Human CD22 inhibits murine B cell receptor activation in a human CD22 transgenic mouse model

KJ Bednar, E Shanina, R Ballet… - The Journal of …, 2017 - journals.aai.org
KJ Bednar, E Shanina, R Ballet, EP Connors, S Duan, J Juan, BM Arlian, MD Kulis…
The Journal of Immunology, 2017journals.aai.org
CD22, a sialic acid–binding Ig-type lectin (Siglec) family member, is an inhibitory coreceptor
of the BCR with established roles in health and disease. The restricted expression pattern of
CD22 on B cells and most B cell lymphomas has made CD22 a therapeutic target for B cell–
mediated diseases. Models to better understand how in vivo targeting of CD22 translates to
human disease are needed. In this article, we report the development of a transgenic mouse
expressing human CD22 (hCD22) in B cells and assess its ability to functionally substitute …
CD22, a sialic acid–binding Ig-type lectin (Siglec) family member, is an inhibitory coreceptor of the BCR with established roles in health and disease. The restricted expression pattern of CD22 on B cells and most B cell lymphomas has made CD22 a therapeutic target for B cell–mediated diseases. Models to better understand how in vivo targeting of CD22 translates to human disease are needed. In this article, we report the development of a transgenic mouse expressing human CD22 (hCD22) in B cells and assess its ability to functionally substitute for murine CD22 (mCD22) for regulation of BCR signaling, Ab responses, homing, and tolerance. Expression of hCD22 on transgenic murine B cells is comparable to expression on human primary B cells, and it colocalizes with mCD22 on the cell surface. Murine B cells expressing only hCD22 have identical calcium (Ca 2+) flux responses to anti-IgM as mCD22-expressing wild-type B cells. Furthermore, hCD22 transgenic mice on an mCD22−/− background have restored levels of marginal zone B cells and Ab responses compared with deficiencies observed in CD22−/− mice. Consistent with these observations, hCD22 transgenic mice develop normal humoral responses in a peanut allergy oral sensitization model. Homing of B cells to Peyer’s patches was partially rescued by expression of hCD22 compared with CD22−/− B cells, although not to wild-type levels. Notably, Siglec-engaging antigenic liposomes formulated with an hCD22 ligand were shown to prevent B cell activation, increase cell death, and induce tolerance in vivo. This hCD22 transgenic mouse will be a valuable model for investigating the function of hCD22 and preclinical studies targeting hCD22.
journals.aai.org