memory B cells [1]. Plasmablasts are short-lived, cycling ASCs that are found in extrafollicular foci in peripheral lymphoid organs. Plasmablasts have often undergone immunoglobulin class switch recombination (CSR), but not somatic hypermutation (SHM) to increase the affinity of the resulting antibodies for the antigen. In a poorly understood process, some activated B cells return to the B cell follicle and proliferate vigorously to form a germinal centre (GC), where the B cells initiate SHM to enable further clonal selection. The GC produces long-lived plasma cells that are non-cycling and home preferentially to the bone marrow [1]. The GC reaction also produces memory B cells. These cells maintain a B cell phenotype but can rapidly differentiate into ASCs following re-exposure to antigen.

The process of B cell terminal differentiation can be studied in vitro, as B cells are capable of both CSR and ASC differentiation in response to T-cell derived stimuli (CD40 ligation and cytokines) or TI-related signals (Toll-like receptor (TLR) ligation by pathogen derived products such as lipopolysaccharide (LPS) and hyper-methylated CpG DNA). B cell responses in vitro thus provide a controlled system to investigate the biology of B cell terminal differentiation on both a cellular and molecular level.

Quantitative analysis of in vitro B cell cultures has revealed a striking relationship between cell division history and CSR and ASC differentiation [3], [4], [5]. The proportion of B cells that undergo either of these differentiation events typically increases with each consecutive division (Fig. 1). The time taken to traverse each cell cycle is highly variable between cells, but does not seem to alter the differentiation rate, implying that cell division itself is playing a critical role in tuning the molecular machinery controlling CSR and the generation of ASCs. Furthermore, T cell cytokines such as IL4 and IL5 alter the probability of CSR and ASC differentiation with division (Fig. 1B). Hasbold et al. found that CSR and development of ASCs behaved as independent stochastic processes, allowing the numbers of class switched ASCs in each division to be predicted for different cytokine concentrations and combinations [5]. These findings have lead to a division-based model of B cell behaviour that describes how stochastic decisions taken at a single cell level result in the controlled generation of a variety of differentiated cell types in the population as a whole [5].

Complementing these cellular studies, the last few years have seen major advances in our understanding of the transcriptional regulation of the B cell to ASC transition [6], [7]. The gene expression changes that are required for this process are regulated by the coordinated activity of a small group of so-called master regulatory transcription factors. These factors can be divided into those, such as Pax5 and Bcl6, which promote and maintain the B cell program, and others such as Blimp1 and Irf4 that control ASC differentiation. The B cell and ASC factors appear to regulate mutually antagonistic transcriptional programs resulting in a gene regulatory network that ensures the separation of the B cell and ASC fates [6].

While most studies to date have proposed that these master regulatory transcription factors function in a dominant manner to either maintain the B cell fate or to drive ASC differentiation, such deterministic behaviour has not yet been reconciled with the evidence supporting stochastic, division-based regulation of differentiation outlined above. In this review we will discuss both the transcriptional and cellular models of B cell terminal differentiation to draw attention to the need for a model of the genetic network of ASC differentiation that more accurately describes the flexible, dynamic and complex regulation of the adaptive immune response.

Section snippets

Factors that promote the B cell fate

One class of transcription factors can be conveniently grouped into those whose primary function is to promote the B cell gene expression program and to prevent ASC differentiation. Here we have focused on the best-characterized B cell factors: Pax5, Bcl6, Bach2 and Oct2/Obf1. It should be noted, however, that a number of other transcriptional regulators, including PU.1 [8], Irf8 [9], MITF [10] and Ets1 [11] are also thought to play roles in the process (Fig. 2).

Irf4

Irf4 is a multi-functional transcriptional regulator that controls many aspects of B cell differentiation including Igκ gene recombination, CSR, GC B cell formation and ASC differentiation [47]. Irf4 also is broadly required for the differentiation of CD4+ T cells [48]. Irf4 can bind to DNA weakly on its own, but displays strong co-operative binding in the presence of PU.1, or the closely related Spi-B [49], [50]. Irf4-PU.1 dimers have been shown to be important in the regulation of the Igh and

Towards describing a gene regulatory network controlling late B cell differentiation

The large amount of data that was summarized in the preceding section was in the whole generated by the analysis of individual transcription factors in isolation. However, this information can also be used to construct gene regulatory networks that attempt to define the crucial processes and interactions that allow the transition from a B cell to an ASC, in a manner analogous to that successfully applied to early haematopoiesis and T cell differentiation [80], [81]. B cell terminal

Blimp1 and Irf4 are dose-dependent regulators of antibody-secreting cell differentiation

While Irf4 emerges as a key component in the decision to undergo CSR and develop into an ASC, it is necessary to contrast its action with the patterned division-linked changes seen at the population level. Questions to be addressed include: why do only a proportion of stimulated cells in vitro develop into ASCs and undergo CSR in each division, despite the presence of Irf4 in all activated B cells? If Irf4 plays a critical role for both CSR and ASC development, why are the two outcomes poorly

Conclusions and future perspectives

Research over the past decade has provided a wealth of data to highlight the key factors in the gene regulatory network driving the terminal differentiation of B cells. Two classes of transcription factors are required, those that promote the B cell state, such as Bcl6 and Pax5 and those including Irf4 and Blimp1, that favour ASC differentiation. On a genetic level, the mutual antagonism between these factors ensures that these key developmental stages in B cell differentiation are kept

Acknowledgements

We would like to thank members of the Walter and Eliza Hall Institute B cell program for discussions. This research was supported by the Pfizer Australia Research Fellowship and ARC Future Fellowship to S.L.N. and National Health and Medical Research Council of Australia Research Fellowships to P.D.H. and L.M.C. This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIIS.

References (96)

  • K. Basso et al.

    Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells

    Blood

    (2010)
  • W. Ci et al.

    The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL

    Blood

    (2009)
  • M. Saito et al.

    A signaling pathway mediating downregulation of BCL6 in germinal center B cells is blocked by BCL6 gene alterations in B cell lymphoma

    Cancer Cell

    (2007)
  • H. Toyama et al.

    Memory B cells without somatic hypermutation are generated from Bcl6-deficient B cells

    Immunity

    (2002)
  • L.M. Corcoran et al.

    Oct-2 is required early in T cell-independent B cell activation for G1 progression and for proliferation

    Immunity

    (1994)
  • T. Decker et al.

    Stepwise activation of enhancer and promoter regions of the B cell commitment gene Pax5 in early lymphopoiesis

    Immunity

    (2009)
  • R. Sciammas et al.

    Graded expression of interferon regulatory factor-4 coordinates isotype switching with plasma cell differentiation

    Immunity

    (2006)
  • H. Kwon et al.

    Analysis of IL-21-induced Prdm1 gene regulation reveals functional cooperation of STAT3 and IRF4 transcription factors

    Immunity

    (2009)
  • A. Paun et al.

    Functional characterization of murine interferon regulatory factor 5 (IRF-5) and its role in the innate antiviral response

    J Biol Chem

    (2008)
  • A. Kallies et al.

    Initiation of plasma-cell differentiation is independent of the transcription factor Blimp-1

    Immunity

    (2007)
  • M. Shapiro-Shelef et al.

    Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells

    Immunity

    (2003)
  • A.L. Shaffer et al.

    Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program

    Immunity

    (2002)
  • H. Yoshida et al.

    XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor

    Cell

    (2001)
  • P. Laslo et al.

    Gene regulatory networks directing myeloid and lymphoid cell fates within the immune system

    Semin Immunol

    (2008)
  • A.L. Shaffer et al.

    XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation

    Immunity

    (2004)
  • A.L. Shaffer et al.

    BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control

    Immunity

    (2000)
  • K.A. Fairfax et al.

    Different kinetics of blimp-1 induction in B cell subsets revealed by reporter gene

    J Immunol

    (2007)
  • P.D. Hodgkin et al.

    B cell differentiation and isotype switching is related to division cycle number

    J Exp Med

    (1996)
  • E.K. Deenick et al.

    Switching to IgG3, IgG2b, and IgA is division linked and independent, revealing a stochastic framework for describing differentiation

    J Immunol

    (1999)
  • J. Hasbold et al.

    Evidence from the generation of immunoglobulin G-secreting cells that stochastic mechanisms regulate lymphocyte differentiation

    Nat Immunol

    (2004)
  • K.L. Calame et al.

    Regulatory mechanisms that determine the development and function of plasma cells

    Annu Rev Immunol

    (2003)
  • C.H. Lee et al.

    Regulation of the germinal center gene program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding protein

    J Exp Med

    (2006)
  • L. Lin et al.

    Active inhibition of plasma cell development in resting B cells by microphthalmia-associated transcription factor

    J Exp Med

    (2004)
  • C. Cobaleda et al.

    Pax5: the guardian of B cell identity and function

    Nat Immunol

    (2007)
  • M. Fuxa et al.

    Reporter gene insertions reveal a strictly B lymphoid-specific expression pattern of pax5 in support of its B cell identity function

    J Immunol

    (2007)
  • S.L. Nutt et al.

    Commitment to the B-lymphoid lineage depends on the transcription factor Pax5

    Nature

    (1999)
  • C. Cobaleda et al.

    Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors

    Nature

    (2007)
  • A. Kallies et al.

    Plasma cell ontogeny defined by quantitative changes in blimp-1 expression

    J Exp Med

    (2004)
  • C. Pridans et al.

    Identification of Pax5 target genes in early B cell differentiation

    J Immunol

    (2008)
  • M.L. Holmes et al.

    Repression of Flt3 by Pax5 is crucial for B-cell lineage commitment

    Genes Dev

    (2006)
  • M.L. Holmes et al.

    The regulation of the B-cell gene expression programme by Pax5

    Immunol Cell Biol

    (2008)
  • C. Duy et al.

    BCL6 is critical for the development of a diverse primary B cell repertoire

    J Exp Med

    (2010)
  • A.L. Dent et al.

    Control of inflammation, cytokine expression, and germinal center formation by BCL-6

    Science

    (1997)
  • B.H. Ye et al.

    The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation

    Nat Genet

    (1997)
  • T. Fukuda et al.

    Disruption of the Bcl6 gene results in an impaired germinal center formation

    J Exp Med

    (1997)
  • C. Tunyaplin et al.

    Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation

    J Immunol

    (2004)
  • M.A. Linterman et al.

    IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses

    J Exp Med

    (2010)
  • D. Zotos et al.

    IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism

    J Exp Med

    (2010)
  • Cited by (184)

    • Mitochondrial respiration in B lymphocytes is essential for humoral immunity by controlling the flux of the TCA cycle

      2022, Cell Reports
      Citation Excerpt :

      Of note, prdm1 RNA (encoding BLIMP1) was lower in DNT-B cells as well (Figure 4J). To exlude the possibility that BLIMP1 was repressed by high abundance of BCL6 (Nutt et al., 2011) we also examined BCL6 protein (Figures 4H and 4I), showing that diminished BLIMP1 was not due to elevated BCL6. To corroborate these findings in vivo and to exclude potentially confounding effects of non-mutant B cells, we labeled 48-h LPS-activated B cells with a proliferation dye and transferred them into rag2−/− mice (Figure S4C).

    View all citing articles on Scopus
    View full text