An effective immune system depends upon regulation of lymphocyte function and homeostasis. In recent years, members of the GTPases of the immunity associated protein (GIMAP) family were proposed to regulate T cell homeostasis. In contrast, little is known about their function and mode of action in B cells. We used a combination of transgenic mice and in vivo and in vitro techniques to conditionally and electively ablate GIMAP1 in resting and activated peripheral B cells. Our data suggest that GIMAP1 is absolutely essential for the survival of peripheral B cells, irrespective of their activation state. Together with recent data showing increased expression of GIMAP1 in B cell lymphomas, our work points to the possible potential of GIMAP1 as a target for manipulation in a variety of B cell–mediated diseases.

The first identified member of the family of GTPases of the immunity associated proteins (GIMAPs) was GIMAP1, which was reported as a malaria-induced gene in mouse splenocytes (1). Since then, genetic association studies implicated human GIMAP genes in autoimmune diseases, including systemic lupus erythematosus (2), Behçet’s disease (3), and type I diabetes (4, 5). Furthermore, their deregulated expression was reported in lymphomas (611). Eight or nine GIMAP family members have been identified in mammals (12). They are a family of septin-related guanine nucleotide-binding G proteins that bear strong resemblance to dynamins (13). Mammalian GIMAPs are expressed prominently within lymphoid compartments, suggesting a role in lymphocyte function (12, 1419). In vivo and in vitro studies implied a role for GIMAPs in lymphoid homeostasis and survival (2030).

GIMAP5 is the most studied GIMAP family member. A mutation in Gimap5 was found to be the cause of lymphopenia seen in the Biobreeding diabetes-prone rat strain (14, 15). In GIMAP5-deficient rats, T cell development appears to occur normally within the thymus, but there are few T cells in the periphery (14, 15, 24, 31, 32). This was attributed to spontaneous apoptosis of T cells, although the mechanism by which this occurs remains unclear (24, 32, 33). Recent work suggested that T cell death may result from the inability of their mitochondria to sequester Ca2+ following capacitative entry (28). A similar paucity of peripheral T cells is seen in GIMAP5-deficient mice, which develop spontaneous colitis, resulting in early mortality (23, 26, 27). Gimap5 deficiency in mice affects various hematopoietic cell types (23, 27, 34) and can lead to a progressive multilineage failure of bone marrow hematopoiesis (34). Knowledge of the extent to which these effects are cell intrinsic awaits the use of conditional alleles in the study of Gimap5.

GIMAP5’s close relative, GIMAP1, is also required for the maintenance of peripheral T cells. Previously, we showed that conditional deletion of Gimap1 from lymphocyte progenitors using Gimap1f/fCD2Cre+ (CD2Cre) mice resulted in normal lymphocyte development but severe reductions in peripheral T cell numbers (22). Surprisingly, we also found a profound deficit of mature peripheral B cells. This study did not address GIMAP1 function in activated B cells. The role that GIMAPs might play in the survival of activated lymphocytes remains unresolved. Although GIMAP5-deficient rat T cells can be activated successfully via their AgRs, GIMAP5-deficient mouse T cells were reported to be unable to proliferate in response to ex vivo stimulation (24, 27, 35). More recently, other studies suggested an important role for GIMAP1 in mature B cells, highlighting its potential role in B cell lymphomas. Diffuse large B cell lymphomas (DLBCLs) show hypomethylation at the Gimap locus, resulting in overexpression of GIMAP1 (10). In addition, the Gimap cluster is found within an early replication fragile site hotspot (6). Early replication fragile site hotspots are proposed to play a mechanistic role in some of the most common genome rearrangements during B cell lymphomagenesis. These studies prompted us to examine, in greater depth, the role that GIMAP1 plays in B cell function. We used a combination of transgenic mice in conjunction with in vivo and in vitro techniques to show that GIMAP1 is required for the maintenance of B cell numbers in the resting peripheral pool, as well as throughout mature B cell activation and differentiation.

Mice were bred and maintained in specific pathogen–free conditions at The Babraham Institute. Husbandry and experimentation complied with existing United Kingdom Home Office and European Union legislation, as well as local standards, as approved by the Babraham Institute Animal Welfare and Ethical Review Body. Gimap1f/f mice [described previously (22)], bearing a “floxed” Gimap1 allele, were crossed with Cd79acre/+ mice (obtained from Michael Reth, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany) to generate Gimap1f/fCd79acre/+ mice, allowing conditional ablation of Gimap1 in the B cell lineage (36). Gimap1f/f mice were also crossed with ERT2Cre+ mice (obtained from Thomas Ludwig, Columbia University, New York, NY) to generate Gimap1f/fERT2Cre+ mice, enabling conditional ablation of Gimap1 upon administration of tamoxifen (37). To conditionally delete Gimap1 in germinal center (GC) B cells, Gimap1f/f mice were crossed with AicdaCre+ mice (38) (obtained from M. Busslinger, Research Institute of Molecular Pathology, Vienna, Austria) to generate Gimap1f/fAicdaCre+ animals. Gimap1f/fCd2cre/+ mice [described previously (22)] were crossed with Eμ-bcl-2-36–transgenic mice expressing human Bcl2 (39) to generate Gimap1f/fCd2cre/+Bcl2tg mice.

Mice were immunized i.p. with 100 μg 4-hydroxy-3-nitrophenylacetyl NP-19–keyhole limpet hemocyanin (KLH; Biosearch Technologies, Nocata, CA) adsorbed to alum in saline. They were bled at the indicated time points before boosting with soluble Ag. In adoptive-transfer experiments, lymph node cells from Gimap1f/fERT2Cre+ and ERT2Cre+ mice were stained with CFSE and CellTrace Violet (CTV; Life Technologies), respectively, and then mixed in a 1:2 ratio (Gimap1f/fERT2Cre+/ERT2Cre+) prior to i.v. injection of 5 × 106 cells/mouse into B6.SJL-Ptprca Pepcb/BoyJ mice. Mice were treated with 200 μg tamoxifen/g body weight or vehicle control i.p. on days 1 and 2 following adoptive cell transfer. On day 13 after cell transfer, mice were killed, and the numbers of transferred cells present in peripheral blood and spleen were determined on the basis of anti-CD45.1, anti-CD45.2, CFSE, CTV, and anti-B220 staining.

Single-cell suspensions were prepared from lymphoid tissues and peripheral blood. Abs were directed against the following surface markers: CD93, B220, CD38, GL7, Fas, IgG1, CD138, IgM, CD45.1, and CD45.2. DAPI and biotinylated nitrophenyl-4-hydroxy-5 iodophenacetic acid (NIP) cells were analyzed using an LSR II or Fortessa (BD Biosciences, Oxford, U.K.), and data were analyzed using FlowJo software (TreeStar, Ashland, OR). To purify GC and follicular B cells, splenocytes from animals immunized 7 d previously with NP-KLH in alum were depleted of CD43+ and CD11c+ cells using biotinylated anti-CD43 and anti-CD11c Abs, followed by anti-biotin beads (MACS; Miltenyi Biotec), prior to depletion on an AutoMACS (Miltenyi Biotec), resulting in >95% CD19+ cells. To fractionate CD19+ cells into GC and follicular B cells, cells were stained with B220, GL7, and CD95 and sorted for GC cells (B220+CD95+GL7+) and for follicular B cells (B220+CD95GL7). To measure levels of apoptosis, splenocytes were stained for B220, GL7, and CD95, and active caspase-3 was determined using a CaspGLOW Fluorescein Active Caspase-3 Staining Kit (BioVision, Milpitas, CA). For 5-ethynyl-2′-deoxyuridine (EdU) incorporation, mice were injected i.p. with 0.5 ml 2 mg/ml EdU in PBS, and splenocytes were harvested 90 min later. EdU incorporation was determined using the Molecular Probes Click-iT EdU Flow Cytometry Assay Kit (Thermo Fisher Scientific).

ELISA plates (Nunc, Paisley, U.K.) were coated with NP23-BSA or NP2-BSA (10 or 2 μg/ml, respectively) as previously described (40). For ELISPOT, MultiScreen-HA mixed cellulose ester plates (Millipore, Watford, U.K.) were coated with NP23-BSA or NP2-BSA in PBS, washed, and blocked before applying serially diluted cells and incubating overnight. Detection was performed using biotinylated anti-IgG1 and anti-IgM (SouthernBiotech, Birmingham, AL), alkaline phosphatase–conjugated streptavidin (R&D Systems), and 5-bromo-4-chloro-3-indoyl phosphate/nitro-blue tetrazolium substrate (R&D Systems), and the number of Ab-secreting cells (ASCs) was determined using a CTL ELISPOT reader and CTL ImmunoSpot Software (Cell Technologies, Shaker Heights, OH).

Lysates were prepared from FACS-purified GC and follicular B cells, and PCR for GIMAP1 and GIMAP8 was performed as previously described (22, 30).

FACS-purified GC and follicular B cells were lysed in Nonidet P-40 lysis buffer, and GIMAP1 and actin protein were detected by Western blot, as previously described (22).

Follicular B cells were purified as described above and cultured on fibroblasts (expressing CD40L and BAFF) with IL-4 in the presence of vehicle control (DMSO) or 50 nM 4-hydroxytamoxifen (4-OHT) for 4 d, prior to reculture in IL-21 for an additional 4 d, as described (41). Cells were counted using a CASY cell counter (Innovatis) and stained for flow cytometric analysis, as described above, using anti-CD138 and anti-IgG1.

We first determined the lineage-intrinsic nature of the B cell requirement for GIMAP1 by crossing our Gimap1f/f allele with CD79a-Cre, which is only active in the B cell lineage (36). Splenocytes, bone marrow, and peritoneal cells were enumerated and stained to determine the numbers of different B cells, as previously described (22). The B cell phenotype of these mice mirrored that of Gimap1f/fCD2Cre+ mice, in which numbers of pro-B, pre-B, and immature B cells were normal (Fig. 1A–C), but mature peripheral B cells were significantly reduced (Fig. 1D–I). In addition, type 2 (T2) and type 3 transitional B cells were reduced, as previously observed in GIMAP1f/fCD2Cre+ mice (Fig. 1J–L) (22). Previous studies suggested that GIMAPs might regulate survival by manipulation of Bcl-2 family members (25, 29, 34, 42). To address this, we crossed Gimap1f/fCD2Cre+ mice (in which Gimap1 is ablated in the lymphoid lineage) with Eμ-bcl-2-36–transgenic mice, which overexpress human Bcl2 (39). To our surprise, hBcl-2 expression did not overcome the effect of Gimap1 deletion on the numbers of mature B cells in bone marrow, spleen, and peritoneum of Gimap1f/fCD2Cre+ mice (Fig. 2). More interestingly, when expression of the hBcl-2 transgene caused an increase in cell numbers in control GIMAP1-sufficient mice (Gimap1f/f), this increase was not seen in GIMAP1-deficient animals (Gimap1f/fCD2Cre+). Signals through the BCR and BAFF receptor are also key for B cell survival. We found no differences in the expression of either of these receptors on GC B cells (Supplemental Fig. 1).

FIGURE 1.

Cell-intrinsic requirement for GIMAP1 in mature B cells. B cell subsets in bone marrow (AD), spleen (E, F, JL), and peritoneal cavity (GI) from Gimap1f/fCD79aCre+ mice (□) and age- and sex-matched Gimap1f/f controls (●) were enumerated. Gating is as previously described (22). Results show the number of cells/organ for individual mice (mean ± SD). *p < 0.05, ****p < 0.00005, unpaired two-tailed Student t test.

FIGURE 1.

Cell-intrinsic requirement for GIMAP1 in mature B cells. B cell subsets in bone marrow (AD), spleen (E, F, JL), and peritoneal cavity (GI) from Gimap1f/fCD79aCre+ mice (□) and age- and sex-matched Gimap1f/f controls (●) were enumerated. Gating is as previously described (22). Results show the number of cells/organ for individual mice (mean ± SD). *p < 0.05, ****p < 0.00005, unpaired two-tailed Student t test.

Close modal
FIGURE 2.

Bcl2 does not prevent cell death in GIMAP1-deficient B cells. B cell subsets in bone marrow (AC), spleen (D and E), and peritoneal cavity (FH) from Gimap1fl/fl (●), Gimap1f/fCD2Cre+ (□), Gimap1fl/flBcl2tg (▪), and Gimap1f/fCD2Cre+Bcl2tg (○) age- and sex-matched mice were enumerated, using previously described gating (22). Results show the number of cells/organ for individual mice (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.00005, unpaired two-tailed Student t test.

FIGURE 2.

Bcl2 does not prevent cell death in GIMAP1-deficient B cells. B cell subsets in bone marrow (AC), spleen (D and E), and peritoneal cavity (FH) from Gimap1fl/fl (●), Gimap1f/fCD2Cre+ (□), Gimap1fl/flBcl2tg (▪), and Gimap1f/fCD2Cre+Bcl2tg (○) age- and sex-matched mice were enumerated, using previously described gating (22). Results show the number of cells/organ for individual mice (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.00005, unpaired two-tailed Student t test.

Close modal

To examine the role of GIMAP1 in mature B cells in vivo, we generated mice, in which the Gimap1 gene can be inducibly deleted, by crossing Gimap1f/f mice with mice expressing Cre recombinase under the control of the estrogen receptor (ERT2Cre+) to create Gimap1f/fERT2Cre+ mice. This allowed conditional deletion of Gimap1 upon administration of tamoxifen or its synthetic derivative, 4-OHT. Deletion of Gimap1 was seen within 2 d of in vitro 4-OHT treatment of lymphocytes derived from Gimap1f/fERT2Cre+ mice (P. Datta, unpublished observations). To control for Cre-mediated toxicity, ERT2Cre+ mice were used alongside Gimap1f/fERT2Cre+ mice (43). In preliminary experiments, we administered tamoxifen to Gimap1f/fERT2Cre+ and ERT2Cre+ mice. We observed reductions in T and B cells in peripheral lymphoid organs (data not shown). To eliminate the possible effect of Gimap1 deletion in nonlymphoid cells, we went on to use an adoptive transfer model in which only 5 × 106 lymph node cells were injected into replete hosts. Using this system, only a small proportion of circulating lymphocytes is susceptible to Gimap1 deletion, which could be tracked by staining cells prior to transfer and by their expression of CD45.2. We adoptively transferred a mixture (1:2 ratio) of Gimap1f/fERT2Cre+ lymphocytes (stained with CFSE) and ERT2Cre+ lymphocytes (stained with CTV) into a replete, congenic CD45.1 host and administered tamoxifen to induce deletion of Gimap1 in transferred Gimap1f/fERT2Cre+ cells. Thirteen days later, splenocytes and peripheral blood were harvested and analyzed. Tamoxifen (but not vehicle) treatment resulted in the loss of nearly all Gimap1f/fERT2Cre+ B cells, with little effect on ERT2Cre+ B cells (Fig. 3). Fig. 3A and 3B show representative flow cytometry plots of CD45.2+B220+ splenocytes taken on day 13 after tamoxifen or vehicle treatment; transferred ErT2Cre+ and Gimap1f/fErT2Cre+ cells are stained with CTV or CFSE, respectively. It shows the disappearance of Gimap1f/fERT2Cre+ B cells and the relative retention of ErT2Cre+ B cells. Unstained cells shown in these FACS plots are transferred cells that had presumably lost their label because they were CD45.2+B220+. These data are representative of three independent experiments, and we found no differential effect of labeling with CTV or CFSE on lymphocyte survival; similar results were obtained when Gimap1f/fERT2Cre+ cells were labeled with CTV and ERT2Cre+ cells were labeled with CFSE (data not shown). This supports the view that the disappearance of Gimap1f/fERT2Cre+ B cells is due specifically to the loss of GIMAP1. The percentages of input Gimap1f/fERT2Cre+ and ERT2Cre+ B cells found in spleen were determined for each mouse (Fig. 3C, 3D). The low frequency of transferred cells in the spleen is typical for this type of experiment and reflects the dispersal of lymphocytes throughout lymphoid organs and multiple other tissues and organs of the body. We also show the ratio of recovered Gimap1f/fERT2Cre+/ERT2Cre+ B220+ cells from blood and spleen following tamoxifen or vehicle treatment (Fig. 3E, 3F). These results show that peripheral B cells intrinsically require continuous GIMAP1 expression for their maintenance.

FIGURE 3.

Mature B cells require GIMAP1 for their survival in the periphery. Lymph node cells from Gimap1fl/flERT2Cre+ and ERT2Cre+ mice were stained with CFSE and CTV, respectively, mixed in a 1:2 ratio (Gimap1fl/flERT2Cre+/ERT2Cre+), and injected into replete B6.SJL-Ptprca Pepcb/BoyJ (CD45.1+ve) mice. Mice were treated with vehicle control or tamoxifen, and spleen and blood were harvested 13 d later. Representative flow cytometry plots from spleen gated on transferred (CD45.2+veB220+ve) cells from vehicle-treated (A) or tamoxifen-treated (B) mice. Percentages of adoptively transferred Gimap1fl/flERT2Cre+ (C) and ERT2Cre+ (D) B220+ cells remaining in the spleen 13 d after vehicle or tamoxifen treatment of individual recipient mice. Each symbol represents the percentage of transferred cells remaining from an individual mouse (mean ± SD). Proportion of Gimap1fl/flERT2Cre+ and ERT2Cre+ cells found in blood (E) and spleen (F) of recipient animals treated with tamoxifen or vehicle control, presented as a ratio of their proportions and standardized by the percentages of Gimap1fl/flERT2Cre+ and ERT2Cre+ cells injected on day 0. Each panel is representative of three independent experiments (n = 4 mice/group). *p < 0.05, **p < 0.005, ****p < 0.00005, unpaired 2-tailed Student t test.

FIGURE 3.

Mature B cells require GIMAP1 for their survival in the periphery. Lymph node cells from Gimap1fl/flERT2Cre+ and ERT2Cre+ mice were stained with CFSE and CTV, respectively, mixed in a 1:2 ratio (Gimap1fl/flERT2Cre+/ERT2Cre+), and injected into replete B6.SJL-Ptprca Pepcb/BoyJ (CD45.1+ve) mice. Mice were treated with vehicle control or tamoxifen, and spleen and blood were harvested 13 d later. Representative flow cytometry plots from spleen gated on transferred (CD45.2+veB220+ve) cells from vehicle-treated (A) or tamoxifen-treated (B) mice. Percentages of adoptively transferred Gimap1fl/flERT2Cre+ (C) and ERT2Cre+ (D) B220+ cells remaining in the spleen 13 d after vehicle or tamoxifen treatment of individual recipient mice. Each symbol represents the percentage of transferred cells remaining from an individual mouse (mean ± SD). Proportion of Gimap1fl/flERT2Cre+ and ERT2Cre+ cells found in blood (E) and spleen (F) of recipient animals treated with tamoxifen or vehicle control, presented as a ratio of their proportions and standardized by the percentages of Gimap1fl/flERT2Cre+ and ERT2Cre+ cells injected on day 0. Each panel is representative of three independent experiments (n = 4 mice/group). *p < 0.05, **p < 0.005, ****p < 0.00005, unpaired 2-tailed Student t test.

Close modal

GIMAP1 expression in GC B cells has not been analyzed. Upon activation, Ag-specific B cells differentiate into long-lived memory B cells and plasma cells (PCs), which are generated within the specialized microenvironment of the GC. Microarray studies from the Immunological Genome Project (https://www.immgen.org) show that GC B cells express GIMAP1 mRNA (Fig. 4A). We looked at GIMAP1 protein expression in GC B cells (Fig. 4B). Although mRNA levels of GIMAP1 are reduced in GC B cells compared with follicular B cells, we found similar levels of GIMAP1 protein expressed in the two cell types, suggesting posttranscriptional regulation of GIMAP1 mRNA in B cells. To examine the role that GIMAP1 plays in B cell activation and differentiation, we specifically ablated Gimap1 in activated B cells by crossing Gimap1f/f mice with AicdaCre+ mice to generate Gimap1f/fAicdaCre+ mice. Aicda is expressed within GC B cells and was used successfully to elucidate the roles of many genes in the GC reaction (38, 44). We purified GC and follicular B cells from these animals and from Gimap1f/f controls by FACS sorting 7 d after NP-KLH immunization. Specific deletion of the Gimap1 gene was evident only in GC B cells (defined as B220+veGL7+veFas+ve) from Gimap1f/fAicdaCre+ mice (Fig. 5A). The nearby Gimap8 gene (analyzed as a control) was intact in GC and follicular B cells from both strains of mice.

FIGURE 4.

GIMAP1 expression in GC B cells. (A) Relative expression of GIMAP1 mRNA in immune cells (adapted from https://www.immgen.org/). (B) Western blot of GIMAP1 protein in follicular and GC B cells from WT mice. B.Fo.Sp, splenic follicular B cell; B.GC.Sp, splenic GC B cell; B.Mz.Sp, splenic marginal zone B cell; B1a.PC, peritoneal cavity B1 B cell.

FIGURE 4.

GIMAP1 expression in GC B cells. (A) Relative expression of GIMAP1 mRNA in immune cells (adapted from https://www.immgen.org/). (B) Western blot of GIMAP1 protein in follicular and GC B cells from WT mice. B.Fo.Sp, splenic follicular B cell; B.GC.Sp, splenic GC B cell; B.Mz.Sp, splenic marginal zone B cell; B1a.PC, peritoneal cavity B1 B cell.

Close modal
FIGURE 5.

GIMAP1 is essential for GC B cell responses. (A) Conditional deletion of Gimap1 in GC B cells. PCR analysis of Gimap1 and Gimap8 in GC B cells FACS-sorted from immunized Gimap1fl/flAicdaCre+ and Gimap1fl/fl mice. Lane 1, 100-bp DNA ladder; lane2, follicular B cells from control Gimap1fl/fl mice; lane3, GC B cells from control Gimap1fl/fl mice; lane4, follicular B cells from Gimap1fl/flAicdaCre+ mice; lane5, GC B cells from Gimap1fl/flAicdaCre+ mice; lane6, H2O control. (B) FACS plots showing NIP-binding IgG1-switched B cells in Gimap1fl/flAicdaCre+ and Gimap1fl/fl mice on day 7 p.i. (C) Enumeration of NIP-binding IgG1-switched (B220+veIgM-veIgD-ve) B cells on day 7 p.i. in Gimap1fl/fl (●) and Gimap1fl/flAicdaCre+ (□) mice. Results show the number of cells/spleen for individual mice (mean ± SD). (D) Titers of NP23-binding (low-affinity) and NP2-binding (high-affinity) IgG1 and IgM Abs on days 7 and 14 after primary immunization. Each symbol represents an individual mouse (●, Gimap1fl/fl; □, Gimap1fl/flAicdaCre+). Data are mean ± SD. (E) Frequency of NP-specific IgG1 and IgM ASCs from Gimap1fl/f (●) and Gimap1fl/flAicdaCre+ (□) mice 14 d p.i., as determined by ELISPOT. Each symbol represents an individual mouse (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, unpaired Student t test.

FIGURE 5.

GIMAP1 is essential for GC B cell responses. (A) Conditional deletion of Gimap1 in GC B cells. PCR analysis of Gimap1 and Gimap8 in GC B cells FACS-sorted from immunized Gimap1fl/flAicdaCre+ and Gimap1fl/fl mice. Lane 1, 100-bp DNA ladder; lane2, follicular B cells from control Gimap1fl/fl mice; lane3, GC B cells from control Gimap1fl/fl mice; lane4, follicular B cells from Gimap1fl/flAicdaCre+ mice; lane5, GC B cells from Gimap1fl/flAicdaCre+ mice; lane6, H2O control. (B) FACS plots showing NIP-binding IgG1-switched B cells in Gimap1fl/flAicdaCre+ and Gimap1fl/fl mice on day 7 p.i. (C) Enumeration of NIP-binding IgG1-switched (B220+veIgM-veIgD-ve) B cells on day 7 p.i. in Gimap1fl/fl (●) and Gimap1fl/flAicdaCre+ (□) mice. Results show the number of cells/spleen for individual mice (mean ± SD). (D) Titers of NP23-binding (low-affinity) and NP2-binding (high-affinity) IgG1 and IgM Abs on days 7 and 14 after primary immunization. Each symbol represents an individual mouse (●, Gimap1fl/fl; □, Gimap1fl/flAicdaCre+). Data are mean ± SD. (E) Frequency of NP-specific IgG1 and IgM ASCs from Gimap1fl/f (●) and Gimap1fl/flAicdaCre+ (□) mice 14 d p.i., as determined by ELISPOT. Each symbol represents an individual mouse (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, unpaired Student t test.

Close modal

In previous work, we were unable to address what role GIMAP1 might play in peripheral B cell function because Gimap1f/fCD2Cre+ mice have very few mature B cells (22), and the lymphopenia within these mice is likely to affect B cell function. By immunizing Gimap1f/fAicdaCre+ mice, we were able to determine whether GIMAP1 was required for peripheral B cell responses to a T-dependent Ag. Gimap1f/fAicdaCre+ mice were generated, and their B cells were examined. Numbers and phenotypes of naive B cells were unaffected by the transgene (Supplemental Fig. 2). Mice were immunized with NP-KLH in alum, and B cell responses were analyzed 7, 14, and 35 d later. We used binding of biotinylated NIP to detect cells bearing an NP-specific BCR (Fig. 5B, 5C). NP-specific IgG1-switched splenic B cells were identified by flow cytometry (B220+IgMIgDNIP+IgG1hi) (45) and enumerated (Fig. 5B, 5C). Gimap1f/fAicdaCre+ mice mounted a very poor response, with relatively few NP-specific switched B cells detectable (Fig. 5B, 5C). We found a similar deficiency in GC B cells, defined as B220+GL7+CD95+ (data not shown). A defect in GC function in the absence of GIMAP1 was also evident when serum Ig levels in Gimap1f/fAicdaCre+ and Gimap1f/f mice were compared (Fig. 5D). At day 7, a small reduction in NP-specific IgG1 was seen in Gimap1f/fAicdaCre+ mice compared with control mice. By day 14, this difference was much greater. This difference could reflect a slow turnover of functional GIMAP1 protein. In addition, we looked at affinity maturation by measuring Ab reactive with NP2 at 35 d postimmunization (p.i.) (Fig. 5D). Again, Gimap1f/fAicdaCre+ mice had very low titers of high-affinity Abs. The number of ASCs was determined by ELISPOT on days 14 and 42 p.i. (Fig. 5E). As expected, no significant differences were seen in the numbers of IgM-secreting cells from the spleen or bone marrow. In contrast, numbers of IgG1-secreting cells specific for NP23 and NP2 from spleen and bone marrow were lower in Gimap1f/fAicdaCre+ mice compared with control animals. In some mice, the number of ASCs was below the level of detection.

The impaired production of class-switched and high-affinity, Ag-specific Abs and the deficit of ASCs and long-lived PCs in Gimap1f/fAicdaCre+ mice suggested a defect in GC B cells. We enumerated splenic GC B cells as NIP-binding, IgG1+ and CD38 cells and looked at the development of the GC response on days 6, 8, and 10 p.i. The effect of Gimap1 deletion was evident by day 6 p.i. (Fig. 6A). By day 8 p.i., GC B cell numbers had decreased further in Gimap1f/fAicdaCre+ mice. A slight recovery was seen at day 10 p.i., possibly reflecting the expansion of cell clones that had escaped Gimap1 deletion.

FIGURE 6.

GC responses fail to develop in the absence of GIMAP1 in Gimap1fl/flAicdaCre+ (●) and control Gimap1fl/fl (□) mice. (A) NIP-binding, IgG1+, CD38 B cells on days 6, 8, and 10 p.i. (B) Percentages of GC cells that had incorporated EdU (data for individual mice). (C) Percentages of GC B cells that were positive for active caspase-3. Results show values for individual mice (mean ± SD). *p < 0.05, **p < 0.005, Student t test.

FIGURE 6.

GC responses fail to develop in the absence of GIMAP1 in Gimap1fl/flAicdaCre+ (●) and control Gimap1fl/fl (□) mice. (A) NIP-binding, IgG1+, CD38 B cells on days 6, 8, and 10 p.i. (B) Percentages of GC cells that had incorporated EdU (data for individual mice). (C) Percentages of GC B cells that were positive for active caspase-3. Results show values for individual mice (mean ± SD). *p < 0.05, **p < 0.005, Student t test.

Close modal

To investigate why GC B cells failed to develop in the absence of GIMAP1, we examined their proliferation and death. EdU incorporation into newly synthesized DNA was used to measure GC B cell proliferation on day 8 p.i. (Fig. 6B). There was a slight, but significant, decrease in the proliferation of these cells in Gimap1f/fAicdaCre+ mice compared with controls. To look at cell death in GC B cells, we measured the percentage of GC B cells expressing active caspase-3 by flow cytometry. We found a small, but significant, difference in the percentage of GC B cells expressing active caspase-3 in cells from Gimap1f/fAicdaCre+ mice (Fig. 6C).

To confirm our in vivo data, we made use of an in vitro system to generate induced GC (iGC) B cells (41). B cells were purified from spleens and cultured with fibroblasts expressing BAFF and CD40L. IL-4 was added to the culture for the first 4 d to generate iGC B cells and then IL-21 was added to induce their differentiation into PCs. B cells from Gimap1f/fERT2Cre+ and control ERT2Cre+ mice were cultured for 8 d in the presence of 4-OHT or vehicle control. We were able to delete GIMAP1 by day 6 of culture and examine cell proliferation, GC development, differentiation to PCs, and cell death. Within 6 d of culture, all cells expressed GL7 and CD95, indicative of a GC-like phenotype (data not shown). The slope of the curves in vehicle-treated Gimap1f/fERT2Cre+ and ERT2Cre+ cells was similar, suggesting that their rate of proliferation was the same. However, 4-OHT–treated Gimap1f/fERT2Cre+ cells, but not control ERT2Cre+ cells, appeared to stop proliferating between days 4 and 8 (Fig. 7A). We found no difference in the percentage of cells that further differentiated into PCs (CD38+IgG1+; Fig. 7B), showing that lack of GIMAP1 has no effect on differentiation. In addition, we looked at DAPI uptake by these cells to determine the percentage of live cells. 4-OHT–treated Gimap1f/fERT2Cre+ cells had significantly fewer live (DAPI) cells at day 8. This suggests that cells initiated apoptosis (evident on day 6) and stopped proliferating, resulting in a reduction in the number of iGC B cells and percentage of live cells on day 8 (Fig. 7C).

FIGURE 7.

In vitro generation of iGC and PCs is compromised by lack of GIMAP1. B cells from Gimap1f/fERT2Cre+ and ERT2Cre+ mice were cultured to induce GC cell and PC differentiation in the presence of 4-OHT or vehicle control. Cells were harvested on days 4, 6, and 8, counted, and stained with anti-IgG1, anti-CD138, and DAPI. (A) Total number of PCs generated from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). (B) Percentages of cells that had differentiated into PCs from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). (C) Percentages of live PCs, as determined by the exclusion of DAPI, from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). Each symbol represents an individual mouse (mean ± SD). *p < 0.05, ***p < 0.0005, unpaired Student t test.

FIGURE 7.

In vitro generation of iGC and PCs is compromised by lack of GIMAP1. B cells from Gimap1f/fERT2Cre+ and ERT2Cre+ mice were cultured to induce GC cell and PC differentiation in the presence of 4-OHT or vehicle control. Cells were harvested on days 4, 6, and 8, counted, and stained with anti-IgG1, anti-CD138, and DAPI. (A) Total number of PCs generated from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). (B) Percentages of cells that had differentiated into PCs from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). (C) Percentages of live PCs, as determined by the exclusion of DAPI, from Gimap1f/fERT2Cre+ and ERT2Cre+ B cells in the presence of 4-OHT (□) or vehicle (●). Each symbol represents an individual mouse (mean ± SD). *p < 0.05, ***p < 0.0005, unpaired Student t test.

Close modal

To determine whether memory B cell responses were also affected in Gimap1f/fAicdaCre+ mice, we looked at secondary responses to NP-KLH. Thirty-five days after primary immunization, Gimap1f/fAicdaCre+ mice and control mice were boosted with NP-KLH. Memory responses were analyzed 7 d later. The numbers of Ag-specific IgG1-switched B cells were significantly reduced in Gimap1f/fAicdaCre+ animals; indeed, in some animals, none could be detected (Fig. 8A). NP-specific IgG1 Ab levels were also reduced, most markedly for high-affinity Ab (as determined by NP2 binding; Fig. 8B). When the numbers of IgG1 ASCs binding to NP23 and NP2 were determined, many Gimap1f/fAicdaCre+ mice had numbers below our detection limit of 1 in 106 cells (Fig. 8C).

FIGURE 8.

Failure to establish B cell memory in the absence of GIMAP1. Secondary responses in Gimap1fl/flAicdaCre+ (□) and control Gimap1fl/f (●) mice. (A) NIP-binding, IgG1-switched splenic B cells enumerated using flow cytometric analysis. (B) Titers of NP23-binding (total-affinity) and NP2-binding (high-affinity) IgG1 and IgM Abs on day 7 of secondary immunization. (C) Frequency of NP-specific IgG1 and IgM ASCs from Gimap1fl/fl (●) and Gimap1fl/flAicdaCre+ (□) mice 7 d p.i., as determined by ELISPOT. Each symbol represents an individual mouse (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, unpaired Student t test.

FIGURE 8.

Failure to establish B cell memory in the absence of GIMAP1. Secondary responses in Gimap1fl/flAicdaCre+ (□) and control Gimap1fl/f (●) mice. (A) NIP-binding, IgG1-switched splenic B cells enumerated using flow cytometric analysis. (B) Titers of NP23-binding (total-affinity) and NP2-binding (high-affinity) IgG1 and IgM Abs on day 7 of secondary immunization. (C) Frequency of NP-specific IgG1 and IgM ASCs from Gimap1fl/fl (●) and Gimap1fl/flAicdaCre+ (□) mice 7 d p.i., as determined by ELISPOT. Each symbol represents an individual mouse (mean ± SD). *p < 0.05, **p < 0.005, ***p < 0.0005, unpaired Student t test.

Close modal

Regulation of lymphocyte function and homeostasis is paramount for an effective and balanced immune system. Dysregulation of this balance frequently leads to autoimmune diseases, inability to fight infection, or cancer. Understanding the mechanisms maintaining this homeostasis may offer opportunities for the selective ablation of lymphocyte pools as a therapeutic strategy. Over the last 15 y, members of the GIMAP family have been implicated as important modulators of peripheral T lymphocyte homeostasis and survival. Very few studies addressed the role of GIMAPs in B cell biology. We showed that deletion of GIMAP8 results in a reduction in the number of recirculating B cells in the bone marrow (30). The generation and characterization of GIMAP1- and GIMAP5-deficient mouse strains further demonstrated that peripheral B cell survival is also influenced by GIMAPs (22, 23, 27, 34). As observed for T cells, deletion of Gimap1 appears to affect only mature cells within the B cell lineage (22). In contrast, GIMAP5-deficient mice show reductions in pro/pre-B cell progenitors, resulting in a more marked reduction in immature and mature B cells. This defect is thought to be due to the effect of GIMAP5 deficiency on hematopoietic stem cells (34). In a separate study, numbers of GIMAP5-deficient mouse B cells were also reduced in the periphery, and severe reductions in B cell responses to immunization were observed (27). As a caveat, there is an important methodological difference between our studies of GIMAP1 deficiency and those of GIMAP5; we used conditional, Cre-mediated ablation of Gimap1, whereas the Gimap5-related studies were conducted using germline mutant animals. In the absence of studies using conditional or elective deletion of the Gimap5 gene, it is not possible to assert whether the B cell changes reported in GIMAP5-deficient animals are B cell intrinsic or mediated via effects on other cells.

We have undertaken the development of mouse strains that enable specific and elective ablation of Gimap1 in B cells. Thus, the effects that we describe cannot be attributed to a “legacy” effect of GIMAP1 (i.e., the result of a function that GIMAP1 performs during B cell development that affects the functional capacity of the cells at a later stage) or to a cell-extrinsic effect acting on B cells. Adoptive transfer of B cells, followed by ablation of their Gimap1 gene, resulted in their disappearance from the periphery. This result reflects an intrinsic requirement for GIMAP1 in mature B cells, most likely to prevent their cell death in the periphery.

Importantly, our in vivo studies showed that B cells further require GIMAP1 during the development of Ab responses. To our knowledge, this is the first study to show the conditional ablation of a GIMAP protein during lymphocyte activation. Our data strongly suggest that B cells require GIMAP1 for their postactivation expansion because, in our in vitro iGC culture, we saw differentiation of cells into iGCs and PCs but not the expected subsequent expansion of these populations. Similar to the results of our in vitro experiments, GIMAP1-deficient GC B cells do not go through normal expansion in vivo. There is also reduced Ab production and affinity maturation, recall IgM response, and an inability to generate a long-lived memory response. These data establish GIMAP1 as absolutely essential for B cell peripheral function.

The deficit of GC B cells in Gimap1f/fAicdaCre+ mice makes elucidation of the mechanism of GIMAP1 action challenging. We were able to detect an increase in active caspase-3 in GC B cells from Gimap1f/fAicdaCre+ mice, indicative of apoptosis, and we also observed a reduction in the percentage of proliferating GC B cells. Although these differences were relatively small, the GC reaction is a rapidly evolving B cell response in which small changes in rates of proliferation and/or cell death can have a large impact on cell numbers (46). It is also likely that the few GC B cells that we detect are a mixture of GIMAP1-deficient GC B cells destined to die and GC B cells that have escaped Gimap1 gene deletion (47).

The mode of action of GIMAP1 remains enigmatic. GIMAP5 was reported to associate with Mcl1 and HSC70 and to promote stabilization of Mcl1 at the mitochondrial membrane (34). Mcl1 is critical for the survival of GC B cells and PCs (48, 49). It is possible that GIMAP1 also plays a role in Mcl1 stabilization that is not compensated by GIMAP5. However, we found no evidence of this in Gimap1-deficient GC cells. Furthermore, expression of the hBcl-2 transgene did not overcome the survival defect of mature B cells in Gimap1f/fCD2Cre+ mice. An hBcl-2 transgene was shown to increase numbers of immature and mature B cell subsets (50). Interestingly, deletion of GIMAP1 overcame this effect of hBcl-2, suggesting that GIMAP1-induced lymphocyte survival uses a mechanism distinct from that of Bcl-2 family members or works downstream of Bcl-2 family members. We also looked at expression levels of IgM and BAFFR, both key mediators of B cell survival. However, no defects were detected in GIMAP1-deficient GC cells. In previous work, we showed that GIMAP5 associates with lysosomes, whereas GIMAP1 associates with the Golgi apparatus (51). Hence, it may be valid to speculate that B cells deficient in GIMAP1 succumb to lysosome-related cell death: lysosomal hydrolases are delivered to late endosomes via the mannose-6-phosphate pathway from the trans-Golgi network, where GIMAP1 is located. GIMAP1 loss may compromise the structural integrity of endosomes, enabling their release into the cytosol. Indeed, apoptosis of human GC B cells was shown to involve lysosomal destabilization, resulting in GC B cell death (52).

The fact that B cells can develop normally in the absence of GIMAP1, yet fail to maintain their peripheral numbers, suggests that the survival mechanisms of developing and mature B cells are distinct. Many proteins involved in B cell survival are required for both developing and mature B cells. However, members of the NF-κB family show B cell knockout phenotypes similar to Gimap1f/fCD2Cre+ mice and are important for the survival of mature B cells, irrespective of their activation status (47, 5357). In Gimap1f/fCD2Cre+ mice, the B cell defect is first evident in the immature transitional T2 cells found in the spleen (22, 53). This implicates a problem in either the differentiation of type 1 cells into T2 cells or a failure of T2 cells to survive. A similar block is seen in mice deficient in members of the NF-κB family (53, 54). NF-κB activation is required for the differentiation of type 1 cells into T2 cells, suggesting that the BCR provides a survival signal mediated by NF-κB (5357). This raises the possibility that tonic signaling through the BCR provides a survival signal to peripheral B cells that also relies upon GIMAP1. Perturbations in NF-κB signaling are seen in GIMAP5-deficient T cells, resulting in their deregulation (58). Dysregulation of the NF-κB signaling pathway is also seen in B cell lymphomas, with DLBCLs being strongly dependent on NF-κB activity (5961). DLBCLs also have a hypomethylated region in the Gimap locus, resulting in overexpression of GIMAP1 and GIMAP5 (10). It is tempting to speculate that GIMAP1 and GIMAP5 regulate cell survival via the NF-κB pathway and that manipulation of either protein can tilt the balance between lymphocyte death and survival (58).

In summary, this work shows that GIMAP1 is required for the establishment and maintenance of the peripheral B cell pool and for all stages of postactivation B cell survival. In the absence of GIMAP1, mature B cells die, irrespective of their activation status or function. Together with our previous work, this establishes GIMAP1 as a key prosurvival factor for mature B lymphocytes and a potential target for the control of B cell–mediated diseases.

We thank Michael Reth for Cd79aCre+ mice, Thomas Ludwig for ERT2Cre mice, Meinrad Busslinger for AicdaCre+ mice, members of the Babraham Institute Biological Services Unit for animal husbandry, members of the Babraham Institute Flow Cytometry Facility for assistance with flow cytometry and cell sorting, and Michelle Linterman for critical review of the manuscript.

This work was supported by a project grant from the Medical Research Council (MR/J001856/1) and Institute Strategic Programme Grant BBS/E/B/000C0409 from the Biotechnology and Biological Sciences Research Council.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ASC

Ab-secreting cell

CTV

CellTrace Violet

DLBCL

diffuse large B cell lymphoma

EdU

5-ethynyl-2′-deoxyuridine

GC

germinal center

GIMAP

GTPase of the immunity associated protein

iGC

induced GC

KLH

keyhole limpet hemocyanin

NIP

nitrophenyl-4-hydroxy-5 iodophenacetic acid

4-OHT

4-hydroxytamoxifen

PC

plasma cell

p.i.

postimmunization

T2

type 2.

1
Krücken
J.
,
Stamm
O.
,
Schmitt-Wrede
H. P.
,
Mincheva
A.
,
Lichter
P.
,
Wunderlich
F.
.
1999
.
Spleen-specific expression of the malaria-inducible intronless mouse gene imap38.
J. Biol. Chem.
274
:
24383
24391
.
2
Hellquist
A.
,
Zucchelli
M.
,
Kivinen
K.
,
Saarialho-Kere
U.
,
Koskenmies
S.
,
Widen
E.
,
Julkunen
H.
,
Wong
A.
,
Karjalainen-Lindsberg
M. L.
,
Skoog
T.
, et al
.
2007
.
The human GIMAP5 gene has a common polyadenylation polymorphism increasing risk to systemic lupus erythematosus.
J. Med. Genet.
44
:
314
321
.
3
Lee
Y. J.
,
Horie
Y.
,
Wallace
G. R.
,
Choi
Y. S.
,
Park
J. A.
,
Choi
J. Y.
,
Song
R.
,
Kang
Y. M.
,
Kang
S. W.
,
Baek
H. J.
, et al
.
2013
.
Genome-wide association study identifies GIMAP as a novel susceptibility locus for Behcet’s disease.
Ann. Rheum. Dis.
72
:
1510
1516
.
4
Jailwala
P.
,
Waukau
J.
,
Glisic
S.
,
Jana
S.
,
Ehlenbach
S.
,
Hessner
M.
,
Alemzadeh
R.
,
Matsuyama
S.
,
Laud
P.
,
Wang
X.
,
Ghosh
S.
.
2009
.
Apoptosis of CD4+ CD25(high) T cells in type 1 diabetes may be partially mediated by IL-2 deprivation.
PLoS One
4
:
e6527
.
5
Heinonen
M. T.
,
Laine
A. P.
,
Söderhäll
C.
,
Gruzieva
O.
,
Rautio
S.
,
Melén
E.
,
Pershagen
G.
,
Lähdesmäki
H. J.
,
Knip
M.
,
Ilonen
J.
, et al
Finnish Pediatric Diabetes Registry
.
2015
.
GIMAP GTPase family genes: potential modifiers in autoimmune diabetes, asthma, and allergy.
J. Immunol.
194
:
5885
5894
.
6
Barlow
J. H.
,
Faryabi
R. B.
,
Callén
E.
,
Wong
N.
,
Malhowski
A.
,
Chen
H. T.
,
Gutierrez-Cruz
G.
,
Sun
H. W.
,
McKinnon
P.
,
Wright
G.
, et al
.
2013
.
Identification of early replicating fragile sites that contribute to genome instability.
Cell
152
:
620
632
.
7
Chadwick
N.
,
Zeef
L.
,
Portillo
V.
,
Boros
J.
,
Hoyle
S.
,
van Doesburg
J. C.
,
Buckle
A. M.
.
2010
.
Notch protection against apoptosis in T-ALL cells mediated by GIMAP5.
Blood Cells Mol. Dis.
45
:
201
209
.
8
Chadwick
N.
,
Zeef
L.
,
Portillo
V.
,
Fennessy
C.
,
Warrander
F.
,
Hoyle
S.
,
Buckle
A. M.
.
2009
.
Identification of novel Notch target genes in T cell leukaemia.
Mol. Cancer
8
:
35
.
9
Wang
H.
,
Zou
J.
,
Zhao
B.
,
Johannsen
E.
,
Ashworth
T.
,
Wong
H.
,
Pear
W. S.
,
Schug
J.
,
Blacklow
S. C.
,
Arnett
K. L.
, et al
.
2011
.
Genome-wide analysis reveals conserved and divergent features of Notch1/RBPJ binding in human and murine T-lymphoblastic leukemia cells.
Proc. Natl. Acad. Sci. USA
108
:
14908
14913
.
10
Chambwe
N.
,
Kormaksson
M.
,
Geng
H.
,
De
S.
,
Michor
F.
,
Johnson
N. A.
,
Morin
R. D.
,
Scott
D. W.
,
Godley
L. A.
,
Gascoyne
R. D.
, et al
.
2014
.
Variability in DNA methylation defines novel epigenetic subgroups of DLBCL associated with different clinical outcomes.
Blood
123
:
1699
1708
.
11
Zenz
T.
,
Roessner
A.
,
Thomas
A.
,
Fröhling
S.
,
Döhner
H.
,
Calabretta
B.
,
Dahéron
L.
.
2004
.
hIan5: the human ortholog to the rat Ian4/Iddm1/lyp is a new member of the Ian family that is overexpressed in B-cell lymphoid malignancies.
Genes Immun.
5
:
109
116
.
12
Krücken
J.
,
Schroetel
R. M.
,
Müller
I. U.
,
Saïdani
N.
,
Marinovski
P.
,
Benten
W. P.
,
Stamm
O.
,
Wunderlich
F.
.
2004
.
Comparative analysis of the human gimap gene cluster encoding a novel GTPase family.
Gene
341
:
291
304
.
13
Schwefel
D.
,
Fröhlich
C.
,
Eichhorst
J.
,
Wiesner
B.
,
Behlke
J.
,
Aravind
L.
,
Daumke
O.
.
2010
.
Structural basis of oligomerization in septin-like GTPase of immunity-associated protein 2 (GIMAP2).
Proc. Natl. Acad. Sci. USA
107
:
20299
20304
.
14
Hornum
L.
,
Rømer
J.
,
Markholst
H.
.
2002
.
The diabetes-prone BB rat carries a frameshift mutation in Ian4, a positional candidate of Iddm1.
Diabetes
51
:
1972
1979
.
15
MacMurray
A. J.
,
Moralejo
D. H.
,
Kwitek
A. E.
,
Rutledge
E. A.
,
Van Yserloo
B.
,
Gohlke
P.
,
Speros
S. J.
,
Snyder
B.
,
Schaefer
J.
,
Bieg
S.
, et al
.
2002
.
Lymphopenia in the BB rat model of type 1 diabetes is due to a mutation in a novel immune-associated nucleotide (Ian)-related gene.
Genome Res.
12
:
1029
1039
.
16
Cambot
M.
,
Aresta
S.
,
Kahn-Perlès
B.
,
de Gunzburg
J.
,
Roméo
P. H.
.
2002
.
Human immune associated nucleotide 1: a member of a new guanosine triphosphatase family expressed in resting T and B cells.
Blood
99
:
3293
3301
.
17
Poirier
G. M.
,
Anderson
G.
,
Huvar
A.
,
Wagaman
P. C.
,
Shuttleworth
J.
,
Jenkinson
E.
,
Jackson
M. R.
,
Peterson
P. A.
,
Erlander
M. G.
.
1999
.
Immune-associated nucleotide-1 (IAN-1) is a thymic selection marker and defines a novel gene family conserved in plants.
J. Immunol.
163
:
4960
4969
.
18
Krücken
J.
,
Epe
M.
,
Benten
W. P.
,
Falkenroth
N.
,
Wunderlich
F.
.
2005
.
Malaria-suppressible expression of the anti-apoptotic triple GTPase mGIMAP8.
J. Cell. Biochem.
96
:
339
348
.
19
Stamm
O.
,
Krücken
J.
,
Schmitt-Wrede
H. P.
,
Benten
W. P.
,
Wunderlich
F.
.
2002
.
Human ortholog to mouse gene imap38 encoding an ER-localizable G-protein belongs to a gene family clustered on chromosome 7q32-36.
Gene
282
:
159
167
.
20
Carter
C.
,
Dion
C.
,
Schnell
S.
,
Coadwell
W. J.
,
Graham
M.
,
Hepburn
L.
,
Morgan
G.
,
Hutchings
A.
,
Pascall
J. C.
,
Jacobs
H.
, et al
.
2007
.
A natural hypomorphic variant of the apoptosis regulator Gimap4/IAN1.
J. Immunol.
179
:
1784
1795
.
21
Schnell
S.
,
Démollière
C.
,
van den Berk
P.
,
Jacobs
H.
.
2006
.
Gimap4 accelerates T-cell death.
Blood
108
:
591
599
.
22
Saunders
A.
,
Webb
L. M.
,
Janas
M. L.
,
Hutchings
A.
,
Pascall
J.
,
Carter
C.
,
Pugh
N.
,
Morgan
G.
,
Turner
M.
,
Butcher
G. W.
.
2010
.
Putative GTPase GIMAP1 is critical for the development of mature B and T lymphocytes.
Blood
115
:
3249
3257
.
23
Schulteis
R. D.
,
Chu
H.
,
Dai
X.
,
Chen
Y.
,
Edwards
B.
,
Haribhai
D.
,
Williams
C. B.
,
Malarkannan
S.
,
Hessner
M. J.
,
Glisic-Milosavljevic
S.
, et al
.
2008
.
Impaired survival of peripheral T cells, disrupted NK/NKT cell development, and liver failure in mice lacking Gimap5.
Blood
112
:
4905
4914
.
24
Hernández-Hoyos
G.
,
Joseph
S.
,
Miller
N. G.
,
Butcher
G. W.
.
1999
.
The lymphopenia mutation of the BB rat causes inappropriate apoptosis of mature thymocytes.
Eur. J. Immunol.
29
:
1832
1841
.
25
Nitta
T.
,
Nasreen
M.
,
Seike
T.
,
Goji
A.
,
Ohigashi
I.
,
Miyazaki
T.
,
Ohta
T.
,
Kanno
M.
,
Takahama
Y.
.
2006
.
IAN family critically regulates survival and development of T lymphocytes.
PLoS Biol.
4
:
e103
.
26
Aksoylar
H. I.
,
Lampe
K.
,
Barnes
M. J.
,
Plas
D. R.
,
Hoebe
K.
.
2012
.
Loss of immunological tolerance in Gimap5-deficient mice is associated with loss of Foxo in CD4+ T cells.
J. Immunol.
188
:
146
154
.
27
Barnes
M. J.
,
Aksoylar
H.
,
Krebs
P.
,
Bourdeau
T.
,
Arnold
C. N.
,
Xia
Y.
,
Khovananth
K.
,
Engel
I.
,
Sovath
S.
,
Lampe
K.
, et al
.
2010
.
Loss of T cell and B cell quiescence precedes the onset of microbial flora-dependent wasting disease and intestinal inflammation in Gimap5-deficient mice.
J. Immunol.
184
:
3743
3754
.
28
Chen
X. L.
,
Serrano
D.
,
Mayhue
M.
,
Wieden
H. J.
,
Stankova
J.
,
Boulay
G.
,
Ilangumaran
S.
,
Ramanathan
S.
.
2013
.
GTPase of the immune-associated nucleotide-binding protein 5 (GIMAP5) regulates calcium influx in T-lymphocytes by promoting mitochondrial calcium accumulation.
Biochem. J.
449
:
353
364
.
29
Yano
K.
,
Carter
C.
,
Yoshida
N.
,
Abe
T.
,
Yamada
A.
,
Nitta
T.
,
Ishimaru
N.
,
Takada
K.
,
Butcher
G. W.
,
Takahama
Y.
.
2014
.
Gimap3 and Gimap5 cooperate to maintain T-cell numbers in the mouse.
Eur. J. Immunol.
44
:
561
572
.
30
Webb
L. M.
,
Pascall
J. C.
,
Hepburn
L.
,
Carter
C.
,
Turner
M.
,
Butcher
G. W.
.
2014
.
Generation and characterisation of mice deficient in the multi-GTPase domain containing protein, GIMAP8.
PLoS One
9
:
e110294
.
31
Pandarpurkar
M.
,
Wilson-Fritch
L.
,
Corvera
S.
,
Markholst
H.
,
Hornum
L.
,
Greiner
D. L.
,
Mordes
J. P.
,
Rossini
A. A.
,
Bortell
R.
.
2003
.
Ian4 is required for mitochondrial integrity and T cell survival.
Proc. Natl. Acad. Sci. USA
100
:
10382
10387
.
32
Ramanathan
S.
,
Poussier
P.
.
2001
.
BB rat lyp mutation and Type 1 diabetes.
Immunol. Rev.
184
:
161
171
.
33
Keita
M.
,
Leblanc
C.
,
Andrews
D.
,
Ramanathan
S.
.
2007
.
GIMAP5 regulates mitochondrial integrity from a distinct subcellular compartment.
Biochem. Biophys. Res. Commun.
361
:
481
486
.
34
Chen
Y.
,
Yu
M.
,
Dai
X.
,
Zogg
M.
,
Wen
R.
,
Weiler
H.
,
Wang
D.
.
2011
.
Critical role for Gimap5 in the survival of mouse hematopoietic stem and progenitor cells.
J. Exp. Med.
208
:
923
935
.
35
Ramanathan
S.
,
Norwich
K.
,
Poussier
P.
.
1998
.
Antigen activation rescues recent thymic emigrants from programmed cell death in the BB rat.
J. Immunol.
160
:
5757
5764
.
36
Hobeika
E.
,
Thiemann
S.
,
Storch
B.
,
Jumaa
H.
,
Nielsen
P. J.
,
Pelanda
R.
,
Reth
M.
.
2006
.
Testing gene function early in the B cell lineage in mb1-cre mice.
Proc. Natl. Acad. Sci. USA
103
:
13789
13794
.
37
de Luca
C.
,
Kowalski
T. J.
,
Zhang
Y.
,
Elmquist
J. K.
,
Lee
C.
,
Kilimann
M. W.
,
Ludwig
T.
,
Liu
S. M.
,
Chua
S. C.
 Jr.
2005
.
Complete rescue of obesity, diabetes, and infertility in db/db mice by neuron-specific LEPR-B transgenes.
J. Clin. Invest.
115
:
3484
3493
.
38
Kwon
K.
,
Hutter
C.
,
Sun
Q.
,
Bilic
I.
,
Cobaleda
C.
,
Malin
S.
,
Busslinger
M.
.
2008
.
Instructive role of the transcription factor E2A in early B lymphopoiesis and germinal center B cell development.
Immunity
28
:
751
762
.
39
Strasser
A.
,
Harris
A. W.
,
Cory
S.
.
1991
.
bcl-2 transgene inhibits T cell death and perturbs thymic self-censorship.
Cell
67
:
889
899
.
40
Rolf
J.
,
Bell
S. E.
,
Kovesdi
D.
,
Janas
M. L.
,
Soond
D. R.
,
Webb
L. M.
,
Santinelli
S.
,
Saunders
T.
,
Hebeis
B.
,
Killeen
N.
, et al
.
2010
.
Phosphoinositide 3-kinase activity in T cells regulates the magnitude of the germinal center reaction.
J. Immunol.
185
:
4042
4052
.
41
Nojima
T.
,
Haniuda
K.
,
Moutai
T.
,
Matsudaira
M.
,
Mizokawa
S.
,
Shiratori
I.
,
Azuma
T.
,
Kitamura
D.
.
2011
.
In-vitro derived germinal centre B cells differentially generate memory B or plasma cells in vivo.
Nat. Commun.
2
:
465
.
42
Filén
S.
,
Lahesmaa
R.
.
2010
.
GIMAP Proteins in T-Lymphocytes.
J. Signal Transduct.
2010
:
268589
.
43
Jullien
N.
,
Goddard
I.
,
Selmi-Ruby
S.
,
Fina
J. L.
,
Cremer
H.
,
Herman
J. P.
.
2008
.
Use of ERT2-iCre-ERT2 for conditional transgenesis.
Genesis
46
:
193
199
.
44
Muramatsu
M.
,
Sankaranand
V. S.
,
Anant
S.
,
Sugai
M.
,
Kinoshita
K.
,
Davidson
N. O.
,
Honjo
T.
.
1999
.
Specific expression of activation-induced cytidine deaminase (AID), a novel member of the RNA-editing deaminase family in germinal center B cells.
J. Biol. Chem.
274
:
18470
18476
.
45
Xu
S.
,
Guo
K.
,
Zeng
Q.
,
Huo
J.
,
Lam
K. P.
.
2012
.
The RNase III enzyme Dicer is essential for germinal center B-cell formation.
Blood
119
:
767
776
.
46
MacLennan
I. C.
1994
.
Germinal centers.
Annu. Rev. Immunol.
12
:
117
139
.
47
Pasparakis
M.
,
Schmidt-Supprian
M.
,
Rajewsky
K.
.
2002
.
IkappaB kinase signaling is essential for maintenance of mature B cells.
J. Exp. Med.
196
:
743
752
.
48
Peperzak
V.
,
Vikström
I.
,
Walker
J.
,
Glaser
S. P.
,
LePage
M.
,
Coquery
C. M.
,
Erickson
L. D.
,
Fairfax
K.
,
Mackay
F.
,
Strasser
A.
, et al
.
2013
.
Mcl-1 is essential for the survival of plasma cells.
Nat. Immunol.
14
:
290
297
.
49
Vikstrom
I.
,
Carotta
S.
,
Lüthje
K.
,
Peperzak
V.
,
Jost
P. J.
,
Glaser
S.
,
Busslinger
M.
,
Bouillet
P.
,
Strasser
A.
,
Nutt
S. L.
,
Tarlinton
D. M.
.
2010
.
Mcl-1 is essential for germinal center formation and B cell memory.
Science
330
:
1095
1099
.
50
Maraskovsky
E.
,
Peschon
J. J.
,
McKenna
H.
,
Teepe
M.
,
Strasser
A.
.
1998
.
Overexpression of Bcl-2 does not rescue impaired B lymphopoiesis in IL-7 receptor-deficient mice but can enhance survival of mature B cells.
Int. Immunol.
10
:
1367
1375
.
51
Wong
V. W.
,
Saunders
A. E.
,
Hutchings
A.
,
Pascall
J. C.
,
Carter
C.
,
Bright
N. A.
,
Walker
S. A.
,
Ktistakis
N. T.
,
Butcher
G. W.
.
2010
.
The autoimmunity-related GIMAP5 GTPase is a lysosome-associated protein.
Self Nonself
1
:
259
268
.
52
van Nierop, K., F. J. Muller, J. Stap, C. J. Van Noorden, M. van Eijk, and C. de Groot. 2006. Lysosomal destabilization contributes to apoptosis of germinal center B-lymphocytes. J. Histochem. Cytochem. 54: 1425–1435
.
53
Kaileh
M.
,
Sen
R.
.
2012
.
NF-κB function in B lymphocytes.
Immunol. Rev.
246
:
254
271
.
54
Baldwin
A. S.
2012
.
Regulation of cell death and autophagy by IKK and NF-κB: critical mechanisms in immune function and cancer.
Immunol. Rev.
246
:
327
345
.
55
Li
Z. W.
,
Omori
S. A.
,
Labuda
T.
,
Karin
M.
,
Rickert
R. C.
.
2003
.
IKK beta is required for peripheral B cell survival and proliferation.
J. Immunol.
170
:
4630
4637
.
56
Kaisho
T.
,
Takeda
K.
,
Tsujimura
T.
,
Kawai
T.
,
Nomura
F.
,
Terada
N.
,
Akira
S.
.
2001
.
IkappaB kinase alpha is essential for mature B cell development and function.
J. Exp. Med.
193
:
417
426
.
57
Kim
S.
,
La Motte-Mohs
R. N.
,
Rudolph
D.
,
Zuniga-Pflucker
J. C.
,
Mak
T. W.
.
2003
.
The role of nuclear factor-kappaB essential modulator (NEMO) in B cell development and survival.
Proc. Natl. Acad. Sci. USA
100
:
1203
1208
.
58
Kupfer
R.
,
Lang
J.
,
Williams-Skipp
C.
,
Nelson
M.
,
Bellgrau
D.
,
Scheinman
R. I.
.
2007
.
Loss of a gimap/ian gene leads to activation of NF-kappaB through a MAPK-dependent pathway.
Mol. Immunol.
44
:
479
487
.
59
Nagel
D.
,
Vincendeau
M.
,
Eitelhuber
A. C.
,
Krappmann
D.
.
2014
.
Mechanisms and consequences of constitutive NF-κB activation in B-cell lymphoid malignancies.
Oncogene
33
:
5655
5665
.
60
Vermeer
M. H.
,
Willemze
R.
.
2014
.
Recent advances in primary cutaneous B-cell lymphomas.
Curr. Opin. Oncol.
26
:
230
236
.
61
Horie
R.
2013
.
Molecularly-targeted strategy and NF-κB in lymphoid malignancies.
J. Clin. Exp. Hematop.
53
:
185
195
.

The authors have no financial conflicts of interest.

Supplementary data