Abstract

In response to antigenic stimulation B cells undergo class switch recombination (CSR) at the immunoglobulin heavy chain (IgH) to replace the primary IgM/IgD isotypes by IgG, IgE, or IgA. CSR is initiated by activation-induced cytidine deaminase (AID) through the deamination of cytosine residues at the switch (S) regions of IgH. B cell stimulation promotes germline transcription (GLT) of specific S regions, a necessary event prior to CSR because it facilitates AID access to S regions. Here, we show that CCCTC-binding factor (CTCF)-deficient mice are severely impaired in the generation of germinal center B cells and plasma cells after immunization in vivo, most likely due to impaired cell survival. Importantly, we find that CTCF-deficient B cells have an increased rate of CSR under various stimulation conditions in vitro. This effect is not secondary to altered cell proliferation or AID expression in CTCF-deficient cells. Instead, we find that CTCF-deficient B cells harbor an increased mutation frequency at switch regions, probably reflecting an increased accessibility of AID to IgH in the absence of CTCF. Moreover, CTCF deficiency triggers premature GLT of S regions in naïve B cells. Our results indicate that CTCF restricts CSR by enforcing GLT silencing and limiting AID access to IgH.

Highlights

  • B cells remodel their immunoglobulin genes first as part of their differentiation program in the bone marrow, and during the immune response, in the context of germinal centers [1]

  • B cells that have been activated by antigen engage in the germinal center reaction where immunoglobulin genes are further diversified by somatic hypermutation (SHM) and class switch recombination (CSR), both of which are initiated by the activation-induced cytidine deaminase (AID) enzyme [4,5,6]

  • To analyze the role of CCCTCbinding factor (CTCF) in de novo germinal center formation, we immunized CTCFfl/fl and CTCFfl/+ mice with sheep red blood cells and analyzed spleen populations by flow cytometry 7 days after immunization. We found that both Fas+GL7+ germinal center B cells and CD138+ plasma cells were virtually absent in CTCFfl/fl mice (Figure 1B), further indicating that CTCF is absolutely required for germinal center formation, and concomitantly, for plasma cell generation

Read more

Summary

Introduction

B cells remodel their immunoglobulin genes first as part of their differentiation program in the bone marrow, and during the immune response, in the context of germinal centers [1]. The heavy (IgH) and the light chain (IgL) immunoglobulin genes are subject to V(D)J recombination, a site specific recombination reaction triggered by RAG recombinases [2, 3]. B cells that have been activated by antigen engage in the germinal center reaction where immunoglobulin genes are further diversified by somatic hypermutation (SHM) and class switch recombination (CSR), both of which are initiated by the activation-induced cytidine deaminase (AID) enzyme [4,5,6]. CSR to a particular isotype is determined by stimulation-driven cues that activate specific S region I promoters, inducing their germline transcription (GLT) [10]. GLT of both the donor and acceptor S regions is an absolute requirement for CSR, presumably by promoting transcription-coupled AID recruitment [11]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call