Review
The many roles of IL-7 in B cell development; Mediator of survival, proliferation and differentiation

https://doi.org/10.1016/j.smim.2012.02.001Get rights and content

Abstract

Interleukin-7 (IL-7) plays several important roles during B cell development including aiding in; the specification and commitment of cells to the B lineage, the proliferation and survival of B cell progenitors; and maturation during the pro-B to pre-B cell transition. Regulation and modulation of IL-7 receptor (IL-7R) signaling is critical during B lymphopoiesis, because excessive or deficient IL-7R signaling leads to abnormal or inhibited B cell development. IL-7 works together with E2A, EBF, Pax-5 and other transcription factors to regulate B cell commitment, while also functions to regulate Ig rearrangement by modulating FoxO protein activation and Rag enhancer activity. Suppressor of cytokine signaling (SOCS) proteins are inhibitors of cytokine activation and, in B cells, function to fine tune IL-7R signaling; ensuring that appropriate IL-7 signals are transmitted to allow for efficient B cell commitment and development.

Highlights

► IL-7R signaling in B cells. ► The role of IL-7 during early B cell development. ► IL-7 regulation of B cell survival, proliferation and maturation. ► IL-7 regulation of V(D)J recombination. ► Regulation, modification and termination of IL-7R signaling.

Introduction

B cells are derived from hematopoietic stem cells (HSCs) in a process by which cells progressively develop B cell traits while repressing traits of other lineages. During development, progenitor B cells undergo rearrangement of their immunoglobulin (Ig) loci in a stepwise and reproducible fashion, whereby recombination of the heavy chain (HC) genes occurs in pro-B cells followed by recombination of the light chain (LC) genes in pre-B cells. Binding of μHC proteins with surrogate LC proteins λ5 and VpreB leads to surface expression of the pre-B cell receptor (pre-BCR) on large pre-B cells, which initiates survival, proliferation and maturation signals via Igα and Igβ signaling subunits. Small resting pre-B cells exit the cell cycle and once again begin the recombination process, leading to rearrangement of the LC proteins. LC proteins associate with μHC proteins to form the B cell receptor (BCR), which is first expressed on immature B cells. Immature B cells undergo positive and negative selection in the bone marrow (BM) and subsequently exit into the periphery where they undergo additional selection prior to becoming functional mature B cells capable of responding to antigen.

B lymphopoiesis takes place in niches within the fetal liver (FL) or BM, which provide the structure for development to proceed, as well as the necessary chemokines and cytokines that regulate signal transduction pathways in receptive cells. Initiation of signaling pathways in turn activate transcription factors (TFs) that function to induce or repress the expression of various target genes that modulate B cell survival, proliferation and differentiation. Interleukin-7 (IL-7) is a key cytokine during B cell development and is produced by stromal cells in the FL, BM, spleen and thymus. IL-7 receptor (IL-7R) signaling leads to the proliferation and survival of B cell progenitors as well as aids in the commitment of cells to the B lineage. Mice with targeted deletions of IL-7 or the IL-7R display a severe block at the early pro-B cell stage of development [1], [2]. The peripheral B cells that exist in these mice appear to have originated during fetal development, a time during which B lymphopoiesis is not absolutely dependent on IL-7. Thymic stromal lymphopoietin (TSLP) is a cytokine that possesses a number of characteristics in common with IL-7 and its receptor is composed of the IL-7Rα chain and TSLPR chain. Fetal derived pro-B and pre-B cells respond to TSLP, while in adult BM, only pre-B cells are TSLP-responsive [3], [4]. TSLP was thought to substitute for IL-7 during fetal development, because transgenic expression of TSLP in IL-7−/− mice restored B cell development during fetal and adult life [5]. However, IL-7−/−/TSLP−/− mice did not show enhanced defects during fetal B cell development and, instead, it was the absence of IL-7 and fms-related tyrosine kinase-3 ligand (Flt-3L) that completely abolished both fetal and adult development of B cells [6], [7]. Flt-3L was also able to recover the development of residual B cells present in IL-7Rα deficient mice [3]. IL-7 transgenic mice displayed increased numbers of immature and mature B cells in the BM, as well as extramedullary B lymphopoiesis whereby pro-B and pre-B cells were observed in the spleen, blood, and lymph nodes, and ultimately lead to lymphoproliferative disorders [8].

IL-7 is absolutely essential for murine B cell development, however, in humans it was thought that this was not the case. Genetic mutations in humans that disrupted the IL-7R led to X-linked severe combined immunodeficiency disease (X-SCID), which is characterised by the absence of T cells and natural killer (NK) cells but normal B cell numbers [9]. However, similar to mice, these B cells appear to be the result of fetal or neonatal development. In vitro, human B cells can be generated from fetal BM as well as cord blood (CB) in the absence of IL-7 [10], [11]. However, human B cell precursors express the IL-7R and displayed increased proliferation and survival in response to IL-7 that was mediated by Signal Transducer and Activator of Transcription-5 (STAT5) [12]. IL-7 also greatly increased the production of B cells in co-cultures containing human BM stroma and either CB or adult BM HSCs, while only neonatal CB was able to give rise to B cell progenitors independent of IL-7 [11]. In humans, TSLP cannot substitute for IL-7, while Flt-3L is able to support IL-7-independent B lymphopoiesis from neonatal CB [11], [13].

As important as the signaling pathways initiated by IL-7 and other cytokines, are the mechanisms that exist to regulate and terminate these signals. The suppressor of cytokine signaling (SOCS) family of proteins are key regulators of cytokine signals and are essential for the development and function of a variety of hematopoietic lineages (reviewed in [14]). SOCS proteins contain three functional domains: a central SH2 domain that is necessary for binding to phosphotyrosine residues on target proteins; a C-terminal domain, termed the SOCS box, which is involved in ubiquitin-mediated proteasomal degradation via elongins B and C; and an N-terminal domain whose function remains largely undefined. SOCS proteins bind directly to Janus Associated Kinase (JAK) and STAT proteins, as well as cytokine receptor chains, and prevent their interaction as well as target them for destruction. SOCS proteins are often induced by the same signaling pathway that they inhibit, providing a negative feedback loop that functions to limit receptor activation.

This review will focus on the signal transduction pathways activated downstream of the IL-7R, as well as how IL-7R signals are translated into the observed physiological outcomes of survival, proliferation and differentiation. We will also discuss the other surface proteins, signaling molecules and TFs that assist in mediating the effects of IL-7 in developing B cells. Finally, we will describe the mechanisms that exist both extrinsically and intrinsically to modulate and terminate IL-7R signals, ensuring that B cell development proceeds efficiently.

Section snippets

JAK/STAT and Src activation

The IL-7R is a heterodimer composed of the IL-7Rα chain and the common γ (γc) chain. The γc chain is a shared component of the receptors for IL-2, IL-4, IL-9, IL-15 and IL-21, while the IL-7Rα chain can also dimerize with the TSLPR chain to form the receptor for TSLP. The γc chain is expressed by a variety of hematopoietic cells and is essential for initiating signals downstream of the IL-7R; however, it is the α chain, which is predominantly expressed by lymphoid cells, that confers receptor

IL-7 synergy with other early co-factors

Developing B cells rely on a variety of factors that provide signals independently or in conjunction with those from the IL-7R. Stem cell factor (SCF, c-Kit-Ligand) binds c-Kit and is required during the earliest stages of hematopoietic development. SCF acts directly on developing B cells and, in vitro, worked synergistically with IL-7 to increase the numbers of pro-B cells in culture [44]. Flt-3L, and its receptor Flt-3, also play important roles in enhancing the survival and proliferation of

Cell survival

One of the main functions of IL-7R signaling during B cell development is to promote cell survival by regulating the localization and interaction of anti-apoptotic (Bcl-2, Bcl-xL and Mcl-1) and pro-apoptotic (Bax, Bad and Bim) factors. Both the JAK/STAT and PI3K/Akt pathways play key roles in mediating survival responses. IL-7R signaling enhances pro-B cell survival by increasing the ratio of Bcl-2 to Bax and, correspondingly, mice deficient for the IL-7R or JAK3 displayed increased levels of

IL-7 regulation of heavy chain recombination

Additional roles for IL-7 in promoting B cell development have been proposed based on studies demonstrating that IL-7R signaling can affect both HC and LC recombination. Several reports have noted that IL-7R activation of STAT5 promoted chromatin accessibility through histone acetylation of the distal VH genes in pro-B cells [88], [89]. In small pre-B cells, which are non-responsive to IL-7, distal VH sites were hypoacetylated and thus non-accessible, providing a possible IL-7-mediated

Modulation of IL-7R signaling by CD45 and the pre-BCR

A variety of signaling pathways are initiated in developing B cells. The factors activated by these pathways can interact with signaling molecules downstream of the IL-7R, modifying the magnitude, duration or quality of the response. B220 is the 220-kDa isoform of the surface membrane phosphatase CD45. CD45 is present on all mouse B cells, except terminally differentiated plasma blasts, as well as in varying isoforms on dendritic cells, T cells, macrophages and NK cells. CD45 dephosphorylates

Conclusion

IL-7 is a key factor that regulates B lymphopoiesis and plays multiple roles in the commitment, survival, proliferation and differentiation of B cell progenitors. During development, B cells receive both activation and inhibition signals from IL-7, which are modified by the amount of IL-7 present and the activation of other signaling pathways (Fig. 1). Certain aspects of the IL-7R signaling pathway have been well characterized, however, others have yet to be fully elucidated, including; the

Acknowledgements

This work was supported by the Canadian Institutes for Health Research. We would like to thank members of the Paige laboratory for helpful discussions and critical reading of this manuscript.

References (133)

  • T. Yasuda et al.

    Erk kinases link pre-B cell receptor signaling to transcriptional events required for early B cell expansion

    Immunity

    (2008)
  • K. Mackarehtschian et al.

    Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors

    Immunity

    (1995)
  • J. Ahsberg et al.

    Interleukin-7-induced Stat-5 acts in synergy with Flt-3 signaling to stimulate expansion of hematopoietic progenitor cells

    J Biol Chem

    (2010)
  • E.M. Mandel et al.

    Transcription control of early B cell differentiation

    Curr Opin Immunol

    (2010)
  • M. Kondo et al.

    Bcl-2 rescues T lymphopoiesis, but not B or NK cell development, in common gamma chain-deficient mice

    Immunity

    (1997)
  • R.P. DeKoter et al.

    PU.1 regulates expression of the interleukin-7 receptor in lymphoid progenitors

    Immunity

    (2002)
  • O.J. Borge et al.

    Lymphoid-restricted development from multipotent candidate murine stem cells: distinct and complimentary functions of the c-kit and flt3-ligands

    Blood

    (1999)
  • C.J. Spooner et al.

    A recurrent network involving the transcription factors PU.1 and Gfi1 orchestrates innate and adaptive immune cell fates

    Immunity

    (2009)
  • J. Downward

    PI 3-kinase, Akt and cell survival

    Semin Cell Dev Biol

    (2004)
  • K. Johnson et al.

    Regulation of immunoglobulin light-chain recombination by the transcription factor IRF-4 and the attenuation of interleukin-7 signaling

    Immunity

    (2008)
  • T.C. Kuo et al.

    Mechanisms controlling expression of the RAG locus during lymphocyte development

    Curr Opin Immunol

    (2009)
  • A.S. Lazorchak et al.

    Sin1-mTORC2 suppresses rag and il7r gene expression through Akt2 in B cells

    Mol Cell

    (2010)
  • H. Jiang et al.

    Ubiquitylation of RAG-2 by Skp2–SCF links destruction of the V(D)J recombinase to the cell cycle

    Mol Cell

    (2005)
  • J.J. Peschon et al.

    Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice

    J Exp Med

    (1994)
  • U. von Freeden-Jeffry et al.

    Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine

    J Exp Med

    (1995)
  • C.A. Vosshenrich et al.

    Thymic stromal-derived lymphopoietin distinguishes fetal from adult B cell development

    Nat Immunol

    (2003)
  • C.A. Vosshenrich et al.

    Pre-B cell receptor expression is necessary for thymic stromal lymphopoietin responsiveness in the bone marrow but not in the liver environment

    Proc Natl Acad Sci U S A

    (2004)
  • C.T. Jensen et al.

    TSLP-mediated fetal B lymphopoiesis?

    Nat Immunol

    (2007)
  • B.E. Rich et al.

    Cutaneous lymphoproliferation and lymphomas in interleukin 7 transgenic mice

    J Exp Med

    (1993)
  • A. Puel et al.

    expression in T(−)B(+)NK(+) severe combined immunodeficiency

    Nat Genet

    (1998)
  • J.R. Pribyl et al.

    In vitro studies of human B lymphopoiesis

    Ann N Y Acad Sci

    (1995)
  • Y.K. Parrish et al.

    IL-7 dependence in human B lymphopoiesis increases during progression of ontogeny from cord blood to bone marrow

    J Immunol

    (2009)
  • S.E. Johnson et al.

    Murine and human IL-7 activate STAT5 and induce proliferation of normal human pro-B cells

    J Immunol

    (2005)
  • S. Giliani et al.

    Interleukin-7 receptor alpha (IL-7Ralpha) deficiency: cellular and molecular bases. Analysis of clinical, immunological, and molecular features in 16 novel patients

    Immunol Rev

    (2005)
  • A. Yoshimura et al.

    SOCS proteins, cytokine signalling and immune regulation

    Nat Rev Immunol

    (2007)
  • P. Macchi et al.

    Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID)

    Nature

    (1995)
  • T. Nosaka et al.

    Defective lymphoid development in mice lacking Jak3

    Science

    (1995)
  • D.C. Thomis et al.

    Defects in B lymphocyte maturation and T lymphocyte activation in mice lacking Jak3

    Science

    (1995)
  • D.C. van der Plas et al.

    Interleukin-7 signaling in human B cell precursor acute lymphoblastic leukemia cells and murine BAF3 cells involves activation of STAT1 and STAT5 mediated via the interleukin-7 receptor alpha chain

    Leukemia

    (1996)
  • L.M. Heltemes-Harris et al.

    The role of STAT5 in the development, function, and transformation of B and T lymphocytes

    Ann N Y Acad Sci

    (2011)
  • M. Mandal et al.

    Ras orchestrates exit from the cell cycle and light-chain recombination during early B cell development

    Nat Immunol

    (2009)
  • Y. Cui et al.

    Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation

    Mol Cell Biol

    (2004)
  • Z. Yao et al.

    Stat5a/b are essential for normal lymphoid development and differentiation

    Proc Natl Acad Sci U S A

    (2006)
  • C.A. Goetz et al.

    STAT5 activation underlies IL7 receptor-dependent B cell development

    J Immunol

    (2004)
  • S. Malin et al.

    Role of STAT5 in controlling cell survival and immunoglobulin gene recombination during pro-B cell development

    Nat Immunol

    (2010)
  • D.E. Isaksen et al.

    Uncoupling of proliferation and Stat5 activation in thymic stromal lymphopoietin-mediated signal transduction

    J Immunol

    (2002)
  • P. Seckinger et al.

    Activation of src family kinases in human pre-B cells by IL-7

    J Immunol

    (1994)
  • E. Ingley et al.

    Cross-regulation of JAK and Src kinases

    Growth Factors

    (2006)
  • A.R. Venkitaraman et al.

    Interleukin-7 induces the association of phosphatidylinositol 3-kinase with the alpha chain of the interleukin-7 receptor

    Eur J Immunol

    (1994)
  • A.E. Corcoran et al.

    The interleukin-7 receptor alpha chain transmits distinct signals for proliferation and differentiation during B lymphopoiesis

    EMBO J

    (1996)
  • Cited by (136)

    • Co-administration of chicken IL-7 or NK-lysin peptide 2 enhances the efficacy of Eimeria elongation factor-1α vaccination against Eimeria maxima infection in broiler chickens

      2022, Poultry Science
      Citation Excerpt :

      The increase in the serum IL-7 level after recombinant chIL-7 administration (VAC 2 and VAC 3) is expected to be due to the autocrine function of IL-7 (Cattaruzza et al., 2009), but further studies are required to understand its exact mechanism on IL-7 enhancement of host immunity in chickens. Since the main function of IL-7 is to play a pivotal role in the development of B and T cells (Gao et al., 2015; Corfe and Paige, 2012), the increased IL-7 level in serum is expected to improve the protective host vaccinal immune response. cNK-2 is a cationic amphiphilic antimicrobial peptide produced by cytotoxic T cells and natural killer cells (Kim et al., 2017).

    View all citing articles on Scopus
    View full text