Abstract Malignant B cells rely on B cell receptor (BCR) signals for their survival and growth. Besides the Immunoglobulin M (IgM) BCR, lymphoma cells can also express non–IgM (IgG) BCRs; however, the role of IgG BCRs in malignant B cell is not well understood. Here, we report poorer disease outcomes in diffuse large B cell lymphoma (DLBCL) expressing high IgM versus those expressing IgG1. Using isogenic lymphoma cells expressing distinct BCRs, we found that IgM expressing cells strongly outcompete their IgG1 counterparts. Mechanistically, IgG1 BCR is associated with a dysfunctional mitochondrial state and reduced cell survival. We show that mitochondrial dysfunction is triggered by accentuated calcium responses downstream of IgG1 BCR. Genetic reversal of IgG1 to IgM, pharmacological dampening of calcium signaling, or treatment with interleukin-21 can correct mitochondrial defects and rescue IgG1 survival. Our findings demonstrate that distinct BCR isotypes are inherently unique and can differentially affect B cell lymphoma pathogenesis. __________________________________________________________________ B cell receptor isotypes play a crucial role in B cell physiology and related pathologies. INTRODUCTION Mature B cell lymphomas are thought to arise from the malignant transformation of B cells undergoing activation in specialized microanatomical sites called the germinal centers (GCs) ([52]1–[53]3). B cells in the GC rapidly proliferate and mutationally diversify their immunoglobulin (Ig) genes through the activity of activation-induced cytidine deaminase (AID) ([54]1, [55]4). AID deaminates cytosines to uracils and, through mutagenic DNA repair pathways, promotes somatic hypermutation (SHM) events to generate high-affinity antibodies ([56]1, [57]4). In addition, AID activity directed to a high density of hotspots in the Ig constant regions, known as switch regions, are processed as DNA double-strand breaks (DNA DSBs) to mediate genetic switching from IgM (or its splice variant IgD) to IgG, IgA, or IgE isotypes ([58]1, [59]2, [60]4). AID activity is often misdirected to non-Ig genes, leading to unwanted genetic lesions that drive the malignant transformation of activated B cells ([61]2, [62]4). One of the features of mature B cell–derived malignancies is their reliance on B cell receptor (BCR) signaling for survival, proliferation, and growth ([63]5–[64]7). Similar to the essential functions of BCR signaling in B cell development, activation, and differentiation, malignant B cells hijack these pathways to propagate growth and survival signals ([65]5–[66]7). Notably, small molecule inhibitors targeting proximal components of BCR signaling, such as Bruton’s tyrosine kinase (Btk) and phosphoinositide 3-kinase (PI3K), have been approved for the treatment of B cell neoplasms ([67]8). Although most studies investigating the role of BCR signaling in malignant B cells have focused on membrane-bound IgM, in line with the GC origin of these neoplasms, expression of non-IgM BCR isotypes have also been reported ([68]9, [69]10). The non-IgM isotypes, such as IgG and IgA, differ greatly in their signaling potential in comparison to IgM ([70]5), but how they contribute to the pathogenesis of mature B cell neoplasms is poorly understood. Among non-IgM BCR isotypes, studies have primarily compared the signaling potential of the IgG subclass of BCRs to IgM ([71]11–[72]14). IgG BCRs can be divided into IgG1, IgG2, IgG3, and IgG4 subclasses in humans and IgG1, IgG2a, IgG2b, and IgG3 subclasses in mice, with IgG1 being the most dominant of the non-IgM isotypes in the serum. IgG BCRs harbor more flexible hinge regions, distinct transmembrane domains, and longer cytoplasmic tails in comparison to IgM ([73]15). Moreover, the observed differences in signaling between IgM and IgG1 BCRs have been mostly attributed to the longer cytoplasmic tail in membrane-bound IgG1 ([74]12, [75]14, [76]16, [77]17). Like IgG BCRs, the membrane-bound forms of IgE and IgA isotypes carry longer cytoplasmic tails ([78]18–[79]21). The shorter cytoplasmic tail of IgM makes these receptors more reliant on cytoplasmic signaling domains of Ig alpha/beta (Igα/β), whereas the longer cytoplasmic tail of IgG1 confers less dependence on Igα/β ([80]14). Moreover, compared to IgM BCR, IgG1 BCR mediates stronger membrane proximal signaling events soon after BCR engagement and forms larger signaling microclusters ([81]13). IgG1 BCRs also display a lower force threshold to trigger signaling than IgM BCRs ([82]16). These key differences in signaling between IgM and IgG1 BCRs are shown to influence fate decisions in differentiating B cells, with IgG1 favoring plasma cell differentiation and IgM preferred during generation of long-term memory cells ([83]22–[84]25). In the context of malignant B cells, both antigen-independent tonic BCR signaling as well as (self) antigen-dependent chronic BCR signaling have been proposed to drive pathogenesis ([85]6, [86]26, [87]27). Related to this, IgM and IgG1 BCRs have been reported to differ in the context of both active and tonic BCR signaling ([88]28) and therefore might be pertinent regardless of the mode of BCR signaling in B cell neoplasms. Mature B cell neoplasms are broadly classified into Hodgkin’s lymphoma (~9000 new cases/year) and the more common non–Hodgkin’s lymphoma (B-NHL ~76,000 new cases/year), most of which arise from GCs. Among B-NHL, diffuse large B cell lymphoma (DLBCL) is the most common adult lymphoid malignancy, accounting for ~40% of all B-NHL cases. Initial classification of DLBCL based on transcriptional signatures defined two main DLBCL subtypes, the activated B cell type (ABC) DLBCL and GC B cell type (GCB) DLBCL ([89]29, [90]30). The classification of patients into these subtypes has immense prognostic value as ABC-DLBCL is associated with a more aggressive disease course and poorer clinical outcomes, whereas GCB-DLBCL follows a less aggressive disease course with better patient prognosis ([91]29, [92]30). More recent studies have used deep sequencing to genetically classify DLBCL into subgroups, including BCL6 and NOTCH2 alterations (BN2/Cluster1; ABC-like), MYD88 and CD79b mutations (MCD/Cluster 5; ABC-like), EZH2 and BCL2 mutations (EZB/Cluster 3; GCB-like), SGK1 and TET2 mutations (ST2), and NOTCH1 mutations ([93]31–[94]33). Even with the most recent iteration of the LymphGen genetic classification, a large proportion of DLBCL cases (~37%) remains unclassified ([95]32, [96]33). Besides the genetic and mutational signatures, the GCB-DLBCL and ABC-DLBCL subtypes also differ in the BCR isotypes that they express, with ABC-DLBCL mainly expressing IgM and GCB-DLBCL having a higher predilection to express non-IgM isotypes, such as IgG and IgA ([97]7, [98]9). The expression of non-IgM BCR isotypes have also been frequently reported in other B cell malignancies, such as, mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL), and double-hit lymphomas ([99]34–[100]36). A previous study in a small cohort of patients with DLBCL highlighted that the expression of non-IgM isotypes, specifically IgG1 and IgA, are associated with better disease prognosis than those expressing IgM ([101]9). However, whether distinct BCR isotypes actively affect the differential pathogenesis of B cell lymphomas is an open question. We sought to investigate the role of distinct BCR isotypes in the pathogenesis of mature B cell lymphomas. We focused on the IgG1 subclass and found that, among human patients with DLBCL, IgG1 expression is associated with better disease outcomes compared to IgM. Using isogenic human lymphoma cells, a mouse model of GC-derived lymphoma, and normal activated B cells, we show that IgM receptors are favored over IgG1 across multiple contexts. Our studies further reveal that IgG1 expressing cells display reduced survival associated with mitochondrial dysfunction and metabolic perturbations, caused by enhanced calcium responses in IgG1 cells compared to IgM. Overall, our work highlights mechanisms by which distinct BCR isotypes can differentially influence B cell fates and the pathogenesis of B cell lymphoma. RESULTS IgM BCR is associated with more aggressive pathogenesis of B cell lymphoma compared to IgG1 BCR To investigate the role of IgM and non-IgM BCR isotypes in lymphoma pathogenesis, we focused on the IgG1 BCR isotype, which is the most common non-IgM/IgD isotype expressed on B cells. To study this in the context of human B cell lymphoma, we categorized 481 patients with DLBCL from The Cancer Genome Atlas (TCGA) into those with high IGHM (n = 111) or IGHG1 (n = 63) (mRNA) expression ([102]Fig. 1A). The patients with DLBCL with high IGHM expression showed significantly poorer disease prognosis as evidenced by a shorter time to progression-free survival (median survival: 4.59 years), in comparison to patients with high IGHG1 expression (median survival: 6.68 years) ([103]Fig. 1B). These differences in patient outcomes could be due to underlying distinctions in DLBCL subtypes within IgM and IgG1 groups. Therefore, we further dissected our dataset into distinct DLBCL genetic subtypes in both IgM high and IgG1 groups. As reported previously ([104]33), IgM high samples were enriched for N1, MCD, and BN2 genetic clusters ([105]Fig. 1C), whereas high IgG1 expression was mainly associated with the EZB and BN2 clusters ([106]Fig. 1C). A large fraction of the patient samples in both IgM and IgG1 high groups did not belong to any known genetic clusters and was designated as unclassified or “other” genetic subtype ([107]Fig. 1C). We compared the impact of IgM and IgG1 expression on disease prognosis within BN2, EZB, and other clusters, all of which were composed of substantial proportions of IgM and IgG1 high samples. Compared to the IgM high cohort, the samples with high IgG1 expression were associated with better progression-free survival and lower clinical stage of disease in BN2 and Other subtypes but not the EZB subtype ([108]Fig. 1, D to F, and fig. S1A). The EZB genetic subtype is known to be associated with IgG1 expression ([109]33); hence, the similar patient prognosis between IgM and IgG1 cohorts within the EZB subtype might be attributed to the genetic mutations coevolving with IgG1 BCRs in this subgroup. We also compared the impact of IgM and IgG1 expression on patient outcomes in a patient cohort predominantly expressing the IGVH3-23 gene family, which presumably reflects comparable antigen-driven selection (fig. S1B). The IgG1 expression within the VH3-23 cohort also appeared to have better progression-free survival when compared to patients with high IgM expression, although these differences were not statistically significant (fig. S1B). In aggregate, we identify a strong association of IgG1 expression with improved survival in DLBCL compared to IgM, and these findings are consistent with a previous report ([110]9). Moreover, our results further highlight the differential impact of IgM and IgG1 BCRs on DLBCL pathogenesis, particularly in a cohort of DLBCL patients which remain genetically unclassified. Fig. 1. IgG1 expression is associated with less aggressive lymphoma pathogenesis. [111]Fig. 1. [112]Open in a new tab (A) IGHM (left) and IGHG1 (right) expression as transcripts per million (TPM) in IgM high (n = 111) or IgG1 high (n = 63) samples. (B) Kaplan-Meir curves displaying progression-free survival (probability) in IgM and IgG1 high samples. (C) Distribution of distinct DLBCL subtypes in IgM and IgG1 high groups. (D to F) Kaplan-Meir curves for progression-free survival (probability) in IgM high and IgG1 high in BN2 (D), EZB (E), and Other (F) genetic subtypes. sgRNA, single guide RNA. (G) Engineering of switched isogenic human lymphoma cell lines using CRISPR-Cas9. (H) IgM and IgG1 expression in BJAB, OCI-Ly7. and HBL-1 cells before and after CRISPR-Cas9 editing. (I) Tumor growth of IgM or IgG1 cells (1 million per mouse) subcutaneously injected into NSG mice (n = 5 mice). (J) Tumor growth curves of IgM or IgG1 expressing BJAB subcutaneously injected into NSG mice. (K) Surface expression of Ig-kappa light chain (Igκ) in IgM BJAB cells (gray), IgM tumors (gray dashed), and IgG1 BJAB cells (red), tumors which grew from IgG1 BJAB cells (red dashed) and BJAB IgM cells stained with secondary Ab (no Ab control, black). (L) Cotransplantation of IgG1 and IgM lymphoma cells mixed at 1:1. Bottom; Representative plots of IgM and IgG1 at days 0 and 21 posttransplant. (M) Quantification of IgM and IgG1 cell frequency in tumors after 21 days (n = 4 for Mino IgG1, Mino IgM, and BJAB IgG1; n = 5 for BJAB IgM). (N and O) Bar graphs quantifying percent IgM and IgG1 BJAB (N) and Mino (O) cells after mixing (day 0) and upon coculture for 5 or 8 days. Statistical significance calculated by log-rank test [(B) and (D) to (F)], t test (I), and two-way ANOVA [(M) to (O)]. Error bars represent ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. [(G) and (L)] Created in BioRender. Shukla, V. (2025) [113]https://BioRender.com/1mfdxwl. See also fig. S1. To directly test the role of IgM and IgG1 BCR isotypes in B cell lymphoma pathogenesis, we used clustered regularly interspaced short palindromic repeats (CRISPR)–Cas9–based genetic engineering to induce isotype switching from IgM to IgG1 in three different cell lines, each derived from lymphoma patients of distinct subtypes: BJAB (ABC-DLBCL/Burkitt-like lymphoma), OCI-Ly7 (GCB-DLBCL), and HBL-1 (ABC-DLBCL) ([114]Fig. 1, G and H). We used lentiviral transduction to deliver three different guide RNAs targeting the 5′ and 3′ of the Sμ region and the 3′ of the Sγ1 in the IGH locus ([115]37) in BJAB, HBL-1, and OCI-Ly7 cells, which normally express IgM BCR. CRISPR-Cas9 targeting followed by single-cell plating yielded isogenic lymphoma cells that were switched to express IgG1 BCRs, without treatment with mitogens or exogenous cytokines ([116]Fig. 1H). We tested the tumorigenic potential of IgM or IgG1 expressing isogenic cells in xenograft models using the NOD-SCID (nonobese diabetic–severe combined immunodeficient) gamma chain–deficient (NSG) mice. The IgM expressing BJAB, OCI-Ly7, and HBL-1 formed aggressive tumors upon subcutaneous injections in mice and rapidly progressed to humane endpoints for euthanasia ([117]Fig. 1I and fig. S1C). However, to our surprise, the IgG1 expressing isogenic cells mostly failed to form tumors in vivo ([118]Fig. 1I and fig. S1C). In follow-up experiments with IgM and IgG1 expressing isogenic BJAB cells, at least two mice originally injected with IgG1 BJAB cells grew tumors ([119]Fig. 1J). Intriguingly, the tumors arising from the IgG1 BJAB cells in these two mice had no surface or intracellular expression of Igκ light chain and were BCR negative (Ig−) ([120]Fig. 1K). Retransplantation of both IgM and Ig− BJAB cells again led to the formation of tumors, which were IgM positive and Ig negative, respectively (fig. S1D). In addition to the DLBCL cell lines (BJAB, OCI-Ly7, and HBL-1), we also induced switching from IgM to IgG1 in Mino cells, an MCL-derived cell line believed to originate from memory-like B cells that have exited the GCs. The CRISPR-Cas9–based approach could not efficiently induce switching to IgG1 in Mino cells, presumably due to altered genomic architecture of the IGH locus in these cells. However, stimulation of Mino cells with anti-CD40, lipopolysaccharide (LPS), and interleukin-4 (IL-4) generated IgG1-switched cells (fig. S1E). Comparable to our results with other lymphoma lines, the IgG1 expressing Mino cells did not form tumors in vivo, whereas their IgM expressing counterparts again formed aggressive tumors (fig. S1F). Next, we performed studies in which IgM and IgG1 expressing lymphoma cells, BJAB, and Mino, were mixed and cotransplanted in NSG mice, with tumors analyzed as they became palpable ([121]Fig. 1, L and M, and fig. S1G). The IgM expressing BJAB and Mino cells outcompeted their IgG1 expressing counterparts and resulted in tumors that primarily expressed IgM and lacked IgG1 ([122]Fig. 1, L and M, and fig. S1G). We further performed similar cellular competition assays with 1:1 mixing of IgM and IgG1 expressing BJAB and Mino cells in culture. As seen in vivo, the IgM expressing BJAB and Mino cells rapidly outcompeted their IgG1 expressing counterparts within 8 days of coculture ([123]Fig. 1, N and O). To examine the effects of BCR signaling on cellular kinetics and proliferation, we performed 5-bromo-2′-deoxyuridine (BrdU) pulse labeling in IgM or IgG1 expressing BJAB and OCI-Ly7 cells. In addition, we also generated BCR negative [Ig(−)] BJAB and OCI-Ly7 cell lines to study the contribution of BCR signaling to cellular proliferation. Notably, the BJAB and OCI-Ly7 cells expressing IgM showed the highest cell numbers over 3 days in culture, followed by IgG1 and Ig(−) cells [IgM > IgG1 > Ig(−)] (fig. S1H). The frequency of cells undergoing S phase, measured by BrdU labeling, followed a subtle but similar trend in OCI-Ly7 cells; and, in BJAB cells, IgG1 expression was associated with the lowest amount of BrdU labeling (fig. S1I). These findings suggest that differences in cellular kinetics mediated by tonic BCR signaling are not solely due to changes in proliferation. However, in the context of induced BCR signaling with an anti-kappa light chain antibody, IgG1 expressing BJAB cells demonstrated a pronounced reduction in BrdU labeling compared to their IgM counterparts (fig. S1J). Together, from these results, we conclude that the IgM BCRs are strongly favored over IgG1 in human lymphoma cells. IgM expressing cells outcompete IgG1 expressing counterparts in a TET-deficient model of B cell lymphoma One possible caveat of using established lymphoma cell lines could be the presence of genetic alterations that are more complementary to and reliant on the IgM BCR. To address this, we used a mouse model in which B cell–specific deletion of Tet2 and Tet3 (CD19 Tet2/Tet3 DKO) leads to the development of a GC-derived lymphoma, which recapitulates several features of human disease ([124]38). Moreover, TET2 (and occasionally TET3) is recurrently mutated in DLBCL, and patients with TET2 mutations are classified into the ST2 DLBCL cluster, which frequently expresses IgG1 BCRs ([125]33). We isolated CD19 DKO B cells at early stages of lymphomagenesis (6-week-old mice) and cultured them on a CD40 ligand (CD40L)– and B cell activation factor (BAFF)–expressing fibroblast cell line (40LB cells) in the presence of IL-4 to induce isotype switching to IgG1 ([126]Fig. 2A) ([127]39). Four days of CD19 DKO B cell coculture with 40LB cells consistently yielded 30 to 40% of IgG1 expressing cells ([128]Fig. 2B). Upon depletion of 40LB cells, CD138+ plasmablasts and plasma cells, and a population of IgE-switched cells, we transferred CD45.2-expressing CD19 DKO B cells into sublethally irradiated CD45.1 expressing immunocompetent host mice. Four weeks posttransfer, the CD45.2+ CD19 DKO B cells expressing a Cre recombinase driven yellow fluorescent protein (YFP) reporter were readily detectable in blood. Beginning at the 4-week time point, the frequency of IgG1 expressing CD19 DKO (YFP+) B cells in blood steadily declined in comparison to IgM expressing B cells ([129]Fig. 2C). At 10 weeks posttransfer, the host mice were euthanized to examine the transferred CD19 DKO B cells in the spleen. Compared to the IgM expressing populations, the frequency and the numbers of IgG1 expressing CD19 DKO B cells were significantly reduced in the spleen ([130]Fig. 2, D and E). The remaining IgG1 expressing TET-deficient B cells displayed a higher turnover than their IgM expressing counterparts, as evidenced by higher frequency of proliferating (Ki67 high) as well as apoptotic (cleaved caspase-3+) cell populations ([131]Fig. 2, F and G). It is also worth noting that, after 10 weeks, a substantial proportion of transferred TET-deficient B cells in the spleen was IgM and IgG1 double-negative cells (fig. S2A). Among the IgM and IgG1 double-negative cells, a large fraction expressed IgG2b or IgD and a smaller fraction expressed IgG3 or IgA BCR isotypes (fig. S2, B and C). We also tested the effect of IgM and IgG1 BCR in the context of wild-type primary B cells stimulated and switched on 40LB cultures. For this, we used congenically marked (CD45.1) wild-type B cells, stimulated them on 40LB cultures in the presence of IL-4, labeled them with CellTrace Violet (CTV), and transferred them into CD45.2-expressing host mice (fig. S2D). Although the frequency and numbers of IgM and IgG1 expressing (CD45.1) B cells in spleen 2 weeks posttransfer were much lower in comparison to CD19 DKO B cells, the cell numbers, frequencies, survival (cleaved caspase-3) and proliferation (CTV dye dilution) of IgG1 expressing cells were again lower than their IgM expressing counterparts (fig. S2, E to I). Together, these studies show that IgG1 expressing normal and pre–malignant B cells are more apoptotic than the IgM expressing counterparts. This provides further evidence that IgM cells are selected over IgG1 cells in a mouse model of GC-derived B cell lymphoma driven by TET deficiency. Fig. 2. IgM expressing TET-deficient B cells outcompete their IgG1 expressing counterparts. [132]Fig. 2. [133]Open in a new tab (A) Schematic of the experimental design. Cd19 cre Tet2/Tet3-deficient DKO B cells (CD45.2+) were isolated from the spleen and cultured for 4 days using an iGB in vitro culture system. TET-deficient B cells (2 million) were transplanted into sublethally irradiated CD45.1+ recipient mice. (B) Representative flow cytometry plot showing the frequency of IgM and IgG1 before transplantation in TET-deficient YFP+, CD45.1− B cells 4 days after iGB culture. (C) Relative frequency of IgM and IgG1 expressing YFP+, CD45.2+ B cells in blood over time. IgM and IgG1 frequencies are normalized to their respective pretransplantation frequency (n = 9 from two independent experiments). (D) Representative flow cytometry plot of YFP+, CD45.1− splenic B cells at week 10 posttransplantation (left). Quantification of IgM and IgG1 frequency (right) (n = 9 from two independent experiments). (E) Quantification of the absolute number of IgM and IgG1 splenic B cells gated from YFP+, CD45.1− B cells. Cell numbers are normalized to IgM (n = 9 from two independent experiments). (F) Representative flow cytometry plots of proliferating IgM and IgG1 cells with Ki67 staining from YFP+, CD45.1− B cells (left). Quantification of Ki67+ from YFP+, CD45.1− splenic B cells (right) (n = 6 from two independent experiments). (G) Representative flow cytometry plots of apoptotic IgM and IgG1 cells measured by cleaved caspase-3 staining from YFP+, CD45.1− cells (left). Quantification of cleaved caspase-3 from YFP+, CD45.1− splenic B cells (right) (n = 6 from two independent experiments). Statistical significance is calculated by ordinary two-way ANOVA test with Šídák multiple comparisons test (C) and paired t test [(D) to (G)]. Error bars represent means ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Schematic in (A) created in BioRender. Shukla, V. (2025) [134]https://BioRender.com/1mfdxwl. See also fig. S2. IgG1 expressing B cells are more sensitive to BCR-induced cell death than IgM expressing cells Because IgM lymphoma cells show increased survival compared to their IgG1 counterparts, we asked whether similar differences are evident in IgM and IgG1 expressing cells during normal B cell activation. To induce B cell activation and class switching in vivo, we immunized wild-type mice with sheep red blood cells (SRBCs) to generate T-dependent polyclonal GC responses ([135]Fig. 3A). Immunization with SRBCs led to robust induction of GC responses with efficient isotype switching to IgG1 ([136]Fig. 3B). Consistent with our observations in TET-deficient lymphoma cells, the IgG1 expressing GC B cells were more apoptotic than IgM expressing GC B cells on day 14 postimmunization ([137]Fig. 3B, right). Fig. 3. IgG1 expressing B cells are more sensitive to BCR-induced apoptosis. [138]Fig. 3. [139]Open in a new tab (A) Schematic for SRBC immunization. (B) Representative plots for gating GC B cells (left). Quantification of cleaved caspase-3 from IgM and IgG1 GC B cells (right). Connecting lines show data points within each mouse. (C) Schematic of iGB cocultures. (D) Representative plots for IgM and IgG1 frequency from CD19+CD138−, 12 hours poststimulation (BCR) with anti-kappa antibody (1 μg/ml). (E) Quantification of IgG1 and IgM B cell frequency with or without BCR stimulation. IgM and IgG1 are normalized to respective frequencies under untreated conditions (n = 7, three independent experiments). (F) Quantification of IgM and IgG1 B cell numbers (n = 7, three independent experiments). (G) Representative flow plots (left) and quantification of cleaved caspase-3 (right). Cleaved caspase-3 is normalized to IgM cells in the untreated group (n = 17 from seven independent experiments). (H) Genome browser tracks of RNA-seq data in the IgH locus (Ighg1 and Ighm genes are highlighted). Black and red tracks are from IgM and IgG1 cells from untreated and BCR-stimulated groups. Tracks are averaged from three replicates. (I) Volcano plot displaying DEGs in IgG1- versus IgM expressing cells from untreated (left) and BCR-stimulated conditions (right). (J) Up-regulated (red) and down-regulated (gray) pathways identified from DEGs in IgG1 compared with IgM using Metascape. The x axis represents the z score. (K) Gene set enrichment analysis (GSEA) of apoptosis and abnormal mitochondrial morphology gene sets. The y axis denotes enrichment score. NES, normalized enrichment score; P nominal P value. Statistical significance is calculated by the paired t test (B) and ordinary two-way ANOVA with Šídák multiple comparisons test [(E) and (F)]. Error bars represent ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. Schematics in (A) and (C) created in BioRender. Shukla, V. (2025) [140]https://BioRender.com/1mfdxwl. See also fig. S3. To further study the effects of IgM and IgG1 BCRs on B cell survival in a more tractable system, we isolated naive B cells from wild-type mice and stimulated them on 40LB cultures with IL-4, mimicking in vivo GCs (induced GC B cell or iGB culture). Following 3 days of activation on iGB cultures, we depleted 40LB stromal, CD138+ plasmablasts and plasma cells, and IgE+ cells and further cultured these B cells with (BCR) or without (untreated) treatment with an anti-kappa light chain antibody to induce active BCR signaling ([141]Fig. 3C). Twelve hours post–BCR stimulation, the IgG1+ cells showed a significant decrease in both frequencies and total cell numbers compared with IgG1+ cells under the untreated condition ([142]Fig. 3, C to E). Although both IgM and IgG1 expressing non-plasmablast B cells (B220+CD19+CD138−) demonstrated a significant increase in apoptosis upon BCR engagement, this effect appeared to be stronger in IgG1 than IgM expressing cells ([143]Fig. 3G). The IgG1 expressing human lymphoma cells (OCI-Ly7 and BJAB) also responded similarly by exhibiting a heightened sensitivity to BCR engagement, leading to higher levels of apoptosis in comparison to IgM expressing lymphoma cells (fig. S3, A to E). In sum, these studies demonstrate enhanced sensitivity of IgG1 expressing normal B cells and lymphoma cells to BCR-induced apoptosis. BCR engagement in IgM and IgG1 expressing cells leads to prominent transcriptional changes To mechanistically interrogate the differential responses of IgM and IgG1 to BCR signaling, we performed transcriptional profiling (in triplicate) with or without BCR engagement. We activated B cells on iGB cultures for 3 days, then depleted 40LB, CD138+, and IgE+ cells, and further cultured IgM and IgG1 expressing B cells in the absence or presence of anti-kappa antibody (1 μg/ml) to stimulate BCR signaling. Following 12 hours of culture with or without BCR stimulation, RNA sequencing (RNA-seq) was performed on flow-sorted populations of IgM or IgG1-positive cells (B220+ CD19+ CD138− and IgE−). Reassuringly, cells sorted for IgM expression under both untreated and BCR-stimulated conditions showed high expression of the Ighm transcript, whereas cells sorted for IgG1 expression displayed high Ighg1 transcripts and minimal Ighm expression ([144]Fig. 3H). We then performed comparative expression analysis between different groups to identify differentially expressed genes (DEGs). The highest number of DEGs was observed between BCR-stimulated IgG1 cells compared with BCR-stimulated IgM cells [299 up-regulated and 374 down-regulated; log[2] fold change > ±1 and FDR (false discover rate) < 0.05] ([145]Fig. 3I). The down-regulated genes in stimulated IgG1 cells (compared to stimulated IgM) included non–receptor tyrosine kinase Syk, E3 ubiquitin ligase Cblb, costimulatory molecule IcosL, and components of Notch signaling ([146]Fig. 3I and fig. S3G). The up-regulated genes in stimulated IgG1 cells included plasma cell differentiation regulators: Prdm1, Xbp1, and Irf4, negative regulators of BCR signaling: Spry2, Tigit, and Pdcd1, and metabolic enzymes: Shmt1, Shmt2, Mthfd1, and Ppat ([147]Fig. 3I and fig. S3, H to K). IgG1 expressing B cells have previously been shown to have an increased propensity toward plasma cell differentiation ([148]22, [149]23), and our findings are consistent with those observations. Moreover, the expression of Aicda, the gene encoding AID, which mediates SHM and CSR in activated B cells, was comparable in BCR-stimulated IgM and IgG1 B cells (fig. S3L). Considerably fewer genes were altered between IgG1 and IgM expressing cells in the non–BCR-stimulated (untreated) groups (45 up-regulated and 145 down-regulated). We also assessed the effects of BCR stimulation within IgM or IgG1 cells in comparison to untreated conditions ([150]Fig. 3I). The IgM expressing cells showed no significant alterations in gene expression upon BCR stimulation for 12 hours, whereas BCR-stimulated IgG1 cells showed similar changes in gene expression to those seen in comparison with BCR-stimulated IgM cells (up-regulated: Prdm1, Irf4, and Xbp1; down-regulated: Notch2, Hes1, and Cblb) ([151]Fig. 3I). We next used pathway enrichment analysis to further study the DEGs identified in comparison between BCR-stimulated IgG1 and IgM cells. The down-regulated pathways in BCR-stimulated IgG1 cells were associated with B cell activation, differentiation, and B cell–mediated immunity ([152]Fig. 3J). On the other hand, the most highly enriched up-regulated pathways in IgG1 cells included one-carbon metabolism associated with nucleotide biosynthesis as well as glycine, glutamine, and folate metabolism ([153]Fig. 3J). These observations are intriguing, particularly in light of previous studies that linked high metabolic stress and mitochondrial dysfunction to increased dependency on one-carbon metabolism ([154]40, [155]41). Prompted by these data, we specifically assessed changes in gene signatures associated with mitochondrial morphology and function. We noted a significant enrichment of gene signatures associated with abnormal mitochondrial morphology in BCR-stimulated IgG1 cells ([156]Fig. 3K, top). In addition, we observed an enrichment of gene signatures linked to apoptosis in BCR-stimulated IgG1 cells, in line with the increased tendency of IgG1 cells to undergo apoptosis ([157]Fig. 3K, bottom). In sum, these studies identified altered differentiation and metabolic pathways as the predominant transcriptional changes associated with BCR engagement in IgG1 B cells. IgG1 expressing cells display lower mitochondrial mass and function than IgM expressing counterparts Our results thus far demonstrated that IgG1 expressing cells are outcompeted by IgM cells, with the transcriptomics data linking BCR stimulation of IgG1 to altered cellular metabolism. Therefore, we asked whether engagement of IgG1 and IgM BCRs could affect mitochondrial dynamics and function. To this end, we measured the mitochondrial mass using a cell-permeant fluorescent dye, MitoTracker Green, in IgM and IgG1 expressing GC B cells from SRBC-immunized mice. IgG1 expressing GC B cells exhibited significantly lower mitochondrial mass than IgM expressing GC B cells ([158]Fig. 4A). To confirm these findings with an analogous approach, we used a flow cytometry–based imaging method (ImageStream) to stain for the mitochondrial membrane protein, translocase of outer mitochondrial membrane 20 (Tomm20), in GC B cells ([159]Fig. 4B). The IgG1 GC B cells showed reduced Tomm20 staining, indicative of lower mitochondrial mass than IgM GC B cells ([160]Fig. 4B). Furthermore, IgG1 cells switched on iGB cultures displayed lower mitochondrial mass compared with IgM expressing cells, and this decrease in mitochondrial mass in IgG1 cells was magnified upon BCR engagement ([161]Fig. 4C). These changes were not an outcome of altered cell size as normalization by cell size in IgM or IgG1 cells did not affect these findings ([162]Fig. 4C, right). Similar to mouse B cells, the human OCI-Ly7 and BJAB lymphoma cells expressing IgG1 BCRs showed lower mitochondrial mass, particularly upon BCR stimulation in BJAB cells (fig. S4, A and B). Fig. 4. IgG1 expressing B cells display lower mitochondrial mass. [163]Fig. 4. [164]Open in a new tab (A) Representative plots of IgM and IgG1 GC B cells (Fas+, CD38low) (left) 14 days postimmunization with SRBCs. Histograms show MitoTracker Green staining in IgM and IgG1 cells (middle). Median fluorescence intensity of MitoTracker Green in IgM and IgG1 cells (right) (n = 8). (B) Representative ImageStream analysis of IgM and IgG1 GC B cells stained with DAPI (nucleus), Tomm20 (PE: mitochondria), IgG1 (FITC), and IgM (PE-Cy7). (C) Histograms showing MitoTracker Green in IgM and IgG1 cells from iGB cultures stimulated with anti-kappa (1 μg/ml) for 12 hours (left). Quantification of relative MitoTracker Green mean fluorescence intensity (MFI) (middle). Values are normalized to untreated IgM from respective experiments. Relative MitoTracker Green MFI also normalized to cell size (right) (n = 6 from two independent experiments). (D) Flow cytometry histograms showing MitoTracker Green staining (top) and quantification of MitoTracker Green MFI (bottom) in IgM and IgG1 splenic B cells from heterozygous IgH^γ1μ/+ mice (n = 3). (E) Representative confocal microscopy images of splenic B cells from heterozygous IgH^γ1μ/+ mice (left) and quantified as mean fluorescence intensity (right). (F) Experimental design with IgG1 cells isolated from expressing homozygous IgH^γ1μ/γ1μ mice and treated with TAT-Cre for 2 hours and stimulated with anti-kappa antibody. (G) Representative flow plots of IgM and IgG1 staining of IgH^γ1μ/γ1μ splenic B cells before TAT-Cre treatment and 24 and 48 hours after TAT-Cre treatment. (H) Histograms showing MitoTracker Green staining (left) and quantification of MitoTracker Green MFI (right) in IgM and IgG1 B cells from IgH^γ1μ/γ1μ mice 48 hours post-BCR anti-kappa stimulation (2 μg/ml) (n = 5). Statistical significance is calculated by the paired t test (A), ordinary two-way ANOVA with Šídák multiple comparisons test [(C), (D), and (H)], and unpaired t test (E). Error bars represent ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. See also fig. S4. Isotype switching is tightly linked to cell activation and proliferation, and we questioned whether the decrease in mitochondrial mass in IgG1 cells could be unique to their activation history and kinetics in comparison to IgM B cells. To account for this, we used a transgenic mouse line in which immunoglobulin heavy chain (IgH) locus is engineered to harbor an Ighg1 allele with Ighm constant region placed in the opposite orientation, flanked by loxP sites (IgH^γ1μ) ([165]14). This mouse line allows reverse switching from IgG1 to IgM upon Cre-mediated recombination ([166]14). In the IgH^γ1μ/γ1μ homozygous mice, all B cells express IgG1 BCRs following VDJ recombination in the bone marrow. In the IgH^γ1μ/+ heterozygous mice, a population of both IgG1 and IgM expressing B cells is readily identified without the need for immunization or exogenous stimulation of B cells. Analysis of IgM and IgG1 expressing B cells from IgH^γ1μ/+ mice again showed lower mitochondrial mass (defined by MitoTracker Green) in both IgG1 expressing naive and BCR-stimulated cells in comparison with IgM cells ([167]Fig. 4D). Confocal imaging of B cells from IgH^γ1μ/+ mice showed a clear reduction in mitochondrial mass (Tomm20 staining) in IgG1 compared to IgM expressing B cells ([168]Fig. 4E). We further tested whether the decreased mitochondrial mass is a direct consequence of IgG1 BCR signaling rather than the distinct developmental dynamics of IgG1 expressing B cells in this model. Toward this, we isolated B cells from IgH^γ1μ/γ1μ homozygous mice and induced a reverse switch from IgG1 to IgM upon treatment with TAT-Cre in vitro ([169]Fig. 4F). Two hours of TAT-Cre treatment and washout was sufficient to induce substantial switching to IgM by 48 hours ([170]Fig. 4G). Under these conditions, the cells were either left untreated or were stimulated with anti-kappa antibody to induce BCR signaling. Notably, a switch from IgG1 to IgM was sufficient to increase the mitochondrial mass and rescue the mitochondrial defects observed in IgG1 B cells ([171]Fig. 4H). To assess the functional consequences of reduced mitochondrial mass in IgG1 expressing cells, we performed a Seahorse mitochondrial stress test to measure mitochondrial function. Consistent with the reduced mitochondrial mass, the IgG1 expressing B cells isolated from IgH^γ1μ/+ mice displayed lower levels of basal respiration [oxygen consumption rate (OCR)] and reduced maximal respiratory capacity [following carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP) treatment] than IgM expressing cells ([172]Fig. 5A). The IgG1 expressing BJAB cells also showed lower basal respiration and respiratory capacity than IgM expressing isogenic cells ([173]Fig. 5B). It is worth pointing out that BCR engagement led to a substantial decrease in basal respiration measured in real time in both IgM and IgG1 BJAB cells, indicating a negative impact of active BCR signaling on mitochondrial respiration ([174]Fig. 5B). Fig. 5. IgG1 signaling impairs mitochondrial function in normal B and lymphoma cells. [175]Fig. 5. [176]Open in a new tab (A) Changes in OCR measured over time in IgM (black) and IgG1 (red) IgH^γ1μ/+ B cells stimulated with anti-kappa antibody, treated with oligomycin, phenylhydrazone (FCCP), and antimycin A and rotenone. (B) Changes in OCR measured over time in IgM and IgG1 BJAB lymphoma cells treated with anti-Igκ antibody, oligomycin, FCCP, antimycin A, and rotenone. (C) Representative flow cytometry plots of IgM and IgG1 splenic GC B cells (Fas+, CD38 low) stained with MitoTracker Green (x axis) and TMRM (y axis). Gated population indicates dysfunctional mitochondria (left), identified as lower TMRM and positive MitoTracker Green staining. Quantification of the frequency of dysfunctional mitochondria in IgM and IgG1 GC B cells (right) (n = 3). (D) Representative flow cytometry plots of IgM and IgG1 B cells from in vitro iGB cultures stained with MitoTracker Green (x axis) and TMRM (y axis) (left). Quantification of the frequency of dysfunctional mitochondria measured by identifying as lower TMRM and positive MitoTracker Green staining (right) (n = 5 from two independent experiments). (E) Frequency of cleaved caspase-3–positive cells in IgM and IgG1 B cells from in vitro iGB cultures (n = 5 from two independent experiments). (F and G) Representative flow cytometry plots of OCI-Ly7 (F) and BJAB (G) IgM and IgG1 lymphoma cells stained with MitoTracker Green and TMRM after BCR treatment for 24 hours (left). Quantification of the frequency of dysfunctional mitochondria (right) (n = 6 from two independent experiments (F) and n = 3 (G) (right). Statistical significance is calculated using a paired t test (C) and ordinary two-way ANOVA with Šídák multiple comparisons test [(D) to (G)]. Error bars represent means ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. Schematic in (F) created in BioRender. Shukla, V. (2025) [177]https://BioRender.com/1mfdxwl. See also fig. S4. Functionally intact mitochondria maintain electrical potential in their inner membranes for efficient energy production through oxidative phosphorylation. Therefore, as another readout for mitochondrial health and function, we stained cells with tetramethylrhodamine (TMRM), a fluorescent dye that labels mitochondria in a membrane potential–dependent manner. IgG1 cells in GCs displayed a higher extent of mitochondrial depolarization than the IgM GC B cells, as measured by lower TMRM staining ([178]Fig. 5C). This depolarized mitochondrial state was noticeable even in IgG1 cells compared with IgM cells cultured under iGB conditions and was strongly associated with BCR stimulation ([179]Fig. 5D). BCR stimulation following iGB cultures led to an ~5-fold increase in IgG1 cells with depolarized mitochondria compared to those with IgM ([180]Fig. 5D). For reference, the difference in IgG1 GC cells with depolarized mitochondria in comparison to IgM GC B cells was more subtle (~1.6-fold) ([181]Fig. 5, C and D). This led us to hypothesize that IgG1 expressing cells in GCs could be somewhat protected from accumulating dysfunctional mitochondria. To test for signals that might contribute to this, we assessed the role of IL-21, a prominent GC-associated cytokine, which is shown to amplify BCR or CD40-dependent activation of AKT (phospho-AKT) and the mammalian target of rapamycin (mTOR) target, phosphorylated S6 ([182]42). BCR stimulation combined with IL-21 rescued the depolarized mitochondrial state in IgG1 cells ([183]Fig. 5D). The protective effect of IL-21 on mitochondrial dysfunction in IgG1 cells was further associated with a rescue of cell survival ([184]Fig. 5E). These results indicate that certain GC associated cytokines could at least partially protect against IgG1-induced mitochondrial dysfunction. Similarly, IgG1 lymphoma cells also showed enhanced mitochondrial depolarization in IgG1 cells in comparison to IgM cells upon BCR engagement ([185]Fig. 5, F and G). Although OCI-Ly7 cells were more sensitive to BCR-induced mitochondrial depolarization than BJAB cells, the IgG1 expressing cells consistently showed more mitochondrial depolarization upon BCR engagement than the IgM counterparts ([186]Fig. 5, F and G). These studies demonstrate that IgG1 BCR signaling profoundly affects mitochondrial dynamics, leading to a depolarized mitochondrial state, which is associated with reduced mitochondrial mass and function in comparison to IgM BCR. IgG1 BCR-induced calcium flux drives mitochondrial dysfunction and apoptosis A recent study showed that chronic BCR stimulation drives mitochondrial dysfunction in a calcium-dependent manner ([187]43). Hence, to test whether altered calcium dynamics could contribute to mitochondrial depolarization and reduced survival in IgG1 cells, we measured intracellular calcium flux downstream of BCR engagement. In primary B cells ([188]Fig. 6A) and all four isogenic lymphoma cell lines ([189]Fig. 6B and fig. S5, A to C) that we analyzed, BCR engagement of IgG1 receptors induced a higher and more sustained calcium flux than their IgM expressing counterparts. These results are consistent with a previous report that has shown enhanced calcium release upon IgG1 BCR engagement ([190]14). We next asked whether dampening of this enhanced calcium flux downstream to IgG1 BCR engagement could rescue defects in mitochondrial depolarization and survival. To interrogate this, we used a cell-permeant calcium chelator, BAPTA AM (BAPTA), to soak up intracellular calcium. For these studies, we used wild-type B cells activated under iGB conditions for 4 days, followed by BCR stimulation in the presence or absence of BAPTA ([191]Fig. 6C). As shown before, BCR stimulation led to enhanced mitochondrial depolarization in IgG1 cells compared to IgM cells ([192]Fig. 6, D and E). In addition, treatment with calcium chelator BAPTA (2 μM) under BCR-stimulated conditions partially reversed the accumulation of dysfunctional mitochondria in IgG1 cells when compared with vehicle [dimethyl sulfoxide (DMSO)] control ([193]Fig. 6, D and E). The decrease in dysfunctional mitochondria upon BCR engagement and BAPTA treatment was accompanied by a rescue in cell survival of IgG1 cells in comparison to BCR engagement alone ([194]Fig. 6, F and G). In summary, these data demonstrate that IgG1 BCRs, in comparison to IgM BCRs, trigger a stronger calcium flux, which promotes mitochondrial depolarization and reduced cell survival ([195]Fig. 6H). Our findings also show that blunting of calcium responses through chelation of intracellular Ca^2+ at least partially rescues mitochondrial function and survival in IgG1 expressing B cells ([196]Fig. 6H). Fig. 6. IgG1 BCR engagement triggers stronger calcium flux and impairs mitochondrial function. [197]Fig. 6. [198]Open in a new tab (A) Calcium flux measured by fluorescence intensity of Fluo4 (y axis) over time in IgM and IgG1 IgH^γ1μ/+ splenic B cells in response to BCR (anti-Igκ) stimulation. (B) Calcium flux measured by fluorescence intensity of Fluo4 in IgM and IgG1 OCI-Ly7 lymphoma cells in response to BCR (anti-Igκ) stimulation. (C) Schematic of the iGB in vitro culture system with treatment groups. (D) Representative flow cytometry plots of IgM and IgG1 B cells derived from iGB in vitro cultures stained with MitoTracker Green (x axis) and TMRM (y axis) 12 hours posttreatment. (E) Quantification of dysfunctional mitochondria frequency in IgM and IgG1 B cells from iGB in vitro cultures (n = 6 from two independent experiments). (F) Representative flow cytometry plots of cleaved caspase-3 from IgM and IgG1 B cells from iGB in vitro cultures 12 hours posttreatment. (G) Quantification of relative frequency of cleaved caspase 3 from IgM and IgG1 B cells from iGB in vitro cultures 12 hours poststimulation. Values are normalized to DMSO IgM and IgG1. (H) Proposed model showing that IgG1 B cell signaling triggers greater calcium flux, leading to increased mitochondrial dysfunction, which contributes to reduced cell survival. Dampening of calcium flux with BAPTA-AM rescues the survival of IgG1 expressing B cells. Statistical significance is calculated by repeated measures two-way ANOVA with Tukey’s multiple comparisons test [(E) and (G)]. Error bars represent means ± SEM; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. Schematics in (C) and (H) created in BioRender. Shukla, V. (2025) [199]https://BioRender.com/1mfdxwl. See also fig. S5. DISCUSSION Our studies here highlight how distinct BCR isotypes govern discrete B cell fates and could influence the pathogenesis of mature B cell–derived neoplasms. A previous study had linked distinct BCR isotypes to different patient outcomes in DLBCL ([200]9), and our studies provide mechanistic insights into how IgM and IgG1 isotypes might alter lymphoma pathogenesis. Although BCR signaling is known to be critical for the growth and survival of mature B cell malignancies, we identify important distinctions between IgM and IgG1 BCRs in lymphoma and normal B cells. The finding that mouse and human lymphoma cells expressing IgM outcompete those expressing IgG1 was unexpected to us, given that IgG1 BCRs signal more strongly and have been shown to better support proliferative responses in GCs ([201]13, [202]16, [203]23, [204]44). In addition to the increased proliferation of IgG1 cells reported previously, we consistently observed reduced survival in IgG1 cells in comparison to IgM. On the basis of these results, we propose a model in which AID activity in GC-derived lymphomas may frequently induce class switching to IgG1 (and other non-IgM) isotypes; however, IgM expressing cells would outcompete those expressing IgG1. That said, under the “right” mutational background (for instance, in the EZB DLBCL), non-IgM receptors may be favored, and it will be interesting to further parse out the mutational signatures enriched in lymphomas expressing IgG1 BCRs. Our studies suggest that categorization of lymphoma patients based on BCR isotypes could add substantial prognostic value for predicting patient outcomes in combination with the current genetic classification. Of specific importance is the clear association of IgG1 expression to better patient outcomes in the cohort of patients with DLBCL, which remain unclassified (Other subtype) even when the most comprehensive genetic classifications are applied. GCB-DLBCL and ABC-DLBCL (and other B cell lymphomas) are proposed to rely on antigen-independent “tonic” and antigen-dependent “active” BCR signaling, respectively ([205]6, [206]26, [207]27). For most of our studies, we used relatively low concentrations (1 μg/ml) of anti-kappa light chain antibody to induce signaling. This allowed us to more easily ascribe functional changes in cell survival, proliferation, transcription, and mitochondrial function to active BCR signaling. Under these conditions, the IgG1 cells are clearly more sensitive to BCR engagement, leading to enhanced cellular turnover, profound transcriptional changes, and increased mitochondrial dysfunction compared to IgM expressing cells. In human DLBCL, the chronically activated BCR signaling is presumed to be driven by recognition of self-antigens ([208]27). Even in the TET-deficient model of B cell lymphoma, our previous study identified features of BCR-driven selection ([209]38). Furthermore, some of our studies do highlight that the effects of IgG1 BCR are also relevant in the context of tonic BCR signaling. For example, in human xenograft studies, we did not exogenously stimulate the BCR, and although we cannot rule out recognition of specific antigen(s) in the NSG mice, our data suggest that IgM BCR is favored over IgG1 even in this seemingly tonic BCR context. This is further supported by a study in which IgG1 BCRs are shown to differ from IgM under conditions of both tonic and active BCR signaling ([210]28). Moreover, patients with different DLBCL subtypes show heterogeneous clinical responses to BCR targeting therapies. For instance, a study observed only a 5% response rate among GCB-DLBCL treated with the Btk inhibitor, ibrutinib, compared with patients with ABC-type DLBCL that showed 37% response rate ([211]45). ABC-DLBCL is shown to be enriched for IgM BCR, whereas GCB-DLBCL displays IgG1 BCR expression ([212]9, [213]27). Given that IgM and IgG1 BCRs greatly differ in their signaling potential, it will be intriguing to examine whether IgM and IgG1 BCRs could mediate differential sensitivities and clinical responses to BCR targeting therapies. The IgM BCR is preferred over IgG1 during normal B cell development in the bone marrow ([214]14). The IgM BCRs are also favored for the generation of long-lived memory cells as memory cells expressing switched IgG BCRs are either purged over time due to high mutational burden or taken over by high avidity IgM B cells from the naive repertoire ([215]22, [216]24, [217]25). In contrast to these scenarios, the IgG1 BCRs are better at promoting plasma cell differentiation than IgM ([218]14, [219]22). Studies by Sundling et al. used an isotype switchable BCR transgenic mouse specific to hen egg lysozyme (HEL) and showed that HEL-specific GC B cells expressing IgG1 are selected over IgM ([220]44). However, removal of the cytoplasmic tail in IgG1 or fusion of the IgG1 cytoplasmic tail to IgM, which is responsible for enhanced BCR signaling, could still not reverse IgG1 selection over IgM in these studies ([221]44). With these results, the authors suggested that perhaps another aspect of IgG receptor structure (such as antigen capture), rather than BCR signaling itself, might drive IgG1 selection in the GCs ([222]44). Besides this explanation, the higher signaling potential and the lower activation threshold of the IgG1 BCR may provide a selective advantage to IgG1 cells over IgM under conditions where BCR specificities are restricted, and antigen is limiting. In such scenarios, additional mechanisms should exist to tolerate pro-apoptotic BCR signaling outputs of IgG1 receptors. In support of this premise, a recent report showed that, in GCs, IgG1 cells depend on transcription factor Miz1 and its downstream target, an anti-apoptotic protein Tmbim4, to safeguard against Ca^2+-induced mitochondrial dysfunction ([223]46). Similarly, the GC cytokine IL-21, produced by T follicular helper cells, enhances the levels of phospho-AKT and the mTOR signaling target, phospho-S6 downstream of BCR and CD40 ([224]42). mTOR activity is a known regulator of mitochondrial biogenesis ([225]47), and we speculate that the protective effect of IL-21 on IgG1 survival observed in our studies is possibly an outcome of increased mitochondrial biogenesis. In alignment, another recent study showed that IL-21 better promotes the growth of IgG1 cells compared to IgM ([226]48). Therefore, in the context of the GC response, distinct protective mechanisms may exist to withstand the unwanted consequences of enhanced BCR signaling through IgG1. On the basis of our work and published literature, we postulate that IgG1 BCR would be favored in several physiological contexts, particularly under conditions where T cell help is available in the form of survival signals, such as IL-21. Unlike conventional GCs, survival signals such as IL-21 are likely limiting in the lymphoma microenvironment, and therefore IgG1 may not provide a strong selective advantage over IgM for malignant B cells. This advantage of IgM over IgG1 was generally true for most DLBCL genetic subtypes, except for EZB DLBCL, where IgG1 BCR showed no disadvantage when compared to IgM. We believe that this is likely due to the overexpression of anti-apoptotic protein BCL2 as a consequence of IGH-BCL2 translocations, which occur frequently in EZB DLBCL. Another important implication of our work is the link between distinct BCR isotypes and the unique metabolic features they impart on normal and malignant B cells. Our studies highlight that, due to their metabolic vulnerabilities, IgG1 expressing normal and malignant B cells may require additional survival signals, such as sustained T cell help (IL-21), for their expansion and persistence. In future work, it will be interesting to investigate how the lymphoma microenvironment differs between ABC-DLBCL and GCB-DLBCL subtypes or genetic subtypes of DLBCL, which show differential enrichment of IgM versus IgG BCRs. For our studies with human lymphoma cells, we primarily relied on CRISPR-Cas9–based editing to induce isotype class switching. One potential limitation of using this method would be that the stable expression of CRISPR-Cas9 could lead to additional mutations that may, in turn, affect cellular fitness. Therefore, to complement our studies with human lymphoma cells, we used primary murine B cells to confirm our findings in the context of normal B cell physiology wherever possible. In our studies with TET-deficient pre–malignant B cells, we also found that IgM cells outcompete IgG1. Nonetheless, our work warrants further investigation to carefully examine the role of different BCR isotypes of both restricted and diverse specificities in the context of normal GC and non–GC B cell responses, as well as autoimmune and malignant B cell pathologies, using additional genetic models. Although we focused on the role of IgG1 BCRs during lymphoma pathogenesis, GC-derived B cells lymphomas can express other IgG (subclasses) and IgA BCRs. Most studies characterizing the signaling differences between IgM and non-IgM BCR isotypes have mainly focused on IgG1 BCRs, and relatively less is known about the signaling properties of IgA, IgE, IgD, and other IgG subclasses, such as IgG2(s), IgG3, and IgG4. The differences in signaling potential of IgG1 to IgM are mainly attributed to its longer cytoplasmic tail ([227]12–[228]15, [229]17). In addition to a longer cytoplasmic tail, the IgG BCRs also harbor more flexible hinge regions, which are likely to support better antigen recognition. Furthermore, IgG1 BCRs are known to be less reliant on Ig α/β heterodimers for their signaling than IgM BCRs ([230]14). This could be an important distinction for lymphoma pathogenesis as gain-of-function mutations in the CD79B gene are common in the MCD genetic subtype, which are enriched in the ABC-DLBCL subgroup expressing IgM BCRs ([231]7, [232]31, [233]32). Together, IgG1 BCRs exhibit lower activation thresholds ([234]16), stronger BCR clustering ([235]13), and higher calcium responses ([236]14) and are associated with increased plasma cell differentiation ([237]23); these features of IgG1 were evident in our work. Similar features of enhanced calcium signaling and plasma cell differentiation have been reported for IgE and IgA BCRs ([238]18–[239]21, [240]49). It is worth pointing out that, in our in vivo studies with TET-deficient pre–malignant B cells, we observed populations of IgG2b-expressing and minor populations of IgA-, IgG3-, and IgD-expressing cells. IgG2 antibody responses are known to be directed toward polysaccharide bacterial antigens, and perhaps under conditions where BCR specificity is restricted to such antigens, IgG2 might be a relevant isotype to investigate further ([241]15). Besides more pronounced calcium responses downstream to IgG1 BCRs, it is quite likely that other differences intrinsic to IgG1 and IgM BCRs may functionally contribute to distinct cellular phenotypes ([242]14, [243]50). Enhanced calcium flux is shown to have a detrimental effect on mitochondrial function in several cellular systems. Mitochondria function as important cellular buffers of calcium, and excessive calcium buildup in mitochondria can cause opening of the mitochondrial permeability transition pore (mPTP), leading to cell death ([244]51). Studies from Akkaya and colleagues demonstrated a role of BCR-induced calcium responses in driving mitochondrial dysfunction ([245]43), and our studies highlight key differences in calcium responses downstream of IgM and IgG1 BCRs. We show that calcium-induced mitochondrial dysfunction in IgG1 cells renders them metabolically less fit in comparison to IgM. We suspect that the enrichment of transcriptional changes associated with one-carbon metabolism upon IgG1 stimulation are an adaptation to this dysfunctional mitochondrial state. Previous studies have identified a higher dependency on one-carbon metabolism through serine catabolism in cells with mitochondrial dysfunction ([246]40, [247]41). One-carbon metabolism is critical for growth of cancer cells, which heavily rely on glycolysis to provide energy and fuel anabolic pathways ([248]52, [249]53). The precise role of one-carbon metabolism in B cells expressing distinct BCR isotypes is an intriguing question for future studies. The ability of IgM cells to outcompete their IgG1 counterparts in these studies is likely related to the metabolic stress experienced by IgG1 cells. Pathways associated with calcium signaling are commonly mutated in B cell lymphomas ([250]54), and additional work will be needed to elucidate how these mutations alter the metabolic landscapes in B cell lymphomas. In summary, we provide exciting evidence that IgM and IgG BCRs are inherently distinct in their activities and differentially control the pathogenesis of B cell lymphomas. MATERIALS AND METHODS Mice Tet2fl/fl and Tet3fl/fl mice were generated as previously described ([251]55, [252]56). C57BL/6J (000664), Cd19-cre (006785), CD45.1 mice (002014, Ptprca), and NOD/SCID IL-2Rγ^−/− (NSG) (JAX#005557) were obtained from the Jackson Laboratory. IgH^γ1μ mice ([253]14) were provided by A.W. (Institute for Molecular Medicine, University of Mainz) to R.C.R. Experiments were performed with mice 8 to 12 weeks of age of both sexes, and experimental mice were age and sex matched. Animals were bred and housed at the specific pathogen–free facility of Sanford Burnham Prebys Medical Discovery Institute and Northwestern University. All procedures were performed under approved protocols by the Institutional Animal Care and Use Committee (IACUC). Primary B cell isolation and cell cultures The protocol for the in vitro iGB culture system was adapted from Haniuda and Kitamura ([254]57). 40LB feeder cells were irradiated with 30 gray. Irradiated 40LB cells were plated at a density of 3 × 10^6 cells per 10-cm dish and incubated overnight in Dulbecco’s modified Eagle’s medium (10-013-CV, Corning) supplemented with 10% fetal bovine serum (FBS) (35-016-CV, Corning), 10 mM Hepes (pH 7.4), 2 mM GlutaMAX, and 55 μM 2-mercaptoethanol (all from Life Technologies) at 37°C with 5% CO[2] to allow for adhesion. 40LB cells were obtained from the D. Kitamura laboratory (Tokyo University of Science). Primary naive B cells were isolated from mouse spleen using the EasySep Mouse B Cell Isolation Kit (19854, STEMCELL Technologies) and seeded on the irradiated 40LB feeder layer at a density of 600K cells per 10-cm dish with rmIL-4 (1 ng/ml; 214-14, PeproTech) in RPMI 1640 medium (11875-093, Gibco) supplemented with 10% heat-inactivated FBS, 10 mM Hepes (pH 7.4), 2 mM GlutaMAX, 1 mM sodium pyruvate, and 55 μM 2-mercaptoethanol (Life Technologies) at 37°C with 5% CO[2]. B cells were expanded for 3 or 4 days, depending on the experiment. To isolate IgM and IgG1 B cells from the culture, 40LB, IgE+, and CD138+ cells were depleted using biotinylated anti-H2Kd, IgE, and CD138 antibodies at a concentration of 1:200 with the EasySep Mouse Streptavidin RapidSpheres Isolation Kit (19860, STEMCELL Technologies). For stimulation experiments, the isolated IgG1 and IgM B cells were then stimulated for 12 hours with anti-kappa light chain antibody (1 μg/ml; 75861S, Cell Signaling Technology) in supplemented RPMI as previously described at a density of –2 million cells/ml. For IL-21 experiments, mouse IL-21 (210-21, PeproTech) was additionally added at a concentration of 10 ng/ml. For BAPTA experiments, mouse primary cells were treated with 2 μM BAPTA-AM (27-872-5, Tocris Bioscience) for 12 hours in RPMI media. Engineering of isogenic lymphoma cell lines, viral transduction, and cell cultures Lentiviral vectors expressing Cas9 and guide RNAs were obtained from T.-C.C. and R.C. ([255]37). Lentivirus was produced by transfecting 293T cells with pLenti lentiviral vectors targeting Sμ 5′ (2.5 μg) and Sy1 3′ (2.5 μg) and pMD2G (1.25 μg) and psPax2 (3.75 μg) packaging vectors. The lentiviral supernatants were added to BJAB, OCI-Ly7, and HBL-1 cells plated at a density of 200 × 10^3/ml of media and centrifuged at 2000 rpm at >20°C for 2 hours with polybrene (8 μg/ml). Transduced cells were selected starting at 48 hours posttransduction with puromycin (2 mg/ml) for at least 72 hours. Single clones were expanded to generate class-switched cell lines. Switching in Mino cells was induced using LPS (25 μg/ml), IL-4 (10 ng/ml), and anti-CD40 (200 ng/ml) for 5 days. Single clones were expanded to generate class-switched cell lines. All cell lines were routinely monitored and immunophenotyped with anti-IgM and anti-IgG1 antibodies to ensure their BCR expression. Isogenic lymphoma cell lines were cultured in RPMI supplemented as described for primary B cell cultures at 37°C with 5% CO[2] for all experiments. For stimulation experiments, BJAB and OCI-Ly7 human lymphoma cell lines were stimulated with anti-kappa antibody (2060-01, Southern Biotech) at a final concentration of 5 and 1 μg/ml, respectively, in supplemented RPMI media at a density of 0.25 million cells/ml for 24 hours (mitochondria staining) or 48 hours (cleaved caspase 3 staining). Transplantation experiments For Cd19 double knockout (DKO) transplantation studies, naive B cells from the spleen of Cd19 DKO mice were cultured using the described iGB culture system for 4 days. CD45.1 mice were sublethally irradiated with 600 centigray 24 hours before transfer of 500,000 Cd19 DKO B cells through the retro-orbital sinus. For wild-type transplantation studies, naïve B cells were isolated from CD45.1 mice and cultured using the described iGB culture system for 4 days. C57/BL6 recipient mice (CD45.2+) were injected with cyclophosphamide (10 mg/kg) 24 hours before transfer of 9 million CD45.1+ B cells through the retro-orbital sinus. Tumor engraftment For tumor engraftment studies with isogenic cell lines, 10^6 cells were injected subcutaneously into NSG mice. Mice health was monitored daily. Tumor volume was measured by (length × width^2/2 [cm^3]). Mice were euthanized when the total tumor volume reached 2 cm^3 or if they were in distress. Immunizations For immunizations with SRBCs, citrated SRBCs (31102, Colorado Serum Company) were washed two times with sterile phosphate-buffered saline (PBS) and injected intraperitoneally at a dose of 200 × 10^6 SRBCs on day 0, followed by a boost on day 5 of 10^9 SRBCs. Mouse spleens were collected and analyzed on day 14. Flow cytometry Primary cells and in vitro cultured cells were stained with appropriate antibodies in fluorescence-activated cell sorting (FACS) buffer [0.5% bovine serum albumin (BSA), 1 mM EDTA, and 0.05% sodium azide in PBS] for 30 min on ice. Cells were washed and fixed with 4% paraformaldehyde (PFA; Affymetrix) for 12 min on ice. For intracellular stains, cells were subsequently permeabilized with the eBioscience Foxp3/Transcription Factor Staining Buffer Set (00-5523, Invitrogen) at 4°C for 2 hours to overnight before staining with appropriate antibodies for 45 min on ice. Flow cytometry experiments were analyzed using the Cytek Aurora spectral cytometer, BD Canto II, and BD LSR Fortessa X20 18-color analytical flow cytometer. Antibodies were from BioLegend, eBioscience, BD Pharmingen, Miltenyi, and Cell Signaling Technology, as listed in [256]Table 1. Data were analyzed with FlowJo 10.9.0 software. Gating for flow cytometry analysis is described in the figure legends. Table 1. Antibodies used for flow cytometry staining. Antibodies (clone) Company Catalog no. Biotin rabbit anti-active caspase-3 BD Biosciences 550557 Anti-human IgG1 PE (IS11-12E4.23.20) Miltenyi Bio 130-119-946 Anti-human IgG1 Bio (IS11-12E4.23.20) Miltenyi Bio 130-119-858 lot no. 5220500500 Anti-human IgM PB BioLegend 314513 Biotin anti-mouse IgE BioLegend 406903 Biotin anti-mouse CD138 BioLegend 142511 Biotin anti-mouse H2kd BioLegend 116604 Anti-mouse IgM APC BioLegend 406509 Anti-mouse IgM BV711 BioLegend 406539 Anti-human kappa antibody Southern Biotech 2060-01 Anti-mouse kappa antibody Cell Signaling Technology 75861S LEAF purified anti-mouse CD40 antibody BioLegend 102810 Anti-mouse IgG1 PE/Cy7 BioLegend 406613 Anti-mouse Ki67 BV 421 BD Biosciences 562899 PE streptavidin BioLegend 405203 Anti-mouse IgD AF-700 BioLegend 405730 Anti-mouse IgA AF-647 Southern Biotech 1040-31 Anti-mouse IgG2b PE BioLegend 406707 Anti-mouse IgG3 PE/Cy7 Southern Biotech 1100-17 Anti-mouse FAS PE/Cy/7 BD Biosciences 557653 Anti-mouse IgG1 BV421 BioLegend 406616 AF-647 streptavidin BioLegend 405237 Anti-mouse IgG1 APC/Cy7 BioLegend 406619 Anti-mouse IgG1 PE BioLegend 406608 Anti-mouse CD138 PE/Cy7 BioLegend 142513 Anti-mouse CD19 PB BioLegend 115526 eBioscience Fixable Viability Dye eFluor 450 Thermo Fisher Scientific 65-0863-18 Anti-mouse B220 BV650 BioLegend 103241 Anti-mouse CD19 BV510 BioLegend 115546 Anti-mouse CD45.1 APC/Cy7 BioLegend 110716 Anti-mouse CD19 FITC BioLegend 115506 Streptavidin PerCP/Cy5.5 BioLegend 405214 Anti-mouse IgG1 FITC (A85-1) BD Biosciences 553443 Anti-mouse IgM PE/Cy7 (II/41) eBioscience 25-5790-82 Tomm20 (D8T4N) Cell Signaling Technology 42406S Anti-mouse IgK (187.1) BD Pharmingen 559750 Anti-mouse CD38 PE (90) BD Pharmingen 553764 Anti-mouse IgM FITC (II-41) eBioscience 11-5790-82 Active/Pro-Caspase-3 AF647 (C92-605) BD Biosciences 560626 Anti-human IgM PE (PJ2-22H3) Miltenyi Bio 130-122-930 Anti-human IgG1 APC (#IS11-12E4.23.20) Miltenyi Bio 130-119-857 Anti-mouse CD45.1 PECy7 BD Pharmingen 560578 Anti-mouse B220 APC (RA3-6B2) BD Pharmingen 553092 Anti-mouse CD45.2 Alexa Fluor 700 (104) BioLegend 109822 Anti-mouse CD45.2 APCCy7 BD Pharmingen 560694 [257]Open in a new tab Measurement of mitochondrial mass and membrane potential For mitochondrial mass staining, cells were incubated with 50 nM MitoTracker Green FM (7514, Invitrogen) in culture media for 25 min at 37°C. Cells were washed twice with FACS buffer (described above) and rapidly run on a flow cytometer. For experiments with primary cells, cells were subsequently stained with surface markers for 30 min and then rapidly run on a flow cytometer. For mitochondrial mass and membrane potential staining, cells were simultaneously incubated with 50 nM MitoTracker Green FM and 500 nM tetramethylrhodamine, methyl ester, perchlorate (TMRM) (T668, Invitrogen) following the same protocol. Dysfunctional mitochondria population was gated on TMRM low and MitoTracker Green–positive cells. Cell proliferation assays The BrdU incorporation assay (552598, BD Biosciences) was used to measure proliferation in the isogenic lymphoma cell lines. A total of 1 × 10^6 cells from each BJAB or OCI-Ly7 cell line were incubated with BrdU for 25 or 15 min, respectively, at a concentration of 10 μM in supplemented RPMI. Following incubation, cells were fixed and permeabilized according to kit instructions and then treated with 30 μg of DNase in PBS for 1 hour. Cells were stained with anti-BrdU APC antibody for 30 min and then run on a flow cytometer. Data were analyzed with FlowJo 10.9.0 software. BrdU+ gating was used to calculate the frequency of cells in the S phase. ImageStream X Cells from the spleen of SRBC-immunized mice were stained and processed as described above for intracellular staining. Samples were run on an Amnis ImageStreamX Mark II imaging flow cytometer at the Sanford Burnham Prebys Flow core facility. Data were analyzed using Inspired acquisition software. Confocal microscopy and analysis IgM and IgG1 expressing cells from IgH^γ1μ mice were incubated on glass slides precoated with poly-l-lysine for 15 min. Cells were stained with 1:100 anti-kappa antibodies for 25 min. Cells were washed twice and fixed with 4% PFA for 10 min at room temperature (RT), followed by two washes with PBS for 5 min. Cells were next permeabilized with 0.5% Triton X-100 for 10 min. Permeabilized cells were washed again two times with PBS for 5 min and then blocked in 500 μl of 1% BSA with PBS for 30 min. The blocking solution was replaced with 250 μl of Tomm20 (Cell Signaling Technology), 1:500 diluted in 1% BSA, and incubated overnight at 4°C. After three washes, cells were incubated in secondary antibody anti-rabbit (PE) 1:2000 in 1% BSA for 1 hour at RT. Cells were counterstained with 4′,6-diamidino-2-phenylindole (DAPI) for 10 min and then washed three times with PBS. A drop of antifade fluorescence mounting medium was added, and a coverslip was placed. All steps were performed in a humidified chamber at 37°C unless otherwise specified. Confocal images were acquired with a Leica SP8 confocal microscope and analyzed in Leica Application Suite X software v3.1.5.16308. Mean fluorescence intensity was quantified using Fiji6. Calcium flux assay Cells were stained with Ca^2+ indicator Fluo-4 ([258]F14201, Thermo Fisher Scientific). Briefly, cells were prepared according to the manufacturer’s guide in HBSS (Hanks’ balanced salt solution) with 1x Hepes, and an equal amount of 2X Fluo-4 was added to the cells. Cells were then incubated for 30 min at 37°C in a water bath. Fluorescence intensity of Fluo-4 was analyzed by flow cytometry using a BD Canto II flow cytometer. Calcium influx was initiated by injection of anti-kappa antibody (5 μg/ml). Cells were kept in a water bath throughout the assay unless acquiring the data. Seahorse OCR was measured using the XF24 extracellular flux analyzer (Agilent) according to the manufacturer’s guidelines. Briefly, cells were harvested and plated at concentrations of 500,000 cells per well (BJAB cell line) or 1.5 million (primary IgH^γ1μB cells) in Seahorse medium with 2 mM l-glutamine and 1 mM sodium pyruvate. Cells were then spun at 300g for 5 min with brakes off to create a monolayer with no gaps in XF24 plates. Injection of compounds occurred at the time points indicated in figures. Changes in OCR measured over time in IgM and IgG1 IgH^γ1μ/+ splenic B cells treated with oligomycin, phenylhydrazone (FCCP), and antimycin A and rotenone. Changes in OCR measured overtime in IgM and IgG1 BJAB lymphoma cells treated with anti-Igκ (5 μg/ml), oligomycin, FCCP, and antimycin A and rotenone. TCGA analysis We downloaded 481 preprocessed tsv files containing the transcriptomic profile, in transcripts per million (TPM) values of patients with DLBCL from TCGA (Project ID: NCICCR-DLBCL). We parsed the files on the basis of their IGHM and IGHG1 mRNA expression and identified 111 samples with high IGHM (IGHM TPM representing >50% of all IGH transcripts) 63 samples with high IGHG1 (IGHG1 TPM representing >20% and IGHM TPM no more than 20% of all IGH transcripts). We applied these filtering criteria taking into account that IGHM are default transcripts expressed on all B cells and will therefore be highly represented in a mixed pool (lymphoma and contaminating naïve cells) of B cells used in these studies. RNA sequencing For RNA-seq experiments, naive B cells were isolated from wild-type C57BL/6J mice spleen and cultured using the in vitro iGB culture system and stimulated with anti-kappa antibody (1 μg/ml) for 12 hours as previously described. After stimulation, B cells were FACS sorted from live cells, CD19+ and CD138− into IgG1+ and IgM+ using BD FACSAria 5L Sorter at the Northwestern University RHLCCC Flow Cytometry Facility. Total RNA was isolated using the RNeasy Plus Kit (74134, Qiagen) according to the manufacturer’s instructions. Quality of RNA was assessed using the Agilent Bioanalyzer RNA 6000 Pico Chip Bioanalyzer. The RNA-seq libraries were prepared using purified RNA (40 ng) with the NEBNext Ultra II RNA Library Prep Kit (E7770, New England Biolabs) according to the manufacturer’s instructions. Samples were sequenced on the Illumina NovaSeq 6000 platform [50–base pair (bp) reads]. RNA-seq analysis Paired-end (50 bp) reads were mapped to the mouse genome mm10/GRCm38 using STAR ([259]58) (2.7.10a) (--genomeLoad LoadAndRemove --outFilterMismatchNmax 4 --outFilterMultimapNmax 100 --winAnchorMultimapNmax 100). Counts were obtained with featureCounts ([260]59) (subread v2.0.1) (-g gene_name -s 0). DEGs were calculated with DESeq2 ([261]60), filtering out genes that did not have any count under any condition; cutoff to define DEGs was an adjusted P value of 0.05 and a log[2] fold change of ≥±1. The enrichment processes and pathway analysis of DEGs in gene ontology function was performed using Metascape ([262]http://metascape.org). Statistical analysis Statistical analyses were performed with GraphPad Prism 10.1.0. The statistical tests used to determine significance are described in the corresponding figure legends. Data are represented as means ± SE. No statistical methods were used to predetermine sample sizes. Parametric tests [t tests, analysis of variance (ANOVA)] were used in experiments where normal distribution could be assumed. No data points were excluded from the analysis, and appropriate animals/samples for each experiment were chosen randomly. Acknowledgments