Abstract
Follicular B (FOB) and marginal zone B (MZB) cells are pivotal in humoral immune responses against pathogenic infections. MZB cells can exacerbate endotoxic shock via interleukin-6 secretion. Here we show that the transcriptional repressor capicua (CIC) and its binding partner, ataxin-1-like (ATXN1L), play important roles in FOB and MZB cell development. CIC deficiency reduces the size of both FOB and MZB cell populations, whereas ATXN1L deficiency specifically affects MZB cells. B cell receptor signaling is impaired only in Cic-deficient FOB cells, whereas Notch signaling is disrupted in both Cic-deficient and Atxn1l-deficient MZB cells. Mechanistically, ETV4 de-repression leads to inhibition of Notch1 and Notch2 transcription, thereby inhibiting MZB cell development in B cell-specific Cic-deficient (Cicf/f;Cd19-Cre) and Atxn1l-deficient (Atxn1lf/f;Cd19-Cre) mice. In Cicf/f;Cd19-Cre and Atxn1lf/f; Cd19-Cre mice, humoral immune responses and lipopolysaccharide-induced sepsis progression are attenuated but are restored upon Etv4-deletion. These findings highlight the importance of the CIC-ATXN1L complex in MZB cell development and may provide proof of principle for therapeutic targeting in sepsis.
Subject terms: Marginal zone B cells, Immunogenetics, Morphogen signalling, Sepsis
Follicular and marginal zone B cells differ in their development and in their role in subverting pathogens with marginal zone B cells bearing more innate-like properties, such as production of proinflammatory cytokines. Here authors show via B-cell specific genomic deletion mouse models that the transcriptional repressor capicua (CIC) and its binding partner, ataxin-1-like (ATXN1L), differentially regulate the development and function of these two main mature B cell populations, while playing a shared role in sepsis mediated by marginal zone B cells.
Introduction
B cells are classified as B-1 or B-2 cells depending on their developmental origin1. B-1 cells, which mediate innate-like immunity by secreting natural antibodies, develop primarily in the fetal liver and are found in the pleural and peritoneal cavities1. Murine B-1 (IgMhiIgDloB220loCD19+ CD23-CD43+) cells are subdivided into B-1a (CD5+) and B-1b (CD5-) cells, depending on the CD5 expression on their surface2. B-2 cell development begins in the bone marrow. Pre-pro B cells derived from bone marrow hematopoietic stem cells acquire characteristic B cell surface markers, such as CD19 and B220, respond to cytokine signaling from molecules such as CXCL12, interleukin (IL)−7, and RANKL, and ultimately develop into immature B cells that express both immunoglobulin (Ig)M and IgD3. Immature B cells migrate to the spleen and differentiate into two distinct mature B cell types: one pathway leads to the production of follicular B (FOB, CD19+CD93-CD21intCD23+) cells via transitional type 1 B (T1B, CD19+CD93+B220+CD21loCD23-IgM+) and T2B (CD19+CD93+B220+CD21loCD23+IgM+) cells, whereas the other pathway leads to the generation of marginal zone B (MZB, CD19+CD93-CD21highCD23lo) cells via MZ progenitor (MZP, CD19+CD93+CD21high) cells following T2B cell differentiation4.
The final development into one of the two mature B cell types is determined by the balance among three signaling pathways: B cell receptor (BCR), B cell activating factor (BAFF), and Notch signaling4. A stronger BCR signal is necessary for differentiation into FOB cells, whereas a weaker BCR signal is sufficient for differentiation into MZB cells4–6. BAFF signaling is essential for the differentiation of immature B cells into both FOB and MZB cells, as well as the maintenance and survival of mature B cells4,7,8. Notch signaling is required for MZB cell development9,10. Of the four mammalian Notch receptors, Notch1 and Notch2 are expressed in mature B cells11,12. When Notch receptors interact with stromal cells that express delta-like ligand 1 (DLL1), their extracellular and intracellular domains are sequentially cleaved by ADAM10 and γ-secretase, respectively11. The liberated Notch intracellular domain translocates to the nucleus, where it forms a transcriptional complex with transcription factors such as RBP-J and MAML13. This complex regulates the expression of target genes, including Dtx1, Hes1, Cr2, and Asb213–15.
FOB cells reside within the B cell follicle, whereas MZB cells are located in the MZ of the spleen, positioned between the white and red pulps16. Because of their distinct localization, FOB cells exhibit specificity in immune responses via interaction with T cells, whereas MZB cells are more likely to encounter blood-borne pathogens and primarily engage in T cell-independent (TI) immune responses17. In most cases, FOB cells differentiate into high-affinity antibody-producing plasma cells or memory B cells through the germinal center (GC) reaction18. MZB cells can also enter the GC and differentiate into plasma cells in response to lipid and polysaccharide pathogens circulating in the bloodstream16,19–21. MZB cells are involved in various diseases, such as pathogen infections and sepsis, through their involvement in cell-to-cell interactions and cytokine secretion22–25.
Capicua (CIC) is an evolutionarily conserved transcriptional repressor that exists in two isoforms: CIC-S and CIC-L, of which CIC-L contains a unique amino-terminal region26,27. CIC directly binds to T(G/C)AATG(A/G)(A/G) sequences via its high mobility group box and C1 domain28,29. Representative CIC target genes include the Pea3 group genes (Etv1, Etv4, and Etv5) and genes encoding negative regulators of the RAS-MAPK pathway, such as Dusp4, Dusp6, Spry4, Spred1, and Spred226. CIC is stabilized by interacting with either ataxin-1 (ATXN1) or its paralog, ataxin-1-like (ATXN1L); ATXN1L plays a predominant role in stabilizing CIC compared with ATXN130,31. CIC functions as a tumor suppressor in various cancer types32–37. CIC also plays a crucial role in the immune system via regulating thymic T cell development and the differentiation of follicular helper T cells and liver-resident memory-like CD8+ T cells38–42. Additionally, CIC regulates B cell development. A CIC deficiency in B cells expands the B-1a cell population at the expense of B-2 cells43,44. This expansion in Cic-deficient mice is attributed to the upregulation of Bhlhe41, a key transcription factor for B-1a cell development, induced by the de-repression of Per2, a CIC target gene43,45. However, the regulatory mechanism of CIC influencing B-2 cell development, including that of FOB and MZB cells, remains unclear.
In this study, we examine the role of the CIC-ATXN1L complex in B-2 cell development. Our findings indicate that the CIC-ATXN1L complex regulates MZB cell development by modulating Notch signaling. In addition, we identify Etv4 as a CIC target gene that inhibits Notch signaling and MZB cell development.
Results
ATXN1L deficiency suppresses MZB cell development
Our previous study has shown that the population of B-1a cells significantly expands in B cell-specific Cic-null (Cicf/f;Cd19-Cre) mice, whereas those of FOB and MZB cells decrease43 (Supplementary Fig. S1a, b). Didonna et al. showed that the B-1 cell frequency increased in the spleens of Atxn1−/− mice, whereas that of MZB cells decreased46, which was confirmed by our data (Supplementary Fig. S1c–f). However, the role of ATXN1L, a protein that stabilizes and interacts with CIC, in B cells remains unclear. To elucidate the role of ATXN1L in B cell development and function, we generated and characterized B cell-specific Atxn1l-null (Atxn1lf/f;Cd19-Cre) mice. In contrast to Cicf/f;Cd19-Cre and Atxn1−/− mice (Supplementary Fig. S1a–c), the B-1 cell populations did not expand in the peritoneal cavities and spleens of Atxn1lf/f;Cd19-Cre mice (Fig. 1a). In the spleens of Atxn1lf/f;Cd19-Cre mice, the MZB and MZP cell populations were significantly reduced, whereas the T1B, T2B, and FOB cell formation remained normal (Fig. 1b–d). These results indicate that ATXN1L deficiency in B cells specifically impairs MZB cell development. Notably, we found that ATXN1L levels were higher in MZB cells than in FOB cells (Fig. 1e).
Fig. 1. ATXN1L deficiency specifically reduces MZP and MZB cell populations.
a Flow cytometry of B-1a (CD19+CD5+CD43+) and B-1b (CD19+CD5-CD43+) cells in the peritoneal cavities and spleens of Cd19-Cre and Atxn1lf/f;Cd19-Cre mice. For peritoneal cavity analysis, N = 5 per both mice. For spleen analysis, N = 7 for Cd19-Cre and N = 5 for Atxn1lf/f;Cd19-Cre mice. Flow cytometry of FOB and MZB cells (b), MZP cells (c), and T1B and T2B cells (d) in the spleens of Cd19-Cre and Atxn1lf/f;Cd19-Cre mice. FOB (CD19+CD93-CD21intCD23+) and MZB (CD19+CD93-CD21highCD23lo) cells were initially gated as CD93- (AA4.1). T1B (CD19+CD93+B220+CD21loCD23-IgM+), T2B (CD19+CD93+B220+CD21midCD23+IgM+), and MZP (CD19+CD93+CD21high) cells were gated as CD93+ (AA4.1). N = 7 for Cd19-Cre and N = 5 for Atxn1lf/f;Cd19-Cre. e Western blotting to detect the levels of CIC, ATXN1, and ATXN1L in FOB, MZB, and B-1a cells. FOB and MZB cells were prepared from the spleens of C57BL/6 mice, whereas B-1a cells were isolated from the peritoneal cavities of the same mice. Data represent 2–3 independent experiments. Statistics: two-tailed Student’s t-test (a–d). Bar graphs present the data as mean ± S.D. Ctrl: Cd19-Cre and A1L cKO: Atxn1lf/f;Cd19-Cre. MZP marginal zone progenitor cells, MZB marginal zone B cells, and FOB follicular B cells. Source data are provided as a Source Data file.
Humoral immune responses to antigens are attenuated in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice
MZB cells respond rapidly to TI antigens; however, they can also engage in a slower T cell-dependent (TD) immune response that is primarily mediated by FOB cells16. To investigate the effects of CIC or ATXN1L deficiency-induced alterations in B cell development on humoral immune responses, Cd19-Cre (control), Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice were immunized with the TI antigen NP-Ficoll and the TD antigen ovalbumin (OVA) via intravenous and intraperitoneal injections, respectively. Seven days after immunization, the mice were analyzed for serum NP-specific or OVA-specific IgM and IgG levels, as well as immune cell frequencies. Upon immunization with NP-Ficoll, the serum NP-specific IgM levels and Blimp1+CD138+ plasma cell populations were significantly decreased in both Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice compared to control mice (Fig. 2a, b), suggesting that ttenuation of MZB cell development caused by the CIC and ATXN1L deficiency suppressed humoral immune responses to TI antigens. Upon immunization with OVA, both OVA-specific IgM and IgG levels were significantly reduced in Atxn1lf/f;Cd19-Cre and Cicf/f;Cd19-Cre mice (Fig. 2c). Consistent with these results, the populations of GCB and plasma cells were reduced in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice (Fig. 2d, e). The immune response to OVA was more robust in Cicf/f;Cd19-Cre mice than that in Atxn1lf/f;Cd19-Cre mice (Fig. 2c–e), potentially attributed to differences in B cell subset formation between Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice.
Fig. 2. Humoral immune responses are attenuated in Cicf/f;Cd19-Cre and Atxn1l f/f;Cd19-Cre mice.
a, b Cd19-Cre, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice were immunized with NP-Ficoll for 7 days. a Serum NP-specific IgM and IgG levels determined using ELISA. b Flow cytometry of splenic plasma cells. N = 4 for Cd19-Cre and Cicf/f;Cd19-Cre groups, and N = 6 for Cd19-Cre and Atxn1lf/f;Cd19-Cre groups. c–e Cd19-Cre, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice were immunized with OVA in alum for 7 days. c Serum OVA-specific IgM and IgG levels determined using ELISA. d, e Flow cytometry of splenic GCB cells (CD19+GL-7+CD95+) (d) and plasma cells (CD19+CD138+Blimp1+) (e). N = 6 for Cd19-Cre and Cicf/f;Cd19-Cre, and N = 5 for Atxn1lf/f;Cd19-Cre. Data represent two independent experiments. Statistics: two-tailed Student’s t-test (a, b) and one-way ANOVA with Tukey’s multiple comparisons test (c–e). Bar graphs present data as mean ± S.D. Ctrl: Cd19-Cre, Cic cKO: Cicf/f;Cd19-Cre, and A1L cKO: Atxn1lf/f;Cd19-Cre. ELISA enzyme-linked immunosorbent assay, OVA ovalbumin, and GCB germinal center B cells. Source data are provided as a Source Data file.
Lipopolysaccharide (LPS)-induced sepsis progression is attenuated in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice
As MZB cells promote LPS-induced sepsis progression via IL-6 secretion induced by Toll-like receptor 4 signaling22, we investigated sepsis progression in control, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice following intravenous LPS injections. Most control mice died 72 h after the injection, whereas more than 50% Cicf/f;Cd19-Cre mice survived (Fig. 3a). The serum IL-6 levels were significantly decreased in Cicf/f;Cd19-Cre mice compared to those in control mice, whereas IL-10 levels were comparable between control and Cicf/f;Cd19-Cre mice 12 h after LPS injection (Fig. 3b). Simultaneously, immune cell infiltration into the lung tissue was significantly reduced in Cicf/f;Cd19-Cre mice compared to that in control mice (Fig. 3c). Similar results were observed in LPS-treated Atxn1lf/f;Cd19-Cre mice (Fig. 3d–f). These data demonstrate that a CIC-ATXN1L complex deficiency in B cells attenuates the inflammatory response during endotoxic shock, thereby alleviating sepsis severity.
Fig. 3. LPS-induced sepsis progression is attenuated in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice.
a–c Female Cd19-Cre and Cicf/f;Cd19-Cre mice were injected intravenously with LPS. Serum samples and lung tissues were collected from Cd19-Cre and Cicf/f;Cd19-Cre mice 12 h after LPS injection. a Survival rate of LPS-treated mice. Mortality was monitored every 24 h. N = 14 per group. b Serum IL-6 and IL-10 levels determined using ELISA. N = 4 per group. c Histology of immune cell infiltration into lung tissue. Arrows indicate immune cell-infiltrated regions. N = 3 per group. Scale bar: 100 µm. d–f Female Cd19-Cre and Atxn1lf/f;Cd19-Cre mice were injected intravenously with LPS. Serum samples and lung tissues were collected from Cd19-Cre and Atxn1lf/f;Cd19-Cre mice 12 h after LPS injection. d Survival rate of LPS-treated mice. Mortality was monitored every 24 h. N = 12 for Cd19-Cre and N = 13 for Atxn1lf/f;Cd19-Cre. e Serum IL-6 and IL-10 levels determined using ELISA. N = 3 per group. f Histology of immune cell infiltration into lung tissue. Arrows indicate immune cell-infiltrated regions. N = 4 for Cd19-Cre and N = 3 for Atxn1lf/f;Cd19-Cre. Scale bar: 100 µm. Data represent 2–3 independent experiments. Statistics: Log-rank (Mantel-Cox) test (a, d) and two-tailed Student’s t-test (b, c, e, f). Bar graphs present data as mean ± S.D. Ctrl: Cd19-Cre, Cic cKO: Cicf/f;Cd19-Cre, and A1L cKO: Atxn1lf/f;Cd19-Cre. LPS lipopolysaccharide, IL interleukin, and ELISA enzyme-linked immunosorbent assay. Source data are provided as a Source Data file.
BCR signaling is attenuated in Cic-null FOB cells but not in Atxn1l-null FOB cells
We found that both FOB and MZB cell populations were reduced in Cicf/f;Cd19-Cre mice, whereas only the MZB cell population was diminished in Atxn1lf/f;Cd19-Cre mice. To examine the specific reduction in FOB cell populations of Cicf/f;Cd19-Cre mice, we characterized T2B and FOB cells from control, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice. Control and Cic-null T2B cells differentiated into FOB cells with similar efficiency upon anti-IgM treatment in vitro (Supplementary Fig. S2a). The apoptosis rate was comparable between control and Cic-null FOB cells in vitro (Supplementary Fig. S2b). The frequency of Ki-67+ FOB cells was higher in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice than in control mice (Supplementary Fig. S2c, d). BAFF receptor signaling promotes B cell survival and T2B cell differentiation into FOB and MZB cells4; BAFF receptor levels were not reduced in Cic-null or Atxn1l-null T2B and FOB cells compared with those of their respective control cells (Supplementary Fig. S2e, f). These data demonstrate that the reduced FOB cell population in Cicf/f;Cd19-Cre mice is not attributed to defects in T2B cell differentiation into FOB cells or in the survival and proliferation of FOB cells.
BCR signaling is crucial for peripheral B cell maintenance47,48; therefore, we examined the BCR signaling strength in FOB cells from control, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice. Both tonic and anti-IgM-stimulated BCR signaling were significantly attenuated in Cic-null FOB cells compared with those in control cells (Fig. 4a). In contrast, these defects were rarely found in Atxn1l-null FOB cells (Fig. 4b). These results suggest that the attenuation of BCR signaling caused by CIC deficiency may decrease the FOB cell population.
Fig. 4. BCR signaling is attenuated in Cic-null FOB cells.
Analysis of anti-IgM-stimulated and tonic BCR signaling in Cic-null (a) and Atxn1l-null (b) FOB cells. Phosphorylated-ERK (pERK), phosphorylated-BTK (pBTK), phosphorylated-BLNK (pBLNK), and phosphorylated-AKT (pAKT) levels were determined using flow cytometry and presented as MFI. For the BCR signaling analysis in control and Cic-null FOB cells, N = 6 per group (a). For the analysis of pERK and pBTK levels in control and Atxn1l-null FOB cells stimulated with anti-IgM, N = 4 per group (b). For the analysis of pBLNK levels in control and Atxn1l-null FOB cells stimulated with anti-IgM, N = 3 per group (b). For the analysis of pAKT levels in control and Atxn1l-null FOB cells stimulated with anti-IgM, N = 4 for Cd19-Cre and N = 3 for Atxn1lf/f;Cd19-Cre (b). For the tonic BCR signaling analysis in control and Atxn1l-null FOB cells, N = 8 for Cd19-Cre and N = 6 for Atxn1lf/f;Cd19-Cre (b). Data represent 2–3 independent experiments. Statistics: two-tailed Student’s t-test (a, b). Bar graphs present data as mean ± S.D. Ctrl: Cd19-Cre, Cic cKO: Cicf/f;Cd19-Cre, and A1L cKO: Atxn1lf/f;Cd19-Cre. BCR B cell receptor, MFI mean fluorescence intensity, and FOB, follicular B cells. Source data are provided as a Source Data file.
Notch signaling is downregulated because of the CIC-ATXN1L complex deficiency
To understand the mechanism underlying the CIC or ATXN1L deficiency inhibiting MZB cell development, we characterized MZB cells from control, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice. Apoptosis rates were significantly reduced in Cic-null or Atxn1l-null MZB cells compared with those in control cells in vitro (Supplementary Fig. S2g, h). Similar to the results for FOB cells (Supplementary Fig. S2c, d), the frequencies of Ki-67+ MZB cells were increased in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice compared with that in control mice (Supplementary Fig. S2i, j). BAFF receptor levels were higher in Cic-null and Atxn1l-null MZB cells than in control cells (Supplementary Fig. S2e, f). The phosphorylation of BLNK and BTK following BCR stimulation showed similar patterns in control, Cic-null, and Atxn1l-null MZB cells (Supplementary Fig. S2k, l). These data suggest that the reduced MZB cell population caused by the CIC and ATXN1L deficiency is not attributed to defects in cell proliferation, cell viability, or BCR signaling.
We then examined the gene expression profiles in control and Cic-null MZB cells using RNA sequencing. In total, 775 genes were differentially expressed in Cic-null MZB cells (505 upregulated and 270 downregulated; Supplementary Data 1) compared with the expression in control cells (log2 fold-change > 0.5 and P < 0.01). Several known CIC target genes, including Etv4, Etv5, Spry4, Spred1, and Dusp4, were among the upregulated genes in Cic-null MZB cells (Fig. 5a and Supplementary Data 1). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and gene ontology (GO) analyses of differentially expressed genes (DEGs) in Cic-null MZB cells revealed that the Notch signaling pathway was significantly downregulated in these cells (Supplementary Fig. S3 and Supplementary Data 2 and 3). Gene set enrichment analysis (GSEA) also showed that the Notch signaling-related genes were downregulated in Cic-null MZB cells (Fig. 5b). We validated the downregulation of genes involved in the Notch signaling pathway, including Notch1, Notch2, Hes1, Dtx1, Asb2, and Cr2, in Cic-null and Atxn1l-null MZB cells using real-time quantitative polymerase chain reaction (RT-qPCR; Fig. 5c, d). At the protein level, NOTCH1, NOTCH2, and CD21 (encoded by Cr2) expression was downregulated in Cic-null and Atxn1l-null MZP and MZB cells (Fig. 5e, f). To evaluate the differentiation potential of Cic-null and Atxn1l-null T1B cells into MZB cells, we co-cultured T1B cells with control or DLL1-expressing OP9 cells and assessed the efficiency of Notch signaling activation in promoting T1B cell differentiation into MZB cells in vitro. Both Cic-null and Atxn1l-null T1B cells were less efficient in differentiating into MZB cells compared with control cells (Fig. 5g, h). Additionally, Atxn1 knockdown further reduced the efficiency of MZB cell differentiation in Atxn1l-null T1B cells (Fig. 5i), suggesting redundancy between ATXN1 and ATXN1L in the regulation of Notch signaling-induced MZB cell development. Overall, these results demonstrate that the CIC-ATXN1L complex promotes MZB cell development via activation of Notch signaling.
Fig. 5. Notch signaling is downregulated by CIC-ATXN1L complex deficiency.
a Volcano plots showing differentially expressed genes (DEGs, log2 fold-change > 0.5) in Cic-deficient MZB cells. Two samples of each genotype were subjected to RNA sequencing. Downregulated Notch signaling-related genes and upregulated CIC target genes are shown next to each corresponding dot. b Gene set enrichment analysis (GSEA) of DEGs in Cic-deficient MZB cells. The gene set database HALLMARK_NOTCH_SIGNALING (MM3870) was employed for GSEA. RT-qPCR analysis showing the downregulation of Notch signaling-related genes in Cic-null (c) and Atxn1l-null (d) MZB cells. For Notch1 and Asb2 levels, N = 5 per group; for Notch2, Dtx1, and Hes1 levels, N = 6 per group; and for Cr2 levels, N = 3 per group (c). N = 3 per group (d). e Surface expression levels of NOTCH1, NOTCH2, and CD21 in MZB and MZP cells from Cd19-Cre and Cicf/f;Cd19-Cre mice. N = 6 per group. f Surface expression levels of NOTCH1, NOTCH2, and CD21 in MZB and MZP cells from Cd19-Cre and Atxn1lf/f;Cd19-Cre mice. N = 6 per group. The MFIs for NOTCH1, NOTCH2, and CD21 were determined using flow cytometry. In vitro MZB cell differentiation assay to assess the differentiation potential of Cic-null (g) and Atxn1l-null (h) T1B cells into MZB cells. Sorted T1B cells were co-cultured with either control or DLL1-expressing OP9 cells for 72 h and subjected to flow cytometry to determine the frequency of MZB (CD21hiCD1dhi) cells. N = 6 per group. i In vitro MZB cell differentiation assay using control and Atxn1l-null T1B cells transfected with either control or Atxn1 siRNAs. T1B cells were co-cultured with DLL1-expressing OP9 cells for 72 h. N = 3 per group. Data represent 2–3 independent experiments. Statistics: Kolmogorov–Smirnov test (b) and two-tailed Student’s t-test (c–i). Bar graphs present the data as mean ± S.D. Ctrl: Cd19-Cre, Cic cKO: Cicf/f;Cd19-Cre, and A1L cKO: Atxn1lf/f;Cd19-Cre. MZB marginal zone B cells, CIC capicua, RT-qPCR quantitative real-time polymerase chain reaction, MZP marginal zone progenitor cells, MFI mean fluorescence intensity, and NES normalized enrichment score. Source data are provided as a Source Data file.
ETV4 is a CIC target gene that suppresses Notch signaling and MZB cell development
Next, we identified the CIC target genes that downregulate Notch signaling in Cic-null and Atxn1l-null MZB cells. Notch signaling is reported to be hyperactivated in the lacrimal gland epithelial cells of Etv1/Etv4/Etv5 triple-knockout mice, accompanied by Notch1, Notch2, and Notch3 overexpression49. Therefore, we hypothesized that the de-repression of Etv4 and Etv5 downregulated the Notch1 and Notch2 expression in Cic-null and Atxn1l-null MZB cells. The RT-qPCR analysis confirmed the significant upregulation of Etv4 and Etv5 expression in both Cic-null and Atxn1l-null MZB cells compared with the expression in control cells (Fig. 6a, b). To determine ETV4- and ETV5-induced regulation of Notch1 and Notch2 expression at the transcription level, we constructed luciferase reporters with either Notch1 or Notch2 promoter regions (upstream 1 kb from the transcription start site; Supplementary Fig. S4a, b). ETV4 suppressed Notch1 and Notch2 promoter activity in HEK293T and HeLa cells, respectively (Supplementary Fig. S4c), but ETV5 did not (Supplementary Fig. S4d and e). ETV4 did not inhibit Notch1 and Notch2 promoter activity when the cell lines were switched (Supplementary Fig. S4f, g), suggesting a cell type-specific suppression of Notch1 and Notch2 expression by ETV4. We also identified ETV4 binding motifs (5′-GGAA-3′) that contributed to ETV4-mediated repression of Notch1 and Notch2 promoter activity. When the second ETV4 binding motif was mutated, ETV4 could no longer repress Notch1 promoter activity (Supplementary Fig. S4a–c). Disruption of both ETV4 binding motifs almost completely abolished the inhibitory effect of ETV4 overexpression on Notch2 promoter activity (Supplementary Fig. S4b, c). To determine whether ETV4 overexpression downregulates Notch1 and Notch2 expression in MZB cells, we infected these cells with a retrovirus expressing ETV4 and analyzed NOTCH1 and NOTCH2 expression using flow cytometry. MZB cells infected with the ETV4-expressing virus exhibited a reduced surface expression of both NOTCH1 and NOTCH2 (Fig. 6c). Additionally, we found that ETV4 binds to the promoter regions of Notch1 and Notch2, which contain ETV4 binding motifs, in Cic-null B-2 cells (Fig. 6d and Supplementary Fig. S4a, b). These results demonstrate that ETV4 functions as a transcriptional repressor of Notch1 and Notch2 expression in MZB cells.
Fig. 6. Etv4 is a CIC target gene that suppresses Notch signaling and MZB cell development.
RT-qPCR analysis of Etv4 and Etv5 levels in MZB cells from Cicf/f;Cd19-Cre (a) and Atxn1lf/f;Cd19-Cre (b) mice. N = 3 per group. c Flow cytometry of NOTCH1 and NOTCH2 levels in MZB cells infected with either a control (NC) or ETV4-expressing (ETV4) retrovirus. N = 6 per group. d ChIP-qPCR analysis of the Notch1 and Notch2 promoter regions containing ETV4 binding motifs. B-2 cells isolated from the spleens of Cicf/f;Cd19-Cre mice were subjected to ChIP using either IgG or an anti-ETV4 antibody. N = 3 per group. Flow cytometry of splenic FOB and MZB cells (e), MZP cells (f), and T1B and T2B cells (g) in Cd19-Cre, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice. N = 7 for Cd19-Cre, N = 8 for Cicf/f;Cd19-Cre, and N = 5 for Etv4−/−;Cicf/f;Cd19-Cre. Ctrl: Cd19-Cre, cKO: Cicf/f;Cd19-Cre, and DKO: Etv4−/−;Cicf/f;Cd19-Cre. Flow cytometry of splenic FOB and MZB cells (h) and MZP cells (i) in Cd19-Cre, Atxn1lf/f;Cd19-Cre, and Etv4−/−;Atxn1lf/f;Cd19-Cre mice. N = 8 for Cd19-Cre, N = 6 for Atxn1lf/f;Cd19-Cre, and N = 3 for Etv4−/−;Atxn1lf/f;Cd19-Cre. Ctrl: Cd19-Cre, cKO: Atxn1lf/f;Cd19-Cre, and DKO: Etv4−/−;Atxn1lf/f;Cd19-Cre. j Surface expression levels of NOTCH1, NOTCH2, and CD21 in MZB and MZP cells from Cd19-Cre, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice. N = 5 per group. k Surface expression levels of NOTCH1, NOTCH2, and CD21 in MZB and MZP cells from Cd19-Cre, Atxn1lf/f;Cd19-Cre, and Etv4−/−;Atxn1lf/f;Cd19-Cre mice. N = 5 for Cd19-Cre and Atxn1lf/f;Cd19-Cre, and N = 3 for Etv4−/−;Atxn1lf/f;Cd19-Cre. The MFIs for NOTCH1, NOTCH2, and CD21 were determined using flow cytometry. Data represent 2–3 independent experiments. Statistics: two-tailed Student’s t-test (a–d) and one-way ANOVA with Tukey’s multiple comparisons test (e–k). Bar graphs present data as mean ± S.D. RT-qPCR quantitative real-time polymerase chain reaction, FOB follicular B cells, MZB marginal zone B cells, MZP marginal zone progenitor cells, and MFI mean fluorescence intensity. Source data are provided as a Source Data file.
To determine the suppression of Notch signaling and MZB cell development via CIC or ATXN1L deficiency-meditated ETV4 de-repression, we generated Etv4 and Cic (Etv4−/−;Cicf/f;Cd19-Cre) or Atxn1l (Etv4−/−;Atxn1lf/f;Cd19-Cre) double mutant mice. The MZB and MZP cell populations were restored to control mouse levels in Etv4−/−;Cicf/f;Cd19-Cre mice (Fig. 6e, f). In contrast, Etv4 allele deletion did not rescue the FOB, GCB, and B-1a cell phenotypes in Cicf/f;Cd19-Cre mice (Fig. 6e and Supplementary Fig. S5), indicating that the regulation of B-1a, MZB, and FOB cell development by CIC was mediated by differential molecular mechanisms43. The frequency of T2B cells was significantly restored in Etv4−/−;Cicf/f;Cd19-Cre mice (Fig. 6g). The MZB and MZP cell populations were also recovered in Etv4−/−;Atxn1lf/f;Cd19-Cre mice (Fig. 6h, i). Furthermore, ETV4 deficiency significantly restored NOTCH1, NOTCH2, and CD21 levels in Cic-null and Atxn1l-null MZB and MZP cells (Fig. 6j, k). Conversely, the loss of Etv5 did not rescue the FOB, MZB, MZP, and T2B cell population decrease caused by CIC deficiency (Supplementary Fig. S6). Notably, the formation of FOB, MZP, and MZB cells and the expression of NOTCH1 and NOTCH2 in these subsets were comparable between wild-type (WT) and Etv4−/− mice (Supplementary Fig. S7), indicating that ETV4 is not essential for the regulation of B cell development and Notch signaling under steady-state conditions. Collectively, these results indicate that ETV4 de-repression due to the CIC-ATXN1L complex deficiency suppresses Notch1 and Notch2 expression, consequently causing defects in MZB cell development.
The diminished IgM production and attenuated sepsis progression in Cicf/f;Cd19-Cre mice are caused by defects in MZB cell development
The defects in MZB cell development in Cicf/f;Cd19-Cre mice were specifically recovered via Etv4 allele deletion; however, it remains unclear whether these defects are the cause for the attenuated humoral immune responses and sepsis progression in Cicf/f;Cd19-Cre mice. For clarification, we analyzed the serum levels of NP-specific or OVA-specific IgM and IgG and the frequencies of immune cells in control, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice immunized with NP-Ficoll or OVA. The decreased Blimp1+CD138+ plasma cell populations and serum NP-specific IgM levels in Cicf/f;Cd19-Cre mice immunized with NP-Ficoll were significantly restored by ETV4 deficiency (Fig. 7a, b), suggesting that the attenuated humoral immune response to TI antigens was caused by defects in MZB cell development in Cicf/f;Cd19-Cre mice. Upon OVA immunization, Etv4−/−;Cicf/f;Cd19-Cre mice did not restore the GCB and plasma cell populations, and the serum anti-OVA IgG levels did not increase, in comparison with the observed effects in Cicf/f;Cd19-Cre mice (Fig. 7c–e). However, the serum anti-OVA IgM levels were significantly restored in Etv4−/−;Cicf/f;Cd19-Cre mice (Fig. 7e), suggesting that the impaired IgM production observed in Cicf/f;Cd19-Cre mice following TD antigen immunization was primarily attributed to MZB cell development defects.
Fig. 7. Restoration of IgM production and sepsis progression in Etv4−/−;Cicf/f;Cd19-Cre mice.
a, b Cd19-Cre, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice were immunized with NP-Ficoll for 7 days. a Flow cytometry of splenic plasma cells. N = 5 for Cd19-Cre and Cicf/f;Cd19-Cre, and N = 4 for Etv4−/−;Cicf/f;Cd19-Cre. b Serum NP-specific IgM and IgG levels determined using ELISA. N = 5 for Cd19-Cre and N = 4 for Cicf/f;Cd19-Cre and Etv4−/−;Cicf/f;Cd19-Cre. c–e Cd19-Cre, Cicf/f;Cd19-Cre, and Etv4−/−;Cd19-Cre mice were immunized with OVA in alum for 7 days. Flow cytometry of splenic GCB cells (c) and plasma cells (d). N = 7 for Cd19-Cre, N = 8 for Cicf/f;Cd19-Cre, and N = 6 for Etv4−/−;Cicf/f;Cd19-Cre. e Serum OVA-specific IgM and IgG levels determined using ELISA. N = 7 for Cd19-Cre, N = 8 for Cicf/f;Cd19-Cre, and N = 5 for Etv4−/−;Cicf/f;Cd19-Cre. f–h Female Cd19-Cre, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice were injected intravenously with LPS. f Survival rate of LPS-treated mice. Mortality was monitored every 24 h. N = 7 for Cd19-Cre and Etv4−/−;Cicf/f;Cd19-Cre, and N = 10 for Cicf/f;Cd19-Cre. g Serum IL-6 and IL-10 levels determined using ELISA. h Histology of immune cell infiltration into lung tissue. Arrows indicate immune cell-infiltrated regions. Scale bar: 100 µm. Serum samples and lung tissues were collected from the mice 12 h after LPS injection. N = 5 per group. Data represent 2–3 independent experiments. Statistics: one-way ANOVA with Tukey’s multiple comparisons test (a–e, g–h) and Log-rank (Mantel-Cox) test (f). The bar graph presents data as mean ± S.D. Ctrl: Cd19-Cre, cKO: Cicf/f;Cd19-Cre, and DKO: Etv4−/−;Cicf/f;Cd19-Cre. ELISA enzyme-linked immunosorbent assay, OVA ovalbumin, and GCB germinal center B cell. Source data are provided as a Source Data file.
We also investigated the LPS-induced sepsis progression in control, Cicf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice. All control and Etv4−/−;Cicf/f;Cd19-Cre mice succumbed to sepsis within 72 h after LPS injection, whereas over 50% of Cicf/f;Cd19-Cre mice survived (Fig. 7f). The diminished IL-6 production and immune cell infiltration into lung tissues observed in Cicf/f;Cd19-Cre mice were significantly restored in Etv4−/−;Cicf/f;Cd19-Cre mice (Fig. 7g, h). These results demonstrate that the mitigation of LPS-induced sepsis progression in Cicf/f;Cd19-Cre mice is caused by defects in MZB cell development.
Discussion
This study investigated the role of CIC and its binding partners, ATXN1 and ATXN1L, in B-2 cell development (Fig. 8). T2B and FOB cell populations were substantially decreased in Cicf/f;Cd19-Cre mice, whereas Atxn1−/− and Atxn1lf/f;Cd19-Cre mice did not exhibit these alterations. This suggests that CIC destabilization caused by the absence of either ATXN1 or ATXN1L may be insufficient to disrupt T2B and FOB cell development. In most tissues and cell types, ATXN1L plays a dominant role in stabilizing CIC compared with ATXN130,31. Consistent with this information, the reduction in MZB cell population was greater in Atxn1lf/f;Cd19-Cre mice (Fig. 1b) than in Atxn1−/− mice (Supplementary Fig. S1d) when compared with their respective control mice. However, the expansion of B-1 cell populations, observed in both Cicf/f;Cd19-Cre and Atxn1−/− mice, was not evident in Atxn1lf/f;Cd19-Cre mice, suggesting that CIC stability may be primarily ensured by ATXN1 rather than ATXN1L in B-1 cells.
Fig. 8. The CIC-ATXN1L complex regulates B-2 cell development.
A schematic diagram illustrates the role of the CIC-ATXN1L complex in B-2 cell development. The left panel shows that CIC deficiency reduces the formation of FOB cells by downregulating BCR signaling. The specific CIC target genes involved in this process have not yet been identified. The right panel indicates that the deficiency of either CIC or ATXN1L downregulates NOTCH1 and NOTCH2 expression by de-repressing ETV4, leading to a decreased MZB cell population. The diagram was created using BioRender.com, which permits the reuse of its content under a CC-BY 4.0 Attribution 4.0 International license.
Our study uncovered the molecular mechanisms underlying the defect in MZB cell development caused by CIC-ATXN1L complex deficiency. Etv4 was the CIC target gene that downregulated Notch signaling in Cic-null and Atxn1l-null MZP and MZB cells. ETV4 also functions as a transcriptional repressor for Notch1 and Notch2 expression in a cell-type-specific manner. Consistent with this result, previous studies have shown that ETV4 overexpression downregulates NOTCH1 expression and upregulates NOTCH2 expression in MDA-MB-231 breast cancer cells50. Many transcription factors play a dual role as transcriptional activators and repressors, depending on their interacting coactivators and corepressors51. Therefore, identifying corepressors that interact with ETV4 and are recruited to Notch1 and Notch2 loci during MZB cell development would be crucial for further understanding the dysregulation of MZB cell development caused by a deficiency in the CIC-ATXN1/ATXN1L complex.
The regulation of Notch signaling by the CIC-ATXN1/ATXN1L complex may involve additional molecular mechanisms beyond ETV4 de-repression. The phosphoinositide-3-kinase (PI3K)-AKT pathway is known to induce Notch-dependent responses in various cell types, including cancer and immune cells52–55. Moreover, Notch signaling can activate the PI3K-AKT pathway through an autocrine loop56. Given the decreased phospho-AKT levels in Cic-null FOB cells, the downregulation of this pathway might contribute to the CIC deficiency-mediated reduction in Notch signaling. On the other hand, it is known that ATXN1 and ATXN1L interact with RBP-J, a key Notch signaling mediator10, and recruit SMRT and HDAC3 to repress the expression of Notch target genes in mammalian cells57. The interaction between RBP-J and ATXN1 or ATXN1L is disrupted by NCID overexpression, leading to the activation of Notch target genes57. Since CIC deficiency destabilizes ATXN1 and ATXN1L30, it is plausible that not only ATXN1L deficiency but also CIC deficiency can enhance RBP-J-mediated activation of Notch signaling. Therefore, the downregulation of Notch signaling observed in Cic-null and Atxn1l-null MZB cells might be the result of these complex molecular alterations.
T2B cells develop into FOB and MZB cells, of which MZB cells specifically require Notch signaling4. Etv4−/−;Cicf/f;Cd19-Cre mice showed that the frequency of T2B cells was restored but the FOB cell population was not (Fig. 6d–f), suggesting that the decrease in FOB cell population was not primarily due to defective T2B cell development in Cicf/f;Cd19-Cre mice. We found that BCR signaling was significantly attenuated in Cic-null FOB cells. However, our previous study has shown that it was enhanced in Cic-null T2B cells43. Similarly, developmental stage-specific attenuation of T cell receptor signaling caused by CIC deficiency has been reported38. The de-repression of CIC target genes that negatively regulated the RAS-MAPK pathway, including Spry4, Spred1, Dusp4, and Dusp6, was proposed as the molecular mechanism behind this phenomenon38. Therefore, we speculate that the impaired FOB cell formation in Cicf/f;Cd19-Cre mice may result from the attenuation of BCR signaling potentially caused by the de-repression of CIC target genes such as Spry4, Spred1, Spred2, and Dusp4, which were observed to be significantly upregulated in Cic-null FOB cells43. T2B cells are normally formed in mice deficient in RBP-J, suggesting that the decreased T2B cell population in Cicf/f;Cd19-Cre mice is not caused by a downregulation of Notch signaling. The observed restoration of T2B cell frequency in Etv4−/−;Cicf/f;Cd19-Cre mice (Fig. 6f) warrants further investigation to elucidate the molecular mechanism underlying the suppression of T2B cell formation via ETV4 overexpression.
MZB cell function conventionally involves protection against blood-borne pathogens via the expression of polyreactive BCRs that bind to multiple microbial molecular patterns. Hence, MZB cells are regarded as innate-like B cells, which mediate the frontline defense against blood-borne microorganisms16. Conversely, MZB cells can exacerbate the progression of LPS- or bacteria-induced sepsis in mice by secreting IL-622,25. While the conventional innate-like features of MZB cells are well-understood, their role in sepsis has not received much attention. Our study confirmed their sepsis-promoting function by assessing sepsis severity in Cicf/f;Cd19-Cre and Atxn1lf/f;Cd19-Cre mice following LPS treatment. Furthermore, the MZB cell population was restored via the deletion of Etv4 alleles, which reversed the attenuation of sepsis progression observed in Cicf/f;Cd19-Cre mice, highlighting the pivotal role of MZB cells in sepsis. However, it remains elusive whether MZB cells promote sepsis progression in humans. Although our study suggests that the CIC-ATXN1/ATXN1L complex and ETV4 are potential therapeutic targets for sepsis, further research is required to investigate the role of human MZB cells in sepsis and the involvement of the CIC-ATXN1/ATXN1L complex in MZB cell development in humans.
Methods
Mice
Cic floxed40,58 (#030555, Jackson Laboratory, USA), Vav1-Cre59 (#035670, Jackson Laboratory), Cd19-Cre60 (#006785, Jackson Laboratory), Atxn1−/−61 (#029025, Jackson Laboratory), and Atxn1l floxed62 (#030717, Jackson Laboratory) mice were generated as described previously. Whole-body Etv4−/− mice were generated using the CRISPR/Cas9 system (Korea Mouse Phenotyping Center, Seoul, Korea). All mice were maintained on a C57BL/6 background. All experiments were conducted with 8–10-week-old mice unless otherwise indicated. For the experimental sepsis model, 8–12-week-old female mice were used. Animals were maintained in a specific pathogen-free animal facility under a standard 12:12 h light/dark cycle and administered standard rodent chow and water ad libitum. All animal procedures performed in this study were ethically approved by the Institutional Animal Care and Use Committee of Pohang University of Science and Technology.
Flow cytometry
Immune cells were obtained from the peritoneal cavity and spleen. The spleen tissue was ground using a Tenbroeck Tissue Grinder (Cat: 357424, WHEATON®, DWK Life Sciences, Wertheim, Germany) to obtain single cells. Red blood cells were removed using RBC lysis buffer (155 mM NH4Cl, 12 mM KHCO3, and 0.1 mM EDTA). Peritoneal cavity cells were harvested using a syringe by injecting 5 ml phosphate-buffered saline (PBS) into the cavity and shaking for 1 min. The process was repeated twice. Ghost DyeTM Violet 510 viability dye (1:1000 dilution, Cat: 13-0870, Tonbo Biosciences, CA, USA) was used to differentiate viable from dead cells. For surface staining, the following antibodies were used at a 1:300 dilution: anti-B220-PerCP (Cat: 103234, BioLegend, CA, USA), anti-CD19-Bv421 (Cat: 562701, BD Biosciences, NJ, USA), anti-CD21/35-PEcy7 (Cat: 25-0211-82, eBioscience, CA, USA), anti-CD23-PE (Cat: 101-607, BioLegend), anti-CD23-FITC (Cat: 101605, BioLegend), anti-CD93-FITC (Cat: 136507, BioLegend), anti-CD93-APC (Cat: 17-5892-81, BioLegend), anti-IgM-APC-eFluor® 780 (Cat: 47-5790-80, eBioscience), anti-NOTCH2-APC (Cat: 130713, BioLegend), anti-NOTCH1-PE (Cat: 130607, BioLegend), anti-CD138-BV421 (Cat: 142507, BioLegend), anti-CD5-PE (Cat: 553023 BD Biosciences), anti-CD43-FITC (Cat: 143204, BioLegend), anti-CD1d-PEdazzle594 (Cat: 123519 BioLegend), anti-GL7-FITC (Cat: 144611, BioLegend), anti-BAFFR-PE (Cat: 134103, BioLegend), and anti-CD24-AlexaFluor 700 (Cat: 101836, BioLegend). Monoclonal antibody anti-CD95 (FAS)-biotin (Cat: 554256, BD Biosciences) was used with Streptavidin-APC (Cat: 554067, BD Biosciences). Surface staining was performed on ice by incubating cells with monoclonal antibody cocktails for 30 min. For intracellular staining, cells were fixed and permeabilized using the FOXP3/Transcription staining kit (Cat: 00-5523-00, eBioscience) as per the manufacturer’s instructions. Permeabilized cells were stained with anti-Blimp1-PE (1:100 dilution; Cat: 150005, BioLegend) and anti-Ki-67-APC (1:100 dilution; Cat: 17-5698-82, eBioscience). All stained cells were analyzed using an LSRFortessa (BD Biosciences) or a Cytoflex (Beckman Coulter, CA, USA) flow cytometer63. The flow cytometry gating strategies used in this study are presented in Supplementary Fig. S8.
In vitro FOB cell differentiation assay
T2B cells (CD19+CD93+B220+CD21midCD23+IgM+) from Cd19-Cre and Cicf/f;Cd19-Cre mice were sorted using a MoFlo Astrios cell sorter (Beckman Coulter). The sorted cells were cultured in Rosewell Park Memorial Institute 1640 (RPMI 1640; Cat: LM011-60, Welgene Inc., Gyeongsangbuk-do, Korea) media supplemented with 10% fetal bovine serum (FBS; Cat: S001-01, Welgene Inc.) and 10 μg/ml anti-IgM F(ab)‘2 (Cat: 16-5092-85, eBioscience) for 72 h. The cells were harvested and stained with a monoclonal antibody cocktail (anti-CD24 and anti-CD21). FOB cells were analyzed as CD24+CD21+ cells.
In vitro MZB cell differentiation assay
T1B cells (CD19+CD93+B220+CD21loCD23-IgM+) from Cd19-Cre, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice were sorted using a MoFlo Astrios cell sorter. The sorted cells were co-cultured with either control or DLL1-expressing OP9 cells in RPMI 1640 media supplemented with 10% FBS and 20 ng/ml of BAFF cytokine (Cat: 8876-BF-010, R&D Systems, MA, USA). After 72 h, the cells were harvested and stained with a monoclonal antibody cocktail (anti-CD1d and anti-CD21). MZB cells were analyzed as CD1dhiCD21+ cells.
BCR signaling analysis
To measure IgM stimulated BCR signaling, splenocytes were stimulated with 10 μg/ml anti-IgM F(ab)‘2 for 5 min in a 37 °C heat block and then washed with cold PBS to stop the activation of BCR signaling. The activated cells were then fixed with Cytofix/Cytoperm™ (Cat: 554655, BD Biosciences) for 30 min and washed with cold PBS. Cells were suspended in ice-cold methanol for cytoplasm permeabilization. After washing with PBS, cells were stained with either anti-p-BTK-PE (Cat: 646903, BioLegend), anti-pBLNK PE (Cat: 558442, BD Biosciences), anti-pERK-PE (Cat: 12-9109-41, Invitrogen, CA, USA), or anti-pAKT-PE (Cat: 560378, BD Biosciences) on ice for 1 h. To measure tonic BCR signaling, cells were immediately permeabilized with ice-cold methanol without anti-IgM treatment. All stained cells were analyzed using an LSRFortessa or a Cytoflex flow cytometer.
In vitro apoptosis assay
The apoptosis assay was performed as previously described43,64. FOB (CD19+CD93-CD21intCD23+) and MZB (CD19+CD93-CD21hiCD23lo) cells from Cd19-Cre, Atxn1lf/f;Cd19-Cre, and Cicf/f;Cd19-Cre mice were sorted using a MoFlo Astrios cell sorter. Sorted cells were cultured in RPMI 1640 media supplemented with 10% FBS, 1% β-mercaptoethanol (Cat: 21985-023, Gibco, Thermo Fisher Scientific, MA, USA), and 1% penicillin-streptomycin (Cat: 15140122, Gibco). After 72 h, the cells were harvested and stained with an Annexin V-PI staining kit (Cat: 640932, BioLegend) as per the manufacturer’s instructions. Stained cells were analyzed using an LSRFortessa or a Cytoflex flow cytometer.
Luciferase assay
Mouse Notch1 and Notch2 promoter regions (−1000 to −1) were amplified via PCR using Pfu-X DNA polymerase (Cat: SPX16-R250, SolGent, Daejeon, South Korea) and cloned into the pGL3-Basic firefly luciferase vector (Cat: E1751, Promega, WI, USA) using KpnI and XhoI restriction enzymes. HEK293T and HeLa cells were seeded into 24-well culture plates and co-transfected with 500 ng of the cloned firefly luciferase vector (pGL3-Notch1 pro or pGL3-Notch2 pro), 50 ng of a Renilla luciferase vector, and 400 ng of the expression vector (MIGR1, MIGR1-ETV4, or MIGR1-ETV5) using FUGENE®HD transfection reagent (Cat: E2311, Promega) as per the manufacturer’s instructions. The cells were lysed 48 h after transfection, and luciferase activity was measured using the Dual-Luciferase Reporter Assay System (Cat: E1960, Promega) as per the manufacturer’s instructions.
Site-directed mutagenesis
Site-directed mutagenesis was performed using a Quick-Change II XL Site-Directed Mutagenesis kit (Cat: 200521, Agilent Technologies, CA, USA) as per the manufacturer’s instructions. Primers used for the mutagenesis of three ETS binding motifs in the Notch1 promoter were as follows: Mut-1 sense, 5′-TTAACTGCAAAGGGATAAGTTAGGCATCTTGGACGCCCAG-3′ and Mut-1 antisense, 5′-CTGGGCGTCCAAGATGCCTAACTTATCCCTTTGCAGTTAA-3′; Mut-2 sense, 5′-TTTAAGGTCAAGCGTTCCGTTAGTCATGGCTAAGAGTGCC-3′ and Mut-2 antisense, 5′-GGCACTCTTAGCCATGACTAACGGAACGCTTGACCTTAAA-3′; Mut-3 sense, 5′-GTAAGGGTTCGCAGTTACCAGGGGCGGAGC-3′ and Mut-3 antisense, 5′-GCTCCGCCCCTGGTAACTGCGAACCCTTAC-3′. Primers used for the mutagenesis of two ETS binding motifs in the Notch2 promoter were as follows: Mut-1 sense, 5′-CGCGCCCAGGGGTTAACCGCAGAAAGAAGC-3′ and Mut-1 antisense, 5′-GCTTCTTTCTGCGGTTAACCCCTGGGCGCG-3′; Mut-2 sense, 5′-GCCCGCGTGTCCTAACGCCTCGGCCTCC-3′ and Mut-2 antisense, 5′-GGAGGCCGAGGCGTTAGGACACGCGGGC-3′. Desired mutations were validated using Sanger sequencing.
RNA sequencing and data analysis
MZB cells were sorted from Cd19-Cre and Cicf/f;Cd19-Cre mice using a MoFlo Astrios cell sorter. Total RNA was purified using RiboEx reagent (Cat: 301-002, GeneAll, Seoul, Korea) as per the manufacturer’s instructions. RNA purity was determined by assaying 1 μl total RNA on a NanoDrop8000 spectrophotometer (Thermo Fisher Scientific). Total RNA integrity was evaluated using an Agilent Technologies 2100 Bioanalyzer. An mRNA sequencing library was constructed using the Truseq Stranded mRNA/Total Library Prep Kit (Illumina Inc., CA, USA) as per the manufacturer’s instructions. Tophat (v2.0.13) software was used to map reads for each sample to the mm10 RefSeq reference genome. Aligned results were then added to Cuffdiff (v2.2.0) to identify DEGs. DEGs with log2 fold-change >0.5 and P < 0.01 were selected for GO and KEGG pathway analyses using the DAVID website (https://david.ncifcrf.gov/). The GO analysis was performed in terms of biological processes, cell components, and molecular function. GSEA was performed using GSEA software (version 4.1.0, https://gsea-msigdb.org/gsea/index.jsp). HALLMARK_NOTCH_SIGNALING (MM3870) was used as the gene set database for GSEA.
RT-qPCR
MZB cells were sorted from Cd19-Cre, Cic f/f;Cd19-Cre, and Atxn1l f/f;Cd19-Cre mice using a MoFlo Astrios cell sorter. Total RNA was purified using the RiboEx reagent and reverse-transcribed into cDNA using a GoScript Reverse Transcription System (Cat: A5004, Promega) as per the manufacturer’s instructions. The SYBR Green real-time PCR master mix (Cat: TOQPK-201, Toyobo, Osaka, Japan) was used for qPCR. Gene expression levels were normalized to 18S rRNA levels. Primer sequences are shown in Supplementary Table 1.
Immunization with TI and TD antigens
Cd19-Cre, Cicf/f;Cd19-Cre, and Atxn1lf/f;Cd19-Cre mice aged 10–12 weeks were injected intravenously with 50 μg TNP-AECM-FICOLL (Cat: F-1300-10, LGC Biosearch Technologies, Hoddesdon, UK) or intraperitoneally with 50 μg OVA (Cat: A2512, Sigma–Aldrich, MO, USA) immersed in alum (Cat: vac-alu-250, InvivoGen, CA, USA). After 7 days, the mice were sacrificed to obtain the serum and spleen tissues. Serum samples were used to measure NP-specific or OVA-specific IgM and IgG levels. Spleen tissues were used to analyze the splenic plasma (CD19+CD138+Blimp1+) and GCB (CD19+GL-7+CD95+) cells.
LPS-induced sepsis
Female Cd19-Cre, Cicf/f;Cd19-Cre, Atxn1lf/f;Cd19-Cre, and Etv4−/−;Cicf/f;Cd19-Cre mice were injected intravenously with 500 μg LPS from Escherichia coli O55:B5 (Cat: L2880-25MG, Sigma–Aldrich). LPS-treated mice were monitored every 24 h for mortality. Those that experienced severe convulsions, loss of motor skills, or a 20% decrease in body weight were euthanized. To obtain serum samples and lung tissues, the mice were sacrificed 12 h after LPS treatment. Mouse serum samples were used to measure IL-6 and IL-10 levels. Lung tissues were utilized to prepare paraffin blocks to identify immune cell infiltration in the organ.
Hematoxylin and eosin staining
Staining was performed as previously described40,65. Lung samples were fixed in a 10% formalin solution (Cat: HT501320-9.5L, Sigma–Aldrich) and agitated overnight. The samples were subjected to sequential dehydration in 70%, 80%, 90%, and 100% ethanol over the following 4 days. The lung samples were then immersed in xylene for 1 h; this process was performed three times. The xylene-soaked tissue was then placed in a paraffin-containing glass vessel and placed in a 60 °C oven overnight to allow paraffin infiltration into the tissue. The next day, paraffin blocks were constructed using an embedding machine (Shandon Histocentre 3, Thermo Fischer Scientific). Tissue sections were prepared using a Semi-Automated Rotary Microtome (Cat: RM2245, Leica Biosystems, Wetzlar, Germany). Sectioned tissues were mounted on non-adherent slides and air-dried on a 50 °C heat plate overnight. Paraffin on the slides was removed via triple immersion in xylene at 7-min intervals. Dehydrated lung tissue sections were rehydrated via sequential immersion in bottles containing 100%, 90%, 80%, and 70% ethanol at 3-min intervals. The sections were then incubated in hematoxylin (Cat: GT0111, Glentham, Life Sciences Ltd., Wiltshire, UK) for 1 min for nuclear staining. The slides were then rinsed with tertiary distilled water and incubated in a 0.1% ammonium hydroxide solution (Cat: 221228, Sigma–Aldrich) for 1 min to complete nuclear staining. After the bluing reaction, the slides were washed with tertiary distilled water and incubated in eosin (Cat: EM000G-5, Cancer Diagnostics, NC, USA) for 1 min to stain the cytoplasm. After cell staining, Canadian balsam solution (Cat: C1795, Sigma–Aldrich) was applied directly onto the lung tissue, and a cover glass was placed over it. The 100× images of the lung sections were obtained using an Olympus BX41TF light microscope (Olympus Corporation, Tokyo, Japan). The immune cell-infiltrated area was quantified using ImageJ software (National Institutes of Health).
Enzyme-linked immunosorbent assay
IL-6 and IL-10 levels in serum samples were quantified using the IL-6 and IL-10 Mouse Uncoated enzyme-linked immunosorbent assay (ELISA) Kits (Cat: 88-7064-22 and 88-7105-22, Invitrogen), respectively. Antibodies captured from the kits were coated onto 96-well half-area clear flat-bottom plates (Cat: 3690, Corning Inc., NY, USA) overnight at 4 °C. The plates were then rinsed with ELISA wash buffer (0.05% Tween-20 in PBS), followed by a 1-h blocking step at room temperature. The reference cytokines and diluted serum samples were incubated in the plates for 2 h at room temperature. The plates were then subjected to several washes and incubated with detection antibodies for 1 h at room temperature. Following additional washing, streptavidin-HRP was added and incubated for 30 min at room temperature. After washing the plates, 50 μl TMB substrate was added, and the plates were incubated for 15 min in a light-blocked environment. Finally, 50 μl of 1 M H2SO4 was added to stop the reaction. Optical density was measured at 450 nm using a spectrophotometer. For the NP-specific Ig ELISA, NP-BSA (Cat: T-5050-10, Biosearch Technologies) was used as the coating antigen. For OVA-specific Ig ELISA, OVA was the coating antigen used. Goat Anti-Mouse IgM, Human ads-HRP (Cat: 1020-05-SBA, Southern Biotech, AL, USA) and Goat Anti-Mouse IgG, Human ads-HRP (Cat: 1031-05-SBA, Southern Biotech) were used as secondary antibodies to capture anti-IgM and anti-IgG in serum, respectively. The overall process was consistent with that of cytokine ELISAs.
Retrovirus preparation
Plat E cells were seeded in 100 mm dishes at a density of ~2 × 10⁶ cells. After 24 h, when the cells reached 70–80% confluency, they were co-transfected with either 4.5 μg of MIGR1 (negative control) or MIGR1-ETV4 expression vector, along with 1.5 μg of the pCL-Eco vector (retrovirus packaging vector), using FUGENE transfection reagent (Cat: E2312, Promega) according to the manufacturer’s instructions. The transfected cells were incubated for 48 h at 37 °C in a humidified atmosphere containing 5% CO2. Afterward, the cell culture supernatant containing the retrovirus particles was collected. The supernatant was mixed with Retro-X concentrator (Cat: 631456, Takara) at a 3:1 ratio and incubated overnight at 4 °C with gentle swirling. The viral mixture was then centrifuged at 1500 × g for 45 min at 4 °C, and the resulting virus pellet was resuspended in RPMI medium.
Retroviral transduction
B-2 cells were isolated from the spleens of WT mice using negative selection. The isolated B-2 cells were then stimulated with anti-IgM (1 μg/ml) and anti-CD40 (1 μg/ml; Cat: 553788, BD bioscience) antibodies for 18 h. For retroviral transduction, the stimulated B-2 cells were mixed with polybrene at a final concentration of 8 μg/ml and the previously prepared retrovirus stock. The cell-virus mixture was then transferred to a 24-well plate, and spin infection was performed by centrifuging the plate at 750 × g for 2 h at room temperature. After spin infection, the medium containing the virus mixture was carefully replaced with fresh RPMI medium to maintain cell viability. The infected cells were then cultured for 48 h at 37 °C in a 5% CO2 incubator. Following the 48-h culture period, the cells were harvested and prepared for flow cytometry.
ChIP-qPCR
B-2 cells were isolated from the spleens of Cicf/f;Cd19-Cre mice using negative selection. 5 × 106 B-2 cells were cross-linked with 1% paraformaldehyde for 20 min under constant agitation. To terminate the cross-linking reaction, 1 M glycine was added, followed by a 5 min incubation. The cross-linked chromatin was rinsed at least twice with cold PBS, then centrifuged at 4200 rpm for 5 min. The pellet was resuspended in Buffer 1 (50 mM HEPES-KOH, pH 7.5; 140 mM NaCl; 1 mM EDTA, pH 8.0; 10% glycerol; and 0.5% NP-40) and rotated at 4 °C for 10 min to lyse the nuclei. After a 5 min centrifugation at 4200 rpm, the pellet was resuspended in MNase digestion buffer (100 mM Tris-HCl, pH 8.0; 2 mM CaCl₂; 0.4% Triton X-100; and MNase [0.001 U/mL, Sigma]) and incubated at 37 °C for 12 min. The reaction was terminated by adding 10 mL of 0.5 M EDTA, followed by centrifugation at 4200 rpm for 5 min. The lysate was then resuspended in Buffer 2 (10 mM Tris-HCl, pH 8.0; 300 mM NaCl; 0.1% sodium deoxycholate; 1% Triton X-100; 1 mM EDTA, pH 8.0; and 0.5 mM EGTA, pH 8.0) and sonicated to shear DNA fragments. Sonication process is comprised with 15 cycles of 30 s eruption and 30 s resting. The supernatant was collected after micro centrifugation at 9400 rpm for 10 min at 4 °C and 10% were stored at −80 °C for input sample. For immunoprecipitation, chromatin was precleared with protein G agarose (Cat: 16-266, Merck, Darmstadt, Germany) in Buffer 2 for 90 min. Post-preclearing, 2 µg of either normal rabbit IgG (Cat: #2729, Cell Signaling Technology, MA, USA) or anti-ETV4 antibody (Cat. 10684-1-AP, Proteintech, IL, USA) was added to the samples, followed by overnight incubation at 4 °C with gentle agitation. The chromatin-antibody complexes were incubated with protein G agarose for an additional 3.5 h at 4 °C. Subsequently, the samples were washed sequentially with low salt buffer (4 mM EDTA, 2% Triton X-100, 40 mM Tris-HCl, pH 8.0, 300 mM NaCl, 0.2% SDS), high salt buffer (4 mM EDTA, 2% Triton X-100, 40 mM Tris-HCl, pH 8.0, 1 M NaCl, 0.2% SDS), LiCl buffer (0.5 M LiCl, 20 mM Tris-HCl, pH 8.0, 2 mM EDTA, 2% NP-40, 2% sodium deoxycholate), and TE buffer (20 mM Tris-HCl, pH 8.0, 2 mM EDTA). Bound chromatin was eluted with elution buffer (0.1 M NaHCO₃ and 0.5% SDS) and reverse-crosslinked with 200 mM NaCl at 65 °C overnight. RNA and proteins were subsequently digested with RNase A and protease K, respectively, and DNA was purified using the Expin CleanUp SV kit (Cat. 113-150, GeneAll). The qPCR primer sequences for the promoter regions of Notch1 and Notch2 were as follows: For Notch1, the forward primer sequence was 5′-CGAACTCCCTTCTACAGAGGC-3′ and the reverse primer sequence was 5′-CTGGGAGCTGTTTGGTCTCG-3′. For Notch2, the forward primer sequence was 5′-GTACGGGGTGCTGCTCACTC-3′ and the reverse primer sequence was 5′-AGGGGTTTCCCGCAGAAAGA-3′.
Western blotting
Using WT C57BL/6 mice, FOB and MZB cells were obtained from the spleen, and B-1a cells were obtained from the peritoneal cavity. Sorted cells were lysed in RIPA buffer (50 mM Tris-HCl [pH 7.4], 150 mM NaCl, 1 mM PMSF, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 2X Complete Protease Inhibitor Cocktail [Cat: 5892953001, Roche, Basel, Switzerland], and 10X PhosSTOP™ [Cat: 4906837001, Roche]). Protein samples were separated using SDS-PAGE with 8% acrylamide gel as the separating gel. For CIC detection, SDS solution was added during the membrane transfer step. The following primary antibodies were used for protein detection: anti-CIC (1:2,000)66, anti-ATXN1L (1:2,000)66, anti-ATXN1 (1:2000; Cat: 2177, Cell Signaling Technology), and anti-GAPDH (1:2,000; Cat: sc-32233, Santa Cruz Biotechnology, TX, USA). Proteins were visualized using Clarity Western ECL Substrate (Cat: BR170-5061, Bio-Rad, CA, USA). Western blots were imaged using the ImageQuant LAS 500 system (GE Healthcare Life Sciences, IL, USA).
siRNA electroporation
siRNA electroporation of T1B cells was performed using the 4D-Nucleofector® X Unit (Cat: AAF-1003X, Lonza, Basel, Switzerland) with the P4 Primary Cell 4D-Nucleofector X Kit S (Cat: V4XP-4032, Lonza). T1B cells, isolated from Cd19-Cre and Atxn1lf/f;Cd19-Cre mice, were resuspended in 20 μl of nucleofection mixture (comprising 14.9 μl nucleofector solution, 3.6 μl supplement solution, and 1.5 μl siRNA at 20 pmol/μl). The cell mixture (20 μl) was then transferred to a cuvette provided in the kit, and electroporation was conducted using the DI-100 program (optimized for mouse B cells). Subsequently, the T1B cells were transferred to a plate pre-seeded with OP9-DLL1 for in vitro MZB cell differentiation. After 72 h, the cells were harvested and analyzed using flow cytometry. The following siRNA sequences were used: siATXN1-sense: 5’-CUGUAUCCAGAUUACUGUATT-3’, siATXN1-antisense: 5’-UACAGUAAUCUGGAUACAGTT-3’, siNC-sense: 5’-CCUACGCCACCAAUUUCGUTT-3’, siNC-antisense: 5’-ACGAAAUUGGUGGCGUAGGTT-3’.
Statistical analysis
All experiments were performed at least two times independently. Datasets were analyzed using the two-tailed Student’s t-test, one-way ANOVA with Tukey’s multiple comparisons test, and Log-rank (Mantel-Cox) test using Prism 10.0 software (GraphPad). Kolmogorov–Smirnov test was used for the GSEA. A P < 0.05 was considered significant. In the bar graphs, bars and error bars indicate mean and S.D., respectively. In the box and whisker plots, the box indicates the median and 25th–75th percentiles, while whiskers represent the 2.5th–97.5th percentiles.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Supplementary information
Description of Additional Supplementary Files
Source data
Acknowledgements
We thank the members of Lee laboratory for their inputs and comments on the study, and Dr. Huda Y. Zoghbi for kindly providing Atxn1−/−, Atxn1l floxed, and Cic floxed mice. This study was supported by the National Research Foundation (NRF) of Korea grants funded by the Korean government (2021R1A6A1A10042944, 2022M3E5F2018020, RS-2023-00260454, and RS-2024-00336114), the Korea Basic Science Institute (National Research Facilities and Equipment Center) grant funded by the Ministry of Education (2021R1A6C101A390), and the BK21 FOUR grant funded by the Ministry of Education, Republic of Korea. (4120240315124). J.L. was supported by a Global PhD Fellowship (NRF-2018H1A2A1059794). Y.H. was supported by the Basic Science Research Program of the NRF of Korea funded by the Ministry of Education (RS-2023-00276340).
Author contributions
Conceptualization: J.S.P., H.H., and Y.L.; Methodology: J.S.P., H.B.K., T.K.K., and Y.L.; Investigation: J.S.P., M.K., H.H., J.L., Y.S., Y.H., and S.K.; Writing—original draft: J.S.P. and Y.L.; Writing—review & editing: T.K.K. and Y.L.; Visualization: J.S.P. and Y.L.; Supervision: Y.L.; Funding acquisition: Y.L.
Peer review
Peer review information
Nature Communications thanks Stephen Yip and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. A peer review file is available.
Data availability
The sequencing data generated in this study have been deposited in the GEO NCBI under the accession code GSE264654. All other data are available in the main text or supplementary materials. Source data are provided with this paper.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
The online version contains supplementary material available at 10.1038/s41467-024-54803-z.
References
- 1.Montecino-Rodriguez, E. & Dorshkind, K. B-1 B cell development in the fetus and adult. Immunity36, 13–21 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Berland, R. & Wortis, H. H. Origins and functions of B-1 cells with notes on the role of CD5. Annu. Rev. Immunol.20, 253–300 (2002). [DOI] [PubMed] [Google Scholar]
- 3.Nagasawa, T. Microenvironmental niches in the bone marrow required for B-cell development. Nat. Rev. Immunol.6, 107–116 (2006). [DOI] [PubMed] [Google Scholar]
- 4.Pillai, S. & Cariappa, A. The follicular versus marginal zone B lymphocyte cell fate decision. Nat. Rev. Immunol.9, 767–777 (2009). [DOI] [PubMed] [Google Scholar]
- 5.Cariappa, A. et al. The follicular versus marginal zone B lymphocyte cell fate decision is regulated by Aiolos, Btk, and CD21. Immunity14, 603–615 (2001). [DOI] [PubMed] [Google Scholar]
- 6.Wen, L. et al. Evidence of marginal-zone B cell-positive selection in spleen. Immunity23, 297–308 (2005). [DOI] [PubMed] [Google Scholar]
- 7.Rauch, M., Tussiwand, R., Bosco, N. & Rolink, A. G. Crucial role for BAFF-BAFF-R signaling in the survival and maintenance of mature B cells. PLoS ONE4, e5456 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Khan, W. N. B cell receptor and BAFF receptor signaling regulation of B cell homeostasis. J. Immunol.183, 3561–3567 (2009). [DOI] [PubMed] [Google Scholar]
- 9.Saito, T. et al. Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development. Immunity18, 675–685 (2003). [DOI] [PubMed] [Google Scholar]
- 10.Tanigaki, K. et al. Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells. Nat. Immunol.3, 443–450 (2002). [DOI] [PubMed] [Google Scholar]
- 11.Garis, M. & Garrett-Sinha, L. A. Notch signaling in B cell immune responses. Front. Immunol.11, 609324 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Moriyama, Y. et al. Delta-like 1 is essential for the maintenance of marginal zone B cells in normal mice but not in autoimmune mice. Int Immunol.20, 763–773 (2008). [DOI] [PubMed] [Google Scholar]
- 13.Bray, S. J. Notch signalling: a simple pathway becomes complex. Nat. Rev. Mol. Cell Biol.7, 678–689 (2006). [DOI] [PubMed] [Google Scholar]
- 14.Gomez Atria, D. et al. Stromal Notch ligands foster lymphopenia-driven functional plasticity and homeostatic proliferation of naive B cells. J. Clin. Investig.132, e158885 (2022). [DOI] [PMC free article] [PubMed]
- 15.Hwang, I. Y., Boularan, C., Harrison, K. & Kehrl, J. H. Galpha(i) signaling promotes marginal zone B cell development by enabling transitional B cell ADAM10 expression. Front. Immunol.9, 687 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cerutti, A., Cols, M. & Puga, I. Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes. Nat. Rev. Immunol.13, 118–132 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Martin, F., Oliver, A. M. & Kearney, J. F. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity14, 617–629 (2001). [DOI] [PubMed] [Google Scholar]
- 18.Klein, U. & Dalla-Favera, R. Germinal centres: role in B-cell physiology and malignancy. Nat. Rev. Immunol.8, 22–33 (2008). [DOI] [PubMed] [Google Scholar]
- 19.Liu, X., Zhao, Y. & Qi, H. T-independent antigen induces humoral memory through germinal centers. J. Exp. Med.219, e20210527 (2022). [DOI] [PMC free article] [PubMed]
- 20.Cinamon, G., Zachariah, M. A., Lam, O. M., Foss, F. W. Jr & Cyster, J. G. Follicular shuttling of marginal zone B cells facilitates antigen transport. Nat. Immunol.9, 54–62 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Porsch, F., Mallat, Z. & Binder, C. J. Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies. Cardiovasc Res117, 2544–2562 (2021). [DOI] [PubMed] [Google Scholar]
- 22.Honda, S. et al. Marginal zone B cells exacerbate endotoxic shock via interleukin-6 secretion induced by Fcalpha/muR-coupled TLR4 signalling. Nat. Commun.7, 11498 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sakaguchi, T. et al. TRPM5 negatively regulates calcium-dependent responses in lipopolysaccharide-stimulated B lymphocytes. Cell Rep.31, 107755 (2020). [DOI] [PubMed] [Google Scholar]
- 24.Liu, D. et al. IL-10-dependent crosstalk between murine marginal zone B cells, macrophages, and CD8alpha(+) dendritic cells promotes Listeria monocytogenes infection. Immunity51, 64–76.e67 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu, J. et al. Fcmicro receptor promotes the survival and activation of marginal zone B Cells and protects mice against bacterial sepsis. Front. Immunol.9, 160 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lee, Y. Regulation and function of capicua in mammals. Exp. Mol. Med.52, 531–537 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jimenez, G., Guichet, A., Ephrussi, A. & Casanova, J. Relief of gene repression by torso RTK signaling: role of capicua in Drosophila terminal and dorsoventral patterning. Genes Dev.14, 224–231 (2000). [PMC free article] [PubMed] [Google Scholar]
- 28.Fores, M. et al. A new mode of DNA binding distinguishes Capicua from other HMG-box factors and explains its mutation patterns in cancer. PLoS Genet.13, e1006622 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shin, D. H. & Hong, J. W. Capicua is involved in Dorsal-mediated repression of zerknullt expression in Drosophila embryo. BMB Rep.47, 518–523 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lee, Y. et al. ATXN1 protein family and CIC regulate extracellular matrix remodeling and lung alveolarization. Dev. Cell21, 746–757 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wong, D. et al. TRIM25 promotes Capicua degradation independently of ERK in the absence of ATXN1L. BMC Biol.18, 154 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Choi, N. et al. miR-93/miR-106b/miR-375-CIC-CRABP1: a novel regulatory axis in prostate cancer progression. Oncotarget6, 23533–23547 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kim, E. et al. Capicua suppresses hepatocellular carcinoma progression by controlling the ETV4-MMP1 axis. Hepatology67, 2287–2301 (2018). [DOI] [PubMed] [Google Scholar]
- 34.Kim, J. W., Ponce, R. K. & Okimoto, R. A. Capicua in human cancer. Trends Cancer7, 77–86 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lee, J. S. et al. Capicua suppresses colorectal cancer progression via repression of ETV4 expression. Cancer Cell Int.20, 42 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Okimoto, R. A. et al. Inactivation of Capicua drives cancer metastasis. Nat. Genet.49, 87–96 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yoe, J., Kim, D., Kim, S. & Lee, Y. Capicua restricts cancer stem cell-like properties in breast cancer cells. Oncogene39, 3489–3506 (2020). [DOI] [PubMed] [Google Scholar]
- 38.Kim, S. et al. Regulation of positive and negative selection and TCR signaling during thymic T cell development by capicua. Elife10, e71769 (2021). [DOI] [PMC free article] [PubMed]
- 39.Park, G. Y. et al. Deletion timing of Cic alleles during hematopoiesis determines the degree of peripheral CD4(+) T cell activation and proliferation. Immune Netw.20, e43 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Park, S. et al. Capicua deficiency induces autoimmunity and promotes follicular helper T cell differentiation via derepression of ETV5. Nat. Commun.8, 16037 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Park, S., Park, J., Kim, E. & Lee, Y. The capicua/ETS translocation variant 5 axis regulates liver-resident memory CD8(+) T-cell development and the pathogenesis of liver injury. Hepatology70, 358–371 (2019). [DOI] [PubMed] [Google Scholar]
- 42.Park, J. et al. ETV5 promotes lupus pathogenesis and follicular helper T cell differentiation by inducing osteopontin expression. Proc. Natl. Acad. Sci. USA121, e2322009121 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hong, H. et al. Postnatal regulation of B-1a cell development and survival by the CIC-PER2-BHLHE41 axis. Cell Rep.38, 110386 (2022). [DOI] [PubMed] [Google Scholar]
- 44.Hong, H. & Lee, Y. Generation of hematopoietic lineage cell-specific chimeric mice using retrovirus-transduced fetal liver cells. STAR Protoc.3, 101526 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kreslavsky, T. et al. Essential role for the transcription factor Bhlhe41 in regulating the development, self-renewal and BCR repertoire of B-1a cells. Nat. Immunol.18, 442–455 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Didonna, A. et al. Ataxin-1 regulates B cell function and the severity of autoimmune experimental encephalomyelitis. Proc. Natl. Acad. Sci. USA117, 23742–23750 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cariappa, A. & Pillai, S. Antigen-dependent B-cell development. Curr. Opin. Immunol.14, 241–249 (2002). [DOI] [PubMed] [Google Scholar]
- 48.Myers, D. R., Zikherman, J. & Roose, J. P. Tonic signals: why do lymphocytes bother? Trends Immunol.38, 844–857 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Garg, A. et al. FGF-induced Pea3 transcription factors program the genetic landscape for cell fate determination. PLoS Genet.14, e1007660 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Clementz, A. G., Rogowski, A., Pandya, K., Miele, L. & Osipo, C. NOTCH-1 and NOTCH-4 are novel gene targets of PEA3 in breast cancer: novel therapeutic implications. Breast Cancer Res.13, R63 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Boyle, P. & Despres, C. Dual-function transcription factors and their entourage: unique and unifying themes governing two pathogenesis-related genes. Plant Signal. Behav.5, 629–634 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.McKenzie, G. et al. Cellular Notch responsiveness is defined by phosphoinositide 3-kinase-dependent signals. BMC Cell Biol.7, 10 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Villegas, S. N. et al. PI3K/Akt cooperates with oncogenic Notch by inducing nitric oxide-dependent inflammation. Cell Rep.22, 2541–2549 (2018). [DOI] [PubMed] [Google Scholar]
- 54.Gutierrez, A. & Look, A. T. NOTCH and PI3K-AKT pathways intertwined. Cancer Cell12, 411–413 (2007). [DOI] [PubMed] [Google Scholar]
- 55.Palomero, T. et al. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat. Med.13, 1203–1210 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Meurette, O. et al. Notch activation induces Akt signaling via an autocrine loop to prevent apoptosis in breast epithelial cells. Cancer Res.69, 5015–5022 (2009). [DOI] [PubMed] [Google Scholar]
- 57.Tong, X. et al. Ataxin-1 and Brother of ataxin-1 are components of the Notch signalling pathway. EMBO Rep.12, 428–435 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lu, H. C. et al. Disruption of the ATXN1-CIC complex causes a spectrum of neurobehavioral phenotypes in mice and humans. Nat. Genet.49, 527–536 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol.33, 314–325 (2003). [DOI] [PubMed] [Google Scholar]
- 60.Rickert, R. C., Roes, J. & Rajewsky, K. B lymphocyte-specific, Cre-mediated mutagenesis in mice. Nucleic Acids Res.25, 1317–1318 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Matilla, A. et al. Mice lacking ataxin-1 display learning deficits and decreased hippocampal paired-pulse facilitation. J. Neurosci.18, 5508–5516 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bowman, A. B. et al. Duplication of Atxn1l suppresses SCA1 neuropathology by decreasing incorporation of polyglutamine-expanded ataxin-1 into native complexes. Nat. Genet.39, 373–379 (2007). [DOI] [PubMed] [Google Scholar]
- 63.Song, Y. & Lee, Y. Brief guide to flow cytometry. Mol. Cells47, 100129 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mikami, M. et al. RUNX1-Survivin Axis Is a Novel Therapeutic Target for Malignant Rhabdoid Tumors. Mol. Cells45, 886–895 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kim, J. A. et al. Temporal transcriptome analysis of SARS-CoV-2-infected lung and spleen in human ACE2-transgenic mice. Mol. Cells45, 896–910 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kim, E. et al. Deficiency of Capicua disrupts bile acid homeostasis. Sci. Rep.5, 8272 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Description of Additional Supplementary Files
Data Availability Statement
The sequencing data generated in this study have been deposited in the GEO NCBI under the accession code GSE264654. All other data are available in the main text or supplementary materials. Source data are provided with this paper.