Skip to main content
The Journal of Biological Chemistry logoLink to The Journal of Biological Chemistry
. 2015 Apr 22;290(28):17056–17072. doi: 10.1074/jbc.M115.645739

Rac-mediated Stimulation of Phospholipase Cγ2 Amplifies B Cell Receptor-induced Calcium Signaling*,

Claudia Walliser ‡,1, Kyrylo Tron §,1, Karen Clauss §, Orit Gutman , Andrei Yu Kobitski , Michael Retlich , Anja Schade , Carlheinz Röcker §, Yoav I Henis ¶,2, G Ulrich Nienhaus ‖,**,‡‡, Peter Gierschik ‡,3
PMCID: PMC4498044  PMID: 25903139

Background: Phospholipase Cγ2 (PLCγ2) is stimulated by Rac GTPases through direct protein-protein interaction.

Results: The Rac-PLCγ2 interaction markedly enhances B cell-receptor-mediated Ca2+ mobilization and nuclear translocation of the Ca2+-regulated transcription factor NFAT in B cells.

Conclusion: Rac-mediated stimulation of PLCγ2 activity amplifies B cell receptor-induced Ca2+ signaling.

Significance: A specific Rac-resistant PLCγ2 variant is used to determine the physiological cell signaling relevance of a functional Rac-PLCγ2 interaction in an appropriate cellular context.

Keywords: calcium, lymphocyte, phospholipase C, Rac (Rac GTPase), signal transduction, B cell, signal amplification

Abstract

The Rho GTPase Rac is crucially involved in controlling multiple B cell functions, including those regulated by the B cell receptor (BCR) through increased cytosolic Ca2+. The underlying molecular mechanisms and their relevance to the functions of intact B cells have thus far remained unknown. We have previously shown that the activity of phospholipase Cγ2 (PLCγ2), a key constituent of the BCR signalosome, is stimulated by activated Rac through direct protein-protein interaction. Here, we use a Rac-resistant mutant of PLCγ2 to functionally reconstitute cultured PLCγ2-deficient DT40 B cells and to examine the effects of the Rac-PLCγ2 interaction on BCR-mediated changes of intracellular Ca2+ and regulation of Ca2+-regulated and nuclear-factor-of-activated-T-cell-regulated gene transcription at the level of single, intact B cells. The results show that the functional Rac-PLCγ2 interaction causes marked increases in the following: (i) sensitivity of B cells to BCR ligation; (ii) BCR-mediated Ca2+ release from intracellular stores; (iii) Ca2+ entry from the extracellular compartment; and (iv) nuclear translocation of the Ca2+-regulated nuclear factor of activated T cells. Hence, Rac-mediated stimulation of PLCγ2 activity serves to amplify B cell receptor-induced Ca2+ signaling.

Introduction

Inositol phospholipid-specific phospholipases C (PLC)4 catalyze the formation of inositol 1,4,5-trisphosphate (InsP3) and diacylglycerol (DAG) from plasma membrane lipid substrate phosphatidylinositol 4,5-bisphosphate (PtdInsP2) (1). Both the rise of the former two and the decline of the latter may serve as intracellular signals to regulate a myriad of cell functions (2). In B lymphocytes, receptors for cell surface immunoglobulins such as the B cell receptors (BCR), cleavage fragments of the third complement component (CD19/CD21) (3), bacterial, viral, or autoimmunity host DNA (toll-like receptors) (4), and even certain G-protein-coupled chemokine receptors (5) mediate activation of PLCγ2, one of the two human PLCγ isoforms. The activity of PLCγ2 controls many B cell functions, such as protein kinase signaling, nucleocytoplasmic trafficking of transcription factors, proliferation, differentiation, cytoskeletal reorganization, cell adhesion and migration, immunological synapse formation, affinity maturation, autoimmunity, homing to and retention in tissue microenvironments, survival, and susceptibility to transformation (6, 7).

Inactivation of the PLCγ2 gene in the mouse caused specific defects in most cell types of hematopoietic origin, except for T cells (8, 9). Mice lacking PLCγ2 showed reduced numbers of mature conventional B cells, a block in pro-B cell differentiation, B1 B cell deficiency, absence of IgM receptor-mediated Ca2+ responses, and B cell-mitogen-induced cell proliferation. PLCγ2 also plays important roles in pre-BCR-mediated early B cell development, in BAFF receptor-mediated survival, and in activation of light-chain loci for recombination as well as receptor editing of self-reactive B cells (1012). Mutationally activated forms of PLCγ2 have been identified in mice subjected to N-ethyl-N-nitrosourea mutagenesis (Ali5 and Ali14) and, more recently, in patients with inherited forms of autoinflammation and immunodeficiency (1316). These defects also lead to deregulation of B cell functions.

Several of the changes described for PLCγ2−/− B cells, e.g. defective Ca2+ signaling, failure to proliferate in response to immunoglobulin receptor stimulation, impediment of B cell development, and failure to mount humoral responses to TD and TI antigens, were also observed in mice carrying deletions in all three genes encoding Vav guanine nucleotide exchange factors of Rho GTPases, Vav1, -2, and -3 (17). These results were difficult to interpret mechanistically because Vav proteins elicit both RhoGEF-dependent and -independent effects (18). However, some of the B cell defects were also observed in mice lacking either Rac2 (19) or both Rac1 and Rac2 (20), including a reduced ability of BCR or CD19 (co)ligation to increase [Ca2+]i, suggesting that at least some of the B cell defects commonly observed in PLCγ2 and Vav1/2/3-null mice were due to loss of Rac activation. At that time, the available evidence suggested that Rac GTPases might activate PLCγ2 indirectly by enhancing the activity of phosphatidylinositol 4-phosphate 5-kinase (21), thus increasing the level of PtdInsP2, the substrate of both PLCγ2 and phosphoinositide 3-kinase (20). Enhanced availability of substrate to the former and enhanced formation of PtdInsP3 by the latter were expected to activate PLCγ2 (22).

We have previously shown that Rac GTPases, but not Cdc42 or RhoA, activate PLCγ2, but not PLCγ1, by direct protein-protein interaction (23). Neither enhanced formation of PtdInsP2 nor PtdInsP3 nor protein tyrosine phosphorylation are involved in this effect. Unlike activation of PLCβ2, which is mediated by Rac interacting with the N-terminal PH domain of this effector, activation of PLCγ2 involves binding of Rac to the bipartite, split PH domain (spPH) juxtaposed between the two halves, X and Y, of the PLCγ2 catalytic domain (24). The three-dimensional structures of the heterodimeric complex between PLCγ2 spPH and GTPγS-activated Rac2, monomeric spPH, and monomeric Rac2 liganded with either GTPγS or GDP allowed us to elucidate the conformational changes that accompany the formation of the signaling active PLCγ2-spPH/Rac2 heterodimer (25). A residue unique for spPH of PLCγ2, but not PLCγ1, Phe-897, was found to be particularly important for the functional and structural PLCγ2-spPH/Rac2 interaction. Replacement of Phe-897 to the corresponding glutamine residue of PLCγ1, F897Q, did not affect the overall three-dimensional structure of the PLCγ2 spPH domain, but it specifically blocked the interaction of the mutant domain with activated Rac2 (24, 25).

Rac GTPases are present at many intracellular crossroads of B cell signaling. They receive inputs from numerous cell surface receptors to regulate and integrate a host of intracellular signaling proteins, including PLCγ2 and many proteins involved in cytoskeletal organization (26). This has made it intrinsically difficult to judge the pertinence of the PLCγ2-Rac interaction observed in cell-free experiments and overexpression studies to cell signaling in more physiologically relevant cellular contexts. In this work, we have used DT40 B cells genetically deficient in PLCγ2 for functional reconstitution with the Rac-resistant PLCγ2 mutant F897Q to study the effects of a specific loss of the PLCγ2-Rac interaction on BCR-mediated cell signaling. The results reveal that loss of this functional interaction causes a marked decrease of BCR-mediated Ca2+ release from intracellular stores, Ca2+ entry from the extracellular compartment, and nuclear translocation of the Ca2+-regulated transcription factor NFAT. Some of these changes have previously been observed in Rac-deficient mice (19, 20). Hence, the results may provide a mechanistic explanation for findings on the role of Rac GTPases in B cell signaling obtained in vivo that have as yet remained largely unexplained. In addition, these insights into BCR-mediated cell signaling may also apply to the mechanisms of action of other B cell receptors such as CD19/CD21, to other cells of hematopoietic origin, e.g. platelets, and to human diseases, such as certain immunodeficiencies. To our knowledge, this is the first time that a Rho-resistant but otherwise normal Rho effector was reintroduced into a genetically Rho effector-deficient background to determine the relevance of the functional Rho effector interaction in a biologically highly relevant context.

Experimental Procedures

Antibodies and Reagents

Mouse monoclonal antibody reactive against the c-Myc epitope (9B11, catalogue no. 2276) was from Cell Signaling. Mouse monoclonal antibody reactive against β-actin (AC-15, catalogue no. A3854), poly-l-lysine (catalogue no. P6282), and ionomycin (catalogue no. I-0634) was from Sigma. Anti-phosphotyrosine antibody (catalogue no. 05-321, 4G10) was purchased from Millipore. Mouse anti-chicken IgM (M-4, catalogue no. 8300-01) was obtained from SouthernBiotech. Alexa Fluor® 488 goat anti-mouse antibody (catalogue no. A-11029), fluo-4 acetoxymethyl ester (catalogue no. F-14201), Pluronic®-F127 (catalogue number P-3000MP), and thapsigargin (catalogue no. T-7459) were from Molecular Probes® (Life Technologies, Inc.). Trypsin (catalogue no. 1418475001) was from Roche Applied Sciences, and puromycin was from InvivoGen.

cDNA Cloning

Because the 5′ end of the mRNA encoding chicken PLCγ2 was unknown at the time, 5′ rapid amplification of cDNA ends (27) was used to gather this information and produce full-length PLCγ2 cDNAs from reverse-transcribed DT40 cell mRNA. Two presumably allelic variants were found, which are identical at the protein level to each other and to database entry XP_414166, except for a Gln to His divergence at position 865. Based on the higher frequency (7/10) of His-865 among PCR products of DT40 cell mRNA, this haplotype was used herein for further studies. A histidine is present at this position in PLCγ2 of numerous species ranging from fish, such as coelacanth, to mammals, such as cattle or sheep. The plasmid NFAT1c-td-RFP611 encodes amino acids 1–400 of mouse NFAT1c fused to a pseudo-monomeric tandem dimer red fluorescent protein, td-RFP611 (28).

Expression Constructs and Reconstitution of PLCγ2−/− DT40 B Cells with Wild-type or F897Q Mutant PLCγ2

The F897Q variant of chicken PLCγ2 was created by site-directed mutagenesis using the primers 5′-GCAACTGATAAAGTAGAAGAACTGCAGGAATGGTACCAAAGTGTCCGTGAA-3′ (sense) and 5′-TTCACGGACACTTTGGTACCATTCCTGCAGTTCTTCTACTTTATCAGTTGC-3′ (antisense). The chicken PLCγ2 deletion mutants PLCγ2ΔPCI and PLCγ2ΔPCIF897Q lacking the phospholipase C inhibitor (PCI) peptide (amino acids 727–734) were constructed by site-directed mutagenesis using the primers 5′-GAGAAGCACCCGCTGCCTGTGACTGAGGAGC-3′ (sense) and 5′-GCTCCTCAGTCACAGGCAGCGGGTGCTTCTC-3′ (antisense). For expression in COS-7 cells, the cDNAs of C-terminally c-Myc epitope-tagged wild-type (WT) PLCγ2, PLCγ2F897Q, PLCγ2ΔPCI, and PLCγ2ΔPCIF897Q were ligated into the BamHI/NotI site of pcDNA3.1(+). For production of recombinant baculoviruses, the cDNAs of c-Myc epitope-tagged PLCγ2 and PLCγ2F897Q were ligated into the BamHI/NotI site of pVL1393. To create stably transfected DT40 cell clones, cDNAs encoding c-Myc epitope-tagged PLCγ2 and PLCγ2F897Q were ligated into the BglII/SmaI site of the expression vector pExpress, in which the expression of a cloned cDNA is controlled by the chicken β-actin promoter and an SV40 poly(A) signal (29). The pExpress cDNA expression cassette was then combined with the vector pLoxPuro by using the restriction site SpeI generating an expression vector, in which only the selectable marker is flanked by mutant loxP sites (29). For electroporation, the plasmids PLCγ2-pLoxPuro and PLCγ2F897Q-pLoxPuro were linearized by the restriction enzyme NotI, precipitated, and resuspended in distilled water to a final concentration of 1 μg/μl. To obtain nontargeted integration of the constructs into the genome of DT40 B cells, 20–30 μg of linearized plasmid DNA were introduced into 20 × 106 PLCγ2−/− DT40 cells by electroporation at 250 V and 950 microfarads resulting in a time constant of 22 ms. For selection, the cells were incubated for 11 days at 37 °C and 10% CO2 in RPMI 1640 medium supplemented with 10% (v/v) heat-inactivated (56 °C, 30 min) fetal calf serum (FCS), 1% (v/v) heat-inactivated chicken serum (catalogue no. C5405, Sigma), 50 μm β mercaptoethanol, 100 units/ml penicillin, and 100 μg/ml streptomycin (henceforth referred to as supplements) and 0.5 μg/ml puromycin. For FRAP experiments, human PLCγ2 fused C-terminally to enhanced green fluorescent protein (GFP) was constructed by inserting the PLCγ2 cDNA into the KpnI site of pEGFPN3 (Clontech). The F897Q variant of human PLCγ2-pEGFPN3 was produced by site-directed mutagenesis using the primers 5′-GACAGGGTGGAGGAGCTCCAGGAGTGGTTTCAGAGCATC-3′ (sense) and 5′-GATGCTCTGAAACCACTCCTGGAGCTCCTCCACCCTGTC-3′ (antisense).

Cell Culture and Analysis of Inositol Phosphate Formation

Chicken WT (RCB1464) and PLCγ2-deficient (PLCγ2−/−; RCB1469) DT40 B cells (all from Cell Bank, RIKEN BioResource Center, Japan) were cultured at 37 °C in a humidified atmosphere of 90% air and 10% CO2 in RPMI 1640 medium containing supplements. DT40 B cells stably reconstituted with either WT or mutant PLCγ2 were cultured in RPMI 1640 medium containing supplements and 0.5 μg/ml puromycin. COS-7 cells were maintained at 37 °C in a humidified atmosphere of 90% air and 10% CO2 in Dulbecco's modified Eagle's medium (Invitrogen, catalogue no. 41965-039) supplemented with 10% (v/v) FCS (Invitrogen, catalogue no. 10270-106), 2 mm glutamine, 100 units/ml penicillin, and 100 μg/ml streptomycin (all from PAA Laboratories). Transfection of COS-7 cells and analysis of inositol phosphate formation to determine PLC activity were carried out as described previously (24).

Preparation of DT40 Cell Lysates

DT40 cells (15 × 106) were lysed in 240 μl of ice-cold buffer A containing 20 mm Tris/HCl, pH 7.5, 2 mm EDTA, 2 μg/ml soybean trypsin inhibitor, 3 mm benzamidine, 0.1 mm PMSF, 1 μm pepstatin, 1 μm leupeptin, and 1 μg/ml aprotinin by forcing the suspension eight times through a 0.45 × 25-mm needle attached to a disposable syringe. Nuclei and unbroken cells were removed from the cell lysate by centrifugation at 300 × g for 10 min at 4 °C. Fifty μg of cell lysate protein were subjected to SDS-PAGE, and immunoblotting was performed using antibodies reactive against the c-Myc epitope and β-actin.

Expression of Proteins in Sf9 Cells and Measurement of PLC Activity in Vitro

The production of crude preparations of isoprenylated, recombinant human Rac2 and chicken PLCγ2 in Sf9 insect cells and the determination of PLC activity in vitro were carried out as described previously (24).

Determination of the Intracellular Calcium Concentration

To monitor the changes in [Ca2+]i in individual DT40 B cells in real time, 3 × 105 DT40 B cells per channel were seeded into six channel microscopy slides (uncoated ibidi® μ-slide VI0.4, ibidi, catalogue no. 80601), which had been coated with poly-l-lysine (0.01% (w/v)), and incubated for 45 min at 37 °C and 10% CO2 to allow for adhesion to the substrate. Next, unbound cells were flushed out with the culture medium, and the adherent cells were loaded with 2 μm fluo-4 AM, 0.02% (v/v) Pluronic® F-127, by incubation at room temperature for 30 min in RPMI 1640 medium containing supplements. Subsequently, cells were washed twice with buffer B (20 mm HEPES/NaOH, pH 7.4, 143 mm NaCl, 6 mm KCl, 1 mm MgSO4, 5.6 mm glucose). To discriminate between mobilization of Ca2+ from intra- and extracellular sources, BCR stimulation was either performed in buffer B or in buffer B supplemented with 1 mm CaCl2. The experiments were performed at room temperature.

Spinning Disc Confocal Fluorescence Microscopy

Fluorescence images were taken with the acquisition software Andor iQ 1.6 using a spinning disk confocal microscope assembled in our laboratory from individual components as described earlier (30). Briefly, its main components are a CSU10 scan head (Yokogawa, Tokyo, Japan), an inverted microscope (Axio Observer, Zeiss) with oil immersion objective (UPlanSApo 60×/1.35 NA, Olympus), environmental control system (PeCon), an image splitter unit (OptoSplit II, Cairn Research), and an EMCCD camera (DV-887, Andor). Diode-pumped solid-state lasers were employed for fluorescence excitation at 473 nm (HB-Laser LSR473-100) and 532 nm (HB-Laser LSR532-250). Time-lapse confocal fluorescence images consisting of 512 × 512 pixels were acquired in a field of 1024 × 1024 μm2 with a depth resolution of ∼2 μm, time resolution of 1.5 frames/s, and exposure time for each frame of 400 ms. The 473 nm excitation laser power measured at the optical fiber output to the spinning disc module was set to ∼4 milliwatts throughout the study.

Quantitative Analysis of the Ca2+ Responses

The fluorescence intensity fluctuations of the Ca2+ -sensitive dye fluo-4 were baseline-corrected and normalized according to the maximal intensity in the presence of ionomycin to account for the differences in fluorescent dye loading between individual cells (Fig. 4A and supplemental Video S1). Then, baseline-corrected and normalized Ca2+ response traces were subjected to further analysis using algorithms developed in-house and implemented in MATLAB (MathWorks). A Ca2+ response peak was defined by a transient positive divergence from the baseline with a minimum of 10% in intensity of the maximum observed in the presence of ionomycin. The number of responding cells was calculated as the number of traces with at least one Ca2+ response peak and expressed as the percentage of the total number of traces in each experiment. Latency was defined as a mean time lapse between the time of ligand addition and the time corresponding to the maximum of the first Ca2+ response peak. Cells that did not show any detectable Ca2+ spike during the initial observation time for the release of Ca2+ from internal stores, 365.1 s, were considered to display maximal latency equal to the time of observation. The integrated intensity was calculated as the mean area under the curve of all individual traces corresponding to single cells. The peak frequency was calculated for cells showing discrete Ca2+ spikes with baseline resolution, i.e. after addition of anti-IgM at concentrations of 4, 40, and 400 ng/ml, and expressed in millihertz (10−3 Hz). The same cells were used to calculate the peak amplitude as the average intensity of the Ca2+ peaks in percentage of the ionomycin maximum. The integrated intensities, peak frequencies, and peak amplitudes of nonresponding cells were set to zero.

FIGURE 4.

FIGURE 4.

Functional reconstitution of B cell receptor-mediated intracellular Ca2+ release and extracellular Ca2+ entry into PLCγ2−/− DT40 B cells. A, normalization of BCR-mediated oscillatory changes in cytosolic Ca2+ in PLCγ2−/− DT40 cells stably expressing wild-type PLCγ2. Left panel, the mean fluorescence intensities monitored inside the three cells selected in supplemental Video S1 and marked in cyan, magenta, and blue were measured on a single-cell level using spinning disc confocal fluorescence microscopy. The right panel shows the same data after correction for basal fluorescence intensity and normalization according to the maximal intensity in the presence of ionomycin. This treatment corrects variations in fluorescent dye loading between cells. The arrowheads indicate the addition of the various extracellular reagents. B, qualitative analysis. Unmodified DT40 cells (panel a; PLCγ2+/+), PLCγ2−/− cells (panel b), PLCγ2−/− cells stably expressing wild-type (panel c), or F897Q mutant PLCγ2 (panel d) (60 cells each) were treated as indicated by arrowheads in the following sequences: 40 ng/ml anti-IgM; no Ca2+ → 40 ng/ml anti-IgM; 1 mm Ca2+ → 4 μm ionomycin; 1 mm Ca2+ in panels a, c, and d, and 400 ng/ml anti-IgM; no Ca2+ → 400 ng/ml anti-IgM; 1 mm Ca2+ → 4 ng/ml trypsin; 1 mm Ca2+ → 4 μm ionomycin; 1 mm Ca2+ in panel b. Insets, snapshots from the corresponding time-lapse image series. C, expression of wild-type PLCγ2 in PLCγ2−/− DT40 cells restores the quantitative features of B cell-receptor-mediated increases in cytosolic Ca2+ to the phenotype observed in wild-type DT40 cells. Top panels, unmodified DT40 cells (PLCγ2+/+) or PLCγ2−/− DT40 cells stably expressing wild-type PLCγ2 (PLCγ2−/− + WT) were sequentially treated as in Fig. 4B, panel a, except that anti-IgM was used at 400 ng/ml. Bottom panels, the intensity traces obtained for both groups of cells were quantitatively analyzed as indicated for the percentage of responding cells, integrated peak intensity, peak frequency, and peak amplitude. A total number of 213 and 196 cells, respectively, was analyzed in three independent experiments (n = 3). D, PLCγ2−/− DT40 cells stably expressing wild-type or F897Q mutant PLCγ2 show similar Ca2+ responses to thapsigargin. PLCγ2−/− cells stably expressing wild-type (left panel) or F897Q mutant PLCγ2 (right panel) (78 cells each) were treated at the times indicated by the arrowheads in the following sequence: 100 nm thapsigargin; no Ca2+ → 100 nm thapsigargin; 1 mm Ca2+ → 4 μm ionomycin; 1 mm Ca2+.

Analysis of Nuclear NFAT1c Translocation

To study the BCR-mediated, Ca2+-dependent translocation of the transcription factor NFAT into the nucleus, 30 μg of cDNA encoding NFAT1c-td-RFP611 was transferred into 107 PLCγ2−/− DT40 B cells per cuvette that had or had not been reconstituted with either WT or F897Q mutant PLCγ2 by nucleofection (Amaxa® Nucleofector® Technology, Cell Line Nucleofector® kit T, program B-023). Forty eight h after nucleofection, 6 × 105 DT40 B cells per channel were allowed to adhere to poly-l-lysine-coated ibidi® 6-channel microscopy slides. After two washing steps with buffer B, the cells were treated for 60 min with either 40 μg/ml of anti-IgM or 2 μm ionomycin in buffer B containing 1 mm Ca2+. The translocation of NFAT1c-td-RFP611 was analyzed at room temperature on a single cell level by spinning disc confocal microscopy over a time period of 60 min using a 532-nm excitation laser for fluorescence imaging.

Indirect Immunofluorescence Staining

For immunofluorescence staining, 3 × 105 DT40 cells per channel were allowed to adhere to tissue culture-treated 6-channel microscopy slides (ibiTreat, ibidi® μ-slide VI0.4, ibidi, catalogue no. 80606,) as described above. The cells were then fixed by incubation for 15 min in 4% (w/v) paraformaldehyde in phosphate-buffered saline (PBS), pH 7.4. After washing the cells twice with PBS, the nonreacted aldehyde groups were quenched by treatment for 10 min with 50 mm NH4Cl in PBS. The cells were then washed four times with PBS, and nonspecific protein-binding sites in the channels were blocked by incubation for 1 h with buffer C (PBS, pH 7.4, 5% (v/v) FCS, 0.02% (w/v) sodium azide, 0.01% (w/v) saponin, 0.1% (v/v) Triton X-100). The cells were then incubated for 45 min with buffer C containing the primary antibody reactive against c-Myc epitope (1:2000). After four washing steps with buffer D (PBS, pH 7.4, 0.01% (w/v) saponin, 0.1% (v/v) Triton X-100), the cells were incubated for 45 min with buffer C containing the secondary goat anti-mouse IgG antibody conjugated with Alexa Fluor 488 (1:1000). After four further washing steps with buffer D, the expression of PLCγ2 protein was analyzed on a single cell level by spinning disk confocal microscopy employing a 473-nm excitation laser for fluorescence and a halogen lamp for transmission light imaging. All steps were done at room temperature.

Analysis of PLCγ2 Phosphorylation by Immunoprecipitation

H2O2 enhances tyrosine phosphorylation of PLCγ2 mediated by the tyrosine kinases Syk and Btk (31). Aliquots (12 × 106 each) of PLCγ2- or PLCγ2F897Q-expressing DT40 cells were treated for 5 min with 2 mm H2O2 in 400 μl of PBS, pH 7.4, at 37 °C in a water bath. After addition of 4 ml of ice-cold PBS and centrifugation at 300 × g for 5 min, the pellet was resuspended in 400 μl of ice-cold lysis buffer (10 mm Tris/HCl, pH 7.4, 150 mm NaCl, 1 mm EDTA, 1 mm EGTA, 1% (v/v) Triton X-100, 0.5% (v/v) Nonidet P-40, 0.2 mm Na3VO4, phosphatase inhibitor mixture 2 (1:100; v/v) (Sigma), 0.1 mm PMSF, 10 μm leupeptin, 2 μm pepstatin, 2 μg/ml soybean trypsin inhibitor, 1 μg/ml aprotinin, 3 mm benzamidine, 1 mm DTT, 16 μg/ml 1-chloro-3-tosylamido-7-amino-2-heptanone, and 16 μg/ml l-1-tosylamido-2-phenylethyl chloromethyl ketone), and the cells were lysed by incubation for 30 min at 4 °C. After centrifugation at 100,000 × g for 1 h at 4 °C, aliquots of the supernatants (1 mg of protein each) were added to anti-c-Myc antibody-protein A complexes, which had been prepared by preincubation of anti-c-Myc antibody (1:1000 (v/v) in 500 μl of PBS) with 30 μl of protein A-agarose beads (50% (v/v) slurry) overnight at 4 °C and three washes with lysis buffer. After incubation for 2 h at 4 °C, the beads were washed three times with lysis buffer and once with PBS and then subjected to SDS-PAGE and Western blotting using anti-phosphotyrosine or anti-c-Myc antibody (1:1000 (v/v) each).

FRAP Experiments

FRAP studies (32, 33) were conducted as described earlier (34). The experiments were performed 24–26 h post-transfection on COS-7 cells transfected with PLCγ2-GFP derivatives. All experiments were conducted at 22 °C, in Hanks' balanced salt solution supplemented with 20 mm HEPES, pH 7.2. The monitoring argon ion laser beam (488 nm, 1.2 microwatts; Innova 70C, Coherent) was focused through the microscope (AxioImager.D1, Carl Zeiss MicroImaging) to a Gaussian spot with a radius ω = 0.77 ± 0.03 μm (×63/1.4 NA oil-immersion objective) or 1.17 ± 0.05 μm (×40/1.2 NA water immersion objective). Experiments were conducted with each beam size (beam size analysis is described in Refs. 35, 36). The ratio between the illuminated areas (ω2(×40)/ω2(×63)) was 2.28 ± 0.17 (n = 59). After a brief measurement at the monitoring intensity, a 5-milliwatt pulse (4–6 ms or 10–20 ms for the ×63 and ×40 objectives, respectively) bleached 50–70% of the fluorescence in the spot. Fluorescence recovery was followed by the monitoring beam. The apparent characteristic fluorescence recovery time (τ) and the mobile fraction (Rf) were derived from the FRAP curves by nonlinear regression analysis, fitting to a lateral diffusion process with a single τ value (37). The significance of differences between τ values measured with the same beam size was evaluated by Student's t test. To compare ratio measurements (τ(×40)/τ(×63) and ω2(×40)/ω2(×63)), we employed bootstrap analysis, which is preferable for comparison between ratios (38), as described by us earlier (36), using 1000 bootstrap samples.

Miscellaneous

SDS-PAGE and immunoblotting were performed according to standard protocols using antibodies reactive against the c-Myc epitope for wild-type and mutant PLCγ2. Immunoreactive proteins were visualized using the ECL Western blotting detection system (GE Healthcare). All experiments were performed at least three times. Similar results and identical trends were obtained each time. Data from representative experiments are shown as means ± S.E. of triplicate determinations, if not stated otherwise in the figure legends. Unless stated otherwise, the significance of differences was assessed by using either the unpaired t test with two-tailed p values or repeated measures analysis of variance with Tukey's post test (Fig. 7B), both contained in GraphPad InStat®, version 3.10. Statistically significant effects are denoted by ***, p < 0.001; **, 0.001 < p < 0.01; and *, 0.01 < p < 0.05. Nonsignificant (ns) changes are denoted by ns, 0.05 < p. In Figs. 1, D and E, and 6, A–C, the data were fitted by nonlinear least squares curve fitting to three or four parameter dose-response equations using GraphPad Prism®, version 5.04. In certain cases, the global curve fitting procedure contained in Prism® was used to determine whether the best fit values of selected parameters differed between data sets. The simpler model was selected unless the extra sum of squares F-test had a p value of less than 0.05. In Fig. 6A, right panel, the data were fitted to the equation of a bell-shaped dose response curve provided by Prism®, with manual adjustments.

FIGURE 7.

FIGURE 7.

BCR-induced translocation of the transcription factor NFAT1c into the nucleus is dependent on a functional interaction between PLCγ2 and Rac. A, PLCγ2−/− cells or PLCγ2−/− cells stably expressing either wild-type or F897Q mutant PLCγ2 were transiently transfected with a vector encoding NFAT1c-td-RFP611. Adherent cells were treated with either anti-IgM (upper panels) or ionomycin (lower panels). Snapshots were taken of the same visual fields at the beginning of the experiment in the absence of ligand (−) and at the end of each experiment in the presence of the stimulus (+).Examples of visible NFAT1c translocation events are marked by arrowheads. B, quantitative analysis of the data shown. ns, not significant.

FIGURE 1.

FIGURE 1.

PLCγ2 mutant F897Q is resistant to stimulation by activated Rac2 in intact cells and in a cell-free system. A, changes in the conformations of the PLCγ2-interacting surface of Rac2 upon activation of Rac2 with GTPγS (gray) and of the PLCγ2 spPH residue Phe-897 upon interaction with activated Rac2. The hydrophobic Rac2 residues that change their positions upon Rac2 activation and interact with PLCγ2 spPH are shown in white (GDP-liganded Rac2) versus red (GTPγS-liganded Rac2). Phe-897 displays flexibility in the known structures of the free spPH domain and adopts two conformations (I and II, pale yellow) (25). This flexibility is stabilized in the complex with activated Rac2 (wheat). The side chain of the polar residue Gln in the F897Q mutant (light blue) is likely to collide with the hydrophobic side chain of Rac2 Leu-67. The structures correspond to database entries 2W2T, 2W2V, 2W2W, and 2W2X (25). The position of Gln-897 was predicted in Swiss-Model. B, COS-7 cells were cotransfected with 1 μg/well of vector encoding either WT PLCγ2 (WT) or its F897Q mutant (F897Q) together with either empty vector (Mock) or with 10 ng/well of vector encoding Rac2 (Rac2) or its constitutively active mutant Rac2G12V (Rac2G12V). Inositol phosphate formation was measured as described under “Experimental Procedures” (upper panel). Cells from one well each were analyzed by SDS-PAGE and immunoblotting (lower panel). C, left panel, soluble fractions of Sf9 cells infected with baculoviruses encoding β-galactosidase (Control), WT PLCγ2 (WT), or its Phe-897 mutant (F897Q) were incubated with phospholipid vesicles containing [3H]PtdInsP2. Right panel, soluble fractions of baculovirus-infected Sf9 cells were adjusted to contain similar basal PLC activity according to the left panel. Aliquots (10 μl) of these samples were reconstituted with aliquots of detergent extracts prepared from membranes of Sf9 cells infected with baculoviruses encoding either β-galactosidase or Rac2 and incubated in the presence of 100 μm GDP or GTPγS with phospholipid vesicles containing [3H]PtdInsP2. Inset, aliquots (10 μl) of the samples were subjected to SDS-PAGE and immunoblotting. Lane 1, control; lane 2, WT, lane 3, F897Q. D, wild-type and F897Q mutant PLCγ2 are similarly sensitive to activation by Ca2+. Aliquots of the soluble fractions of baculovirus-infected Sf9 cells were incubated at increasing concentrations of free Ca2+ with phospholipid vesicles containing [3H]PtdInsP2. For both wild-type and F897Q mutant PLCγ2, half-maximal and maximal stimulatory effects were observed at ∼140 nm and 10 μm free Ca2+. E, PLCγ2 variants ΔPCIWT and ΔPCIF897Q lacking the PCI peptide have similar constitutive activities but show a striking difference in their sensitivity to Rac2. Tyrosine phosphorylation-induced activation of PLCγ is mediated by removal of intramolecular autoinhibition caused by an octapeptide (726YRKMRLRY in human PLCγ2; PCI) within the C-terminal Src homology domain contained in the Src homology domain tandem between the two catalytic subdomains X and Y. Deletion of the PCI peptide causes activation of PLCγ2 (88). Upper left panel, COS-7 cells were transfected with increasing amounts of vector encoding ΔPCIWT or ΔPCIF897Q or with a single amount (1000 ng) of vector encoding either wild-type PLCγ2 or its F897Q mutant. Upper right panel, COS-7 cells were cotransfected with vectors encoding either of the ΔPCI variants together with empty vector (Mock) or with 50 ng/well of vector encoding Rac2 or Rac2G12V. Lower panel, expression of wild-type and mutant PLCγ2 isozymes in upper left panel. Cells from one well each were analyzed by SDS-PAGE and immunoblotting. F, wild-type and the F897Q mutant PLCγ2 do not differ in their ability to be tyrosine-phosphorylated by treatment of DT40 cells with H2O2. PLCγ2 phosphorylation in PLCγ2−/− DT40 cells stably transfected with wild-type or the F897Q mutant PLCγ2 was analyzed in the absence (−) or presence (+) of H2O2. PLCγ2 was immunoprecipitated from cell extracts using anti-c-Myc antibody, and Western blotting was performed with either anti-phosphotyrosine (pY) antibody (top panel) or anti-c-Myc antibody (bottom panel).

FIGURE 6.

FIGURE 6.

A, functional interaction of PLCγ2 with activated Rac enhances the proportion of cells responding to BCR ligation with an increase in [Ca2+]i. PLCγ2−/− stably expressing either wild-type or F897Q mutant PLCγ2 were treated in the absence (left panel) or presence (right panel) of 1 mm extracellular Ca2+ with medium supplemented with increasing concentrations of anti-IgM. The data correspond to the means ± S.E. of three independent measurements for each ligand concentration, each performed on 60–80 cells. The best fit values obtained for the EC50 values in A and the corresponding half-maximal proportional Ca2+ responses as well as the EC50 value estimated in the right panel are marked by dashed lines. B and C, functional interaction of PLCγ2 with activated Rac enhances the BCR-mediated increase in integrated Ca2+ fluorescence activity in the absence and in the presence of extracellular Ca2+. PLCγ2−/− stably reconstituted with either WT (left panels) or F897Q mutant of PLCγ2 (right panels) were treated in the absence (B) or presence (C) of 1 mm extracellular Ca2+ with increasing concentrations of anti-IgM. Data obtained for single cells are shown. Nonresponding cells are shown individually below the abscissae. The mean values were fit by nonlinear least squares curve fitting. The standard errors of the best fit values of the maximal intensities are indicated by shading. B, 202, 173, 195, 174, and 185 (WT) and 121, 186, 201, 192, and 196 (F897Q) single cells were analyzed with increasing concentrations of anti-IgM. C, these cell numbers were 201, 174, 195, 173, and 185 (WT) and 121, 187, 200, 191, and 196 (F897Q). D, latency (time between anti-IgM addition and first Ca2+ peak) was plotted as a function of ligand concentration in the absence of extracellular Ca2+. Cells that showed no Ca2+ response during the observation time of 365.1 s were considered to have maximal latency equal to the time of observation. Data are shown as mean ± S.E. of 121–202 values obtained by analysis of all individual traces (corresponding to single cells) from three independent experiments in each group for each ligand concentration. E, peak frequency (left panel) and peak amplitude (right panel) calculated for the cells treated with anti-IgM in concentrations of 4, 40, and 400 ng/ml in the absence of extracellular Ca2+.

Results

Point Mutation F897Q Renders the DT40 Cell PLCγ2 Orthologue Specifically Resistant to Stimulation by Activated Rac2

Mutational and structural analyses have shown that the stimulation of human PLCγ2 by activated Rac GTPases is due to an interaction of the Rac switch I and II regions with specific residues in the C-terminal half of the PLCγ2 spPH domain (24, 25). Fig. 1A illustrates the importance of the PLCγ2 residue Phe-897, contained within the C-terminal α helix of spPH, in this respect. Phe-897 forms the core of a hydrophobic pocket on PLCγ2 spPH, which interacts with several hydrophobic residues of the Rac2 switch I and II regions, such as Val-36 (I), Phe-37 (I), Trp-56 (II), Leu-67 (II), and Leu-70 (II). Rac2-PLCγ2 complex formation is thought to have two consequences as follows: (i) conversion of GTP-bound Rac2 from conformational state 1 to state 2. These states were initially described for H-Ras as having low versus high affinity, respectively, toward Ras effectors (39). (ii) Stabilization of the Phe-897 side chain was in one of the two conformational states observed in the crystal structures of free spPH (25). Replacement of Phe-897 in human PLCγ2 spPH by the corresponding human PLCγ1 residue glutamine is expected to reduce the hydrophobic momentum of the Rac2 binding pocket on PLCγ2 spPH and, possibly, interfere with the reorientation of the Leu-67 side chain upon complex formation (Fig. 1A). Consistent with this view, the F897Q substitution blocked activation of human PLCγ2 by constitutively active Rac2 and abolished binding of GTPγS-activated Rac2 to PLCγ2 spPH, while leaving the overall fold of PLCγ2 spPH unaffected (24, 25).

To minimize the differences between the cellular system to be functionally reconstituted and analyzed here and unmodified DT40 B cells, a cDNA encoding chicken PLCγ2 was produced using mRNA from DT40 cells as a template. The encoded protein shares 83% identical residues with human PLCγ2. Within the C-terminal helices of the two spPH domains, 14 of 17 residues, including Phe-897, are identical, and three are conserved. The experiments shown in Fig. 1, B and C, examined the consequences of the F897Q mutation on the ability of the DT40 PLCγ2 orthologue (henceforth referred to as PLCγ2) to interact with activated Rac2. Fig. 1B shows that constitutively active Rac2G12V, but not WT Rac2, caused a marked (∼19-fold) stimulation of PLCγ2 activity in COS-7 cells, but it was ineffective in control cells lacking PLCγ2 and in cells expressing the F897Q mutant. These observations were confirmed in a cell-free system (Fig. 1C). The F897Q mutation did not affect activation of PLCγ2 by calcium ions or by loss of autoinhibition (Fig. 1, D and E) and had no influence on the extent of PLCγ2 protein tyrosine phosphorylation in response to H2O2 (Fig. 1F).

To measure the effect of the F897Q mutation on the interactions of PLCγ2 with the plasma membrane in intact cells, FRAP experiments were performed on GFP-tagged WT and the F897Q mutant PLCγ2. PLCγ2-GFP fluorescence recovery occurred with a fluorescence recovery time (τ) of 0.079 s (Fig. 2A, left panel). Coexpression of constitutively active Rac2G12V enhanced the interaction of the enzyme with the plasma membrane, resulting in an ∼1.5-fold increase in τ of PLCγ2-GFP to 0.115 s (Fig. 2A, right panel). Interestingly, FRAP beam size analysis experiments showed that the WT enzyme and the variant F897Q did not differ in their modes of membrane interaction, as the fluorescence recovery of both PLCγ2-GFP and PLCγ2F897Q-GFP occurred by a mixture of binding to and dissociation from the membrane, referred to as exchange, and of stable association with the membrane, resulting in lateral diffusion. In the presence of Rac2G12V, the fluorescence recovery of WT PLCγ2-GFP was shifted toward lateral diffusion, as the ratio between the fluorescence recovery times with the two beam sizes (×40 and ×63 objectives) was 2.3 (Fig. 2, B and C). This ratio is indistinguishable from that expected for recovery by pure lateral diffusion (2.28 ± 0.17, the ratio between the areas illuminated by the two beam sizes employed). Consequently, in the presence of Rac2G12V, the interaction of PLCγ2-GFP with the plasma membrane is dominated by lateral diffusion, suggesting that the exchange rate becomes much slower than the lateral diffusion rate (i.e. a shift to stable membrane interactions). In marked contrast, Rac2G12V had no effect at all on the mode of membrane association of the mutant PLCγ2F897Q-GFP, which correlates well with our observations that Rac2G12V cannot interact with and fails to stimulate PLCγ2F897Q. We conclude that PLCγ2 is not compromised in its overall activity and in its mode of membrane interaction by the F897Q mutation, in contrast to its specific loss of regulation by activated Rac.

FIGURE 2.

FIGURE 2.

FRAP beam size analysis demonstrates that wild-type and F897Q mutant PLCγ2 display similar membrane interactions, but only that of wild-type PLCγ2 is augmented by Rac2G12V. A, COS-7 cells were cotransfected with a vector encoding GFP-tagged wild-type PLCγ2 (WT) together with either empty vector (left panel) or vector encoding Rac2G12V (WT + Rac2G12V; right panel). The typical FRAP curves shown were obtained using a ×63 objective. The solid lines represent the best fit obtained by nonlinear regression analysis. The best fit τ values are depicted in each panel. The mobile fractions (Rf) were above 0.93 in all cases and therefore are not shown. B and C, FRAP beam size analysis. Transfection was as in A, except that in some studies the GFP-tagged mutant (F897Q) replaced its wild-type counterpart (WT). Bars represent the means ± S.E. of 30–60 measurements. The studies employed ×40 and ×63 objectives, yielding a beam size ratio of 2.28 ± 0.17 (n = 59). Thus, this τ(×40)/τ(×63) ratio is expected for FRAP by lateral diffusion (C, upper dashed line). A τ ratio of 1 (C, lower dotted line) indicates recovery by exchange (35). In the absence of Rac2G12V, both the τ values (comparing values measured with the same beam size (B) and the τ(×40)/τ(×63) ratios (C) of WT and F897Q mutant PLCγ2 were similar (p > 0.4 in all cases; Student's t test), suggesting similar lateral diffusion in and exchange rate from the plasma membrane. Although coexpression of Rac2G12V had no significant effect on either the τ values or τ ratio of the F897Q mutant (p > 0.4), it significantly enhanced the membrane association of wild-type PLCγ2, reflected in a slower τ(×40) (**, p < 0.002) and in a significant increase of its τ ratio (*, p < 0.02; bootstrap analysis) from an intermediate value (characterizing recovery by a mixture of lateral diffusion and exchange) to 2.3, similar to the value expected for pure lateral diffusion.

Functional Reconstitution of Wild-type and F897Q Mutant PLCγ2 into PLCγ2−/− DT40 B Cells

Next, genetically PLCγ2-deficient DT40 cells were stably reconstituted with either isogenic WT PLCγ2 or the PLCγ2F897Q mutant such that the resultant cell clones were indistinguishable in terms of enzyme expression and subcellular distribution (Fig. 3). Spinning disc confocal fluorescence microscopy was then used to determine the regulatory influence of Rac on the kinetic parameters of BCR-mediated changes in [Ca2+]i at the level of individual DT40 B cells. Cells were loaded with the fluorescent Ca2+ indicator fluo-4, and authentic PLCγ2+/+ DT40 cells were compared with PLCγ2−/− cells, before and after stable reconstitution of the latter with either WT or F897Q mutant PLCγ2, for their ability to respond to BCR ligation with increases in [Ca2+]i. In accordance with earlier studies (40), cells were first treated with 40 ng/ml anti-IgM in the absence of extracellular Ca2+ to measure the BCR-mediated Ca2+ release from intracellular stores. After some time, the effect of the same concentration of anti-IgM was determined in the presence of 1 mm extracellular Ca2+ to also allow for the entry of extracellular Ca2+ into the cells. Finally, the Ca2+ ionophore ionomycin was added to normalize the fluorescence intensities obtained for the individual cells (Fig. 4A). The latter values were subsequently used to normalize the individual intensity time traces. Sixty cells from each cell type were analyzed individually in each experiment (Fig. 4B). Each of the curves shown represents the fluorescence intensity time trace of a single cell in the observation area. Fig. 4B, upper left panel, shows that addition of anti-IgM to WT DT40 caused oscillations of [Ca2+]i in most but not all cells. In responding cells, the oscillations commenced after a lag time of ≥∼2 min. Although some cells displayed a less oscillatory and more monophasic increase in [Ca2+]i upon addition of extracellular Ca2+ accompanied by a loss of spiking, most responding cells showed a relatively homogeneous oscillatory behavior in the absence and presence of extracellular Ca2+. In PLCγ2−/− cells, by contrast, none of the cells responded to addition of anti-IgM (400 ng/ml), regardless of whether Ca2+ was absent from or present in the incubation medium (Fig. 4B, upper right panel). Addition of 4 ng/ml trypsin to these cells in the presence of extracellular Ca2+ caused the appearance of a single [Ca2+]i peak, presumably by activation of G-protein-coupled PAR2 receptors endogenously present in DT40 cells, followed by activation of endogenous PLCβ (41). In this case, almost all cells responded, and the lag time was much shorter (∼10 s). Fig. 4B, lower panels, shows that although both PLCγ2 and PLCγ2F897Q were able to reconstitute oscillatory Ca2+ responses to PLCγ2−/− DT40 cells following BCR ligation, substantial quantitative differences were apparent already at the level of visual assessment of the two sets of time traces. Specifically, the proportion of cells responding to anti-IgM as well as both the peak amplitudes and the frequencies of the spikes were considerably lower, and the mean latencies of the overall responses were distinctly longer for cells expressing PLCγ2F897Q rather than WT PLCγ2. A more detailed, quantitative characterization of this difference will be presented below in Figs. 5 and 6. A quantitative comparison of the [Ca2+]i transients observed in PLCγ2+/+ DT40 cells versus PLCγ2−/− cells reconstituted with WT PLCγ2 in response to 400 ng/ml anti-IgM is shown in Fig. 4C (upper panels). There were no significant differences between the two cell types in the proportions of responding cells, integrated peak intensities, peak frequencies, and amplitudes (Fig. 4C, lower panels). Thus, the PLCγ2-deficient cells reconstituted with WT PLCγ2 bear a close resemblance to their native counterparts. Fig. 4D shows that PLCγ2−/− cells reconstituted with WT versus F897Q mutant PLCγ2 do not appreciably differ in [Ca2+]i responses triggered by addition of thapsigargin, an inhibitor of the sarco-/endoplasmic reticulum Ca2+-ATPase (40, 41). Thus, the F897Q mutation and, by extension, interaction of PLCγ2 with activated Rac have no effect on BCR-independent [Ca2+]i mobilization in DT40 B cells.

FIGURE 3.

FIGURE 3.

Stable reconstitution of PLCγ2−/− DT40 B cells with wild-type versus F897Q mutant PLCγ2. PLCγ2-deficient DT40 cells (89) were stably reconstituted with either isogenic wild-type or F897Q mutant PLCγ2. A, aliquots of the lysates (50 μg of protein) of native DT40 cells (PLCγ2+/+), PLCγ2-deficient DT40 cells (PLCγ2−/−), and from three independent clones PLCγ2−/− DT40 cells stably expressing similar quantities of either wild-type (A–C) or F897Q mutant PLCγ2 (a–c) were subjected to SDS-PAGE and immunoblotting (upper panel). The same membrane was subsequently probed with an anti-β-actin antibody to control for equal loading of samples (lower panels). All six clones were used for experimentation in this study, with no differences detected between clones A–C and a–c, respectively. B, cells from the clones A and c were analyzed by indirect fluorescence staining (left panels). Right panels, corresponding phase contrast images. C, mean fluorescence intensities of the three images each, as shown in B, were corrected for background staining. Similar results were obtained for other pairs of clones.

FIGURE 5.

FIGURE 5.

Rac-insensitive mutant PLCγ2F897Q is quantitatively impaired in its ability to restore B cell receptor-mediated Ca2+ flux in PLCγ2−/− DT40 cells. PLCγ2−/− cells stably expressing wild-type (left column) or F897Q mutant PLCγ2 protein (right column) were treated with increasing concentrations of anti-IgM. The treatment was performed as indicated by the arrowheads in the following sequence: anti-IgM; no Ca2+ → anti-IgM; 1 mm Ca2+ → 4 μm ionomycin; 1 mm Ca2+. Forty nine to 60 cells were analyzed in each single experiment.

Concentration Dependence on Anti-IgM of [Ca2+]i Response Impairment in DT40 B Cells Expressing Wild-type Versus Rac-insensitive F897Q Mutant PLCγ2

To determine the influence of the Rac-PLCγ2 interaction on [Ca2+]i changes in DT40 B cells in response to increasing extents of BCR ligation, PLCγ2−/− DT40 cells stably reconstituted with either WT or F897Q mutant PLCγ2 were treated with increasing concentrations of anti-IgM in the absence of extracellular Ca2+, followed by the same concentration of anti-IgM in the presence of 1 mm extracellular Ca2+. Cells were treated with 4 μm ionomycin at the end of each experiment for baseline correction and normalization of the [Ca2+]i responses in single cells. Fig. 5, left panels, shows that oscillatory [Ca2+]i responses developed in cells reconstituted with WT PLCγ2 at anti-IgM concentrations between 4 and 400 ng/ml. At 40–400 ng/ml anti-IgM, the responses were similar in both their frequencies and intensities, in the absence or presence of extracellular Ca2+. At 4 μg/ml anti-IgM (Fig. 5, next-to-lowest left panel), individual Ca2+ oscillations appeared to coalesce into single major asymmetric peaks in many traces. These peaks reached their maximum intensities within seconds after anti-IgM addition in the absence of extracellular Ca2+ to gradually decline to base levels over the next 6 min. Addition of extracellular Ca2+ to anti-IgM at this time point led to a second, more symmetric [Ca2+]i wave, reaching a maximum after about 1.5 min and declining with similar kinetics thereafter. At still higher concentrations of anti-IgM, 40 μg/ml, the pattern was qualitatively similar but somewhat reduced in quantitative terms, both in the absence and presence of extracellular Ca2+. In cells expressing the Rac-insensitive mutant F897Q of PLCγ2 (Fig. 5, right panels), there was a striking loss of the [Ca2+]i responses, in particular at low concentrations of anti-IgM, e.g. 40 ng/ml. In each case, this effect was clearly evident both with and without extracellular Ca2+. At high (4 and 40 μg/ml) concentrations of anti-IgM, there also was a loss of coalescence of individual oscillations in responding cells and a decrease in the overall duration of the [Ca2+]i response to BCR ligation, in particular in the absence of extracellular Ca2+.

The results of further quantitative analyses of the differences between the [Ca2+]i responses of cells expressing WT versus F897Q PLCγ2 are shown in Fig. 6. Thus, in the absence of extracellular Ca2+, there was a marked reduction in the anti-IgM sensitivity of the latter cells by an order of magnitude, but no change in the maximal extent, when the proportion of responding cells was used as a response parameter (Fig. 6A, left panel). In the presence of extracellular Ca2+, the major difference was a distinct loss of the proportion of responding cells, which was similar (∼55%) at low and intermediate anti-IgM concentrations, 4–400 ng/ml, and even more striking (∼80%) at high anti-IgM concentrations (Fig. 6A, right panel). There was no apparent change in the sensitivity of the cells to anti-IgM. Integration of the fluorescence intensities in single cells over time and evaluation of their concentration dependence on anti-IgM at the level of the means showed a reduction of the maximal mean intensity in cells expressing the F897Q mutant by 64% in the absence of extracellular Ca2+ (Fig. 6B). In its presence, this loss amounted to 82% (Fig. 6C).

Fig. 6D shows that resistance of PLCγ2 to regulation by Rac also resulted in a longer latency of the [Ca2+]i response to addition of anti-IgM. This became evident at low anti-IgM concentrations (40 ng/ml) and more prominent at all higher concentrations. [Ca2+]i peak frequencies and amplitudes were only analyzed for the three lowest ligand concentrations, 4–400 ng/ml, and in the absence of extracellular Ca2+, mostly because of peak coalescence at higher anti-IgM concentrations. By assigning a peak frequency of zero to nonresponding cells, we determined a reduction of this parameter by ∼86 and ∼55% in cells expressing the F897Q mutant rather than WT PLCγ2 (Fig. 6E, left panel). Likewise, decreases of peak amplitudes amounting to ∼84 and ∼58% were observed for the cells harboring the mutant at 40 and 400 ng/ml anti-IgM, respectively (Fig. 6E, right panel). Collectively, these results indicate that the regulation of PLCγ2 by Rac is not an absolute requirement for BCR-mediated Ca2+ release at most BCR ligand concentrations tested herein, both in the absence and presence of extracellular Ca2+. However, although the qualitative patterns of [Ca2+]i responses to BCR ligation are similar in the absence and presence of PLCγ2 regulation by Rac, there are striking quantitative differences. These are readily evident at the level of the proportion of responding cells and their sensitivity to anti-IgM, the latency of the response after addition of anti-IgM, as well as both the peak frequency and amplitude of the oscillatory [Ca2+]i responses.

Dependence of BCR-mediated Nuclear Translocation of the Transcription Factor NFAT1c on Functional Rac-PLCγ2 Interaction

Within minutes after BCR-mediated B cell activation, several transcription factors, including the Ca2+-dependent transcription factor NFAT1c, are translocated into the nucleus to induce transcription of regulatory genes involved in B cell-fate decisions (6). To determine the role of a functional Rac-PLCγ2 interaction in this response, a C-terminally truncated NFAT1c construct consisting of the N-terminal transactivation and the calcineurin-binding domain (amino acids 1–400) fused to the red fluorescent protein td-RFP611 was introduced into PLCγ2−/− DT40 B cells and PLCγ2−/− DT40 B cells stably expressing either WT or F897Q mutant PLCγ2, and its nuclear translocation following BCR ligation was analyzed by spinning disk fluorescence microscopy (Fig. 7). Treatment of cells with ionomycin, resulting in PLCγ2-independent nuclear translocation of NFAT1c-td-RFP611 in almost all transfected cells, was used as a positive control (Fig. 7, A, lower panels, and B, right panel). In PLCγ2-deficient cells, there was little, if any, nuclear translocation of the fluorescent reporter protein in response to anti-IgM (Fig. 7A, upper panels), consistent with the concept that this event is absolutely dependent on PLCγ2-induced Ca2+ mobilization. In cells expressing WT PLCγ2, NFAT1c-td-RFP611 translocated into the nuclei in about 80% of the cells (Fig. 7B, left panel). Importantly, only 16% of the cells expressing the Rac-insensitive PLCγ2 mutant F897Q displayed nuclear NFAT1c-td-RFP611, such that uncoupling of PLCγ2 from Rac caused an 80% decrease of a cellular response directly related to altered gene transcription and, by extension, cell fate decisions in B cells.

Discussion

This study shows that specific interference with a functional Rac-PLCγ2 interaction in intact DT40 B cells precipitates several clear alterations in BCR-mediated Ca2+ signaling. First, the sensitivity of Ca2+ release from internal stores to the BCR ligand anti-IgM is reduced by an order of magnitude. Second, the extent of the Ca2+ signal in the presence of extracellular Ca2+, also involving entry of extracellular Ca2+ into the cells, is greatly diminished. Third, Ca2+-regulated, NFAT-mediated transcriptional regulation is largely decreased.

Several lines of evidence have already suggested that PLCγ2 and Rac are both activated by BCR ligation, although a direct interaction of the two signaling molecules has received relatively little attention (6, 26, 42, 43). Thus, several elements of the canonical B cell receptor signaling cascade, e.g. Syk (44), Btk (45), and BLNK (46, 47), are known to physically interact with and activate the Rac activator Vav. BCR cross-linking caused activation of both Rac1 and Rac2 within minutes (48). Rac cooperated with PLCγ2 in BCR-mediated activation of c-Jun N-terminal kinase, p38 mitogen-activated protein kinase, SRF, and NFAT (49, 50). Dominant negative Rac1 suppressed these cooperative relationships. These results were interpreted to suggest that the signaling pathways controlling the activities of Rac and PLCγ2 are activated by BCR ligation in parallel to converge at points distal to enhanced formation of InsP3 and DAG. Btk, BLNK, Vav, and PLCγ2 form highly coordinated microsignalosomes in a process dependent on Syk and Lyn (51). Formation of these complexes is important for amplification of signaling and, hence, for appropriate B cell activation, in particular at low antigen concentrations. The results presented here strongly suggest that signal amplification is caused within or in the vicinity of these sites by convergence of signals emanating from activated BCR through a direct interaction of activated Rac with PLCγ2.

Calcium oscillations arise from a complex interplay between PLC-mediated generation of InsP3 and DAG, InsP3 receptor-mediated release of intracellular Ca2+, and entry of extracellular Ca2+ mediated by channels that are either store-operated, activated by DAG, or subject to other regulatory controls (52). These incremental changes may be rapidly overturned by InsP3 metabolism and Ca2+ efflux from the cytosol (6, 53). All three InsP3 receptors are present in DT40 B cells and display differential sensitivities to InsP3 in these cells (54). Expression of only one of the three in InsP3 receptor-deficient cells resulted in very distinct oscillatory responses to similar degrees of BCR activation. This finding suggests that InsP3 receptors are a major regulatory site of Ca2+ oscillations in these cells. In addition, feedback mechanisms may exist in B cells giving rise to spatiotemporal dynamics of the levels of InsP3 (55) and, by extension, of DAG. For example, the transient receptor potential cation channel type 3-mediated increases in [Ca2+]i were shown in B cells to promote translocation of PLCγ2 to the plasma membrane and further activation of the enzyme, by a process involving physical interaction with TRPC3 cation channels (56). Interestingly, some aspects of the latter interaction appeared to be independent of the phospholipase C activity of PLCγ2 (40, 56). Selective and direct inhibition of TRPC3 channels by pyrazole-3 eliminated the Ca2+ influx-dependent PLCγ2 plasma membrane translocation and the late oscillatory phase of the BCR-induced Ca2+ response (57). The same TRPC3 inhibitor also suppressed Rac1 activation without affecting total Rac1 protein abundance in cardiomyocytes (58), whereas activated Rac1 enhanced the rapid vesicular translocation and membrane insertion of another transient receptor potential cation channel, TRPC5 (59). Thus, the interaction of PLCγ2 with Rac may influence BCR-mediated Ca2+ oscillations in several ways.

Previous studies have shown that the interaction of PLCγ2 with Rac2G12V markedly enhances its enzymatic activity in vitro and in intact cells and coincides with a translocation of PLCγ2 from the soluble to the particulate fraction of intact cells (23). Here, we demonstrate that binding of activated Rac to WT PLCγ2 also changes the mode of plasma membrane association of the latter from a mixture of exchange and lateral diffusion to almost pure lateral diffusion (Fig. 2). This suggests that Rac-PLCγ2 interaction reduces the exchange rate between membrane-associated and cytoplasmic WT PLCγ2 such that it becomes negligible relative to its lateral diffusion rate, demonstrating a stronger membrane association. Hence, in intact cells, Rac-activated WT PLCγ2 mainly travels along the plasma membrane, allowing for a spatio-temporal pattern of encounter with both its lipid substrate and its protein interaction partners, e.g. TRPC3, that is different from that of its unliganded or Rac-resistant counterparts.

The dependence of the Ca2+ oscillations in cells expressing WT PLCγ2 on low to intermediate concentrations of anti-IgM (≤4 μg/ml) suggests that signal transduction occurs mostly through frequency modulation, although there is also a clear increase in the amplitude with increasing BCR ligation. It is also evident that, in this concentration range of anti-IgM, the spiking behavior of the cells is largely independent of the presence of extracellular Ca2+. The relative independence of oscillation frequency on external Ca2+ suggests that DT40 B cells are highly efficient at recycling their internal Ca2+ (53). In aggregate, these findings indicate that a minimal model based on reversible desensitization of InsP3 receptors may suffice to explain the oscillatory pattern observed at low to intermediate anti-IgM concentrations (53, 60). According to the minimal model, [Ca2+]i oscillations are driven by a coupled process of Ca2+-induced activation and obligatory intrinsic inactivation of the Ca2+-sensitized state of InsP3 receptors. Ca2+ spikes are initiated by the Ca2+-mediated conversion of low affinity InsP3 receptors from their low to the high affinity type (60). In this paradigm, the decrease in latency and the increase in oscillation frequency observed in Fig. 6, D and E, would be due to the decrease in the time required to generate a sufficient Ca2+ trigger signal to initiate the first or next Ca2+ release spike, respectively. Enhanced and accelerated translocation of PLCγ2 to the plasma membrane containing its phospholipid substrate by activated Rac would allow for a decrease in the time necessary for reaching this threshold level, such that considerably lower levels of activated BCR are required to elicit a given [Ca2+]i response. Of note, enhanced association of PLCγ2 with the plasma membrane following interaction with activated Rac is indeed observed in the FRAP beam size analysis experiments (Fig. 2), and the absence of this effect on the PLCγ2 mutant defective in Rac interactions (F897Q) is in accord with the much lower quantitative effect of anti-IgM in cells expressing this mutant on the Ca2+ oscillations (Fig. 6E). In addition to temporal changes of PLCγ2 activation by Rac, spatial changes may come into play. Given that the Ca2+ conductances are similar for the three InsP3-R subtypes (61), the increase in amplitude observed with increasing anti-IgM concentrations may be due to expression of the receptors at different cellular levels in DT40 B cells, with higher abundance of subtypes with lower InsP3 sensitivity (InsP3-R1 (4.7 μm); InsP3-R2 (0.35 μm); InsP3-R3 (18.6 μm); EC50 values for InsP3-mediated Ca2+ release in permeabilized cells expressing a single InsP3-R subtype in parentheses (54)).

When extracellular Ca2+ is provided to cells incubated without Ca2+ for several minutes at high concentrations of anti-IgM, the main effect of a loss of Rac regulation by PLCγ2 is a marked reduction in the number of responding cells, which cannot be recuperated by increasing the concentration of anti-IgM. Several scenarios may explain this observation: (i) the interaction of PLCγ2 with activated Rac could strengthen the productive interaction of PLCγ2 with plasma membrane channels such as TRPC3 allowing entry of extracellular Ca2+, either by altered temporal or spatial interaction of PLCγ2 with the membrane (cf. Fig. 2). If TRPC3 channels were involved in Rac activation in B cells, as they appear to be in cardiomyocytes (see above), this would further reinforce this process. (ii) Cells could be desensitized to BCR-mediated Ca2+ signaling with continued anti-IgM exposure. They can become less sensitive to this course of action if the Rac-PLCγ2 interaction is intact. Both homologous and heterologous desensitization of B cell membrane-immunoglobulin-mediated Ca2+ mobilization have long been known to occur within minutes of anti-immunoglobulin exposure (62). Interestingly, anti-IgM-treated B cells are hyper-responsive to AlF4 and mastoparan (63). Although the two reagents are known as activators of heterotrimeric G proteins, they also appear to activate Rho GTPases, including Rac (64, 65). If this was to occur in B cells, the results presented here and by Cambier et al. (63) may indicate that anti-IgM-mediated B cell activation affects a function(s) of the macromolecular complex involving PLCγ2, without impinging upon the activation of the enzyme by Rac, and that the complex is protected, at least in part, when the Rac-PLCγ2 interaction is intact. The markedly diminished effect of high concentrations of anti-IgM on [Ca2+]i observed in cells lacking the functional Rac-PLCγ2 interaction does not appear to require exposure of B cells to anti-IgM in the absence of extracellular Ca2+, because the inhibitory effect was also evident at the level of nuclear NFAT1c-td-RFP611 translocation, which was assayed in the continued presence of extracellular Ca2+ (Fig. 7).

Under these conditions, the insensitivity of PLCγ2 to activated Rac led to a marked reduction in BCR-mediated nuclear translocation of NFAT1c-td-RFP611 (Fig. 7), strongly suggesting that the changes observed at the level of [Ca2+]i are effectively transduced to the transcriptional level. In B lymphocytes, the activity of the Ca2+-regulated transcriptional regulators c-Jun N-terminal kinase (JNK), NFκB, and NFAT are differentially regulated by the amplitude and duration of oscillatory Ca2+ signals (66). NF-κB and JNK are selectively activated by a large transient [Ca2+]i rise, whereas NFAT is activated by a low, sustained Ca2+ plateau. These results and findings obtained in T lymphocytes (67) and basophilic leukemia cells (68) are consistent with the notion that nuclear translocation of NFAT functions as a working memory of Ca2+ signals by decoding Ca2+ oscillations (69). In general, this process appears to be more cost-effective than translation of continuous Ca2+ signals. In this study, nuclear translocation and, hence, activation of NFAT1c-td-RFP611 was studied at 40 μg/ml anti-IgM, allowing for similar numbers of responders (∼92%) when cells expressing either WT or F897Q mutant PLCγ2 were analyzed for BCR-mediated Ca2+ release from internal stores (Fig. 6A), with an overall reduction of the integrated fluorescence intensity in the latter cells by 64% (Fig. 6B). In the presence of extracellular Ca2+, both the proportion of responding cells and the mean integrated single cell fluorescence intensity were reduced by more than 80% (Fig. 6, A and C). Because NFAT activation is highly dependent on both InsP3-induced intracellular Ca2+ release and store-operated Ca2+ entry mediated by CRAC channels (50, 70), it is likely that reductions of both responses participate in reducing nuclear NFAT translocation. Furthermore, analysis of the Ca2+ oscillatory behavior in the absence of extracellular Ca2+ at 40 μg/ml anti-IgM revealed that the majority of responses (>75%) consisted of more than one Ca2+ spike in cells expressing WT PLCγ2, whereas this parameter was reduced to 35% in cells expressing the F897Q mutant, showing only a single Ca2+ spike in most cases (cf. Fig. 5, lower panels). Thus, reduced [Ca2+]i spiking from intracellular sources may also contribute to the substantial decrease in nuclear NFAT translocation caused by resistance of PLCγ2 to activation by Rac.

Because Rac GTPases are activated in B cells by quite a number of cell surface receptors in addition to the BCR, the signaling pathway convergence shown in this work is very well suited to detect coincident extracellular signals and to ensure signaling reliability by correctly interpreting the emergence of signals according to the particular cellular context and the specific (patho)physiological circumstances (71). Thus, agonist activation of receptors for integrins, chemokines, and pathogen-derived ligands has also been shown to activate Rac, which provides the GTPase with the potential to act as a central B cell signaling hub. For integrins, it is interesting to note that coligation of the β2αL integrin LFA-1 and BCR has been shown to cause an ∼10-fold reduction in the amount of BCR ligand required for B cells to make a tight contact with target membranes containing the receptor ligands (72). Using a similar experimental design, Henderson et al. (73) showed that Rac2 plays an important role in outside-in signaling from LFA-1 that leads to firm adhesion of murine B cells to ICAM-1. Interestingly, the Rac2 deficiency could be bypassed by treatment of B cells with phorbol ester and ionomycin, thus mimicking enhanced activation of PLCγ2. Although the molecular mechanisms of Rac activation by LFA-1 in B cells are unknown, they may be similar to those encountered in T cells (7476), although a non-Vav RacGEF(s) may be involved in this case (73). Hence, simultaneous activation of both BCR and LFA-1, e.g. at the immunological synapse, may give rise to locally enhanced Rac activation, allowing for an increased potency of BCR ligand to mediate integrin activation, at least in part by enhanced activity of PLCγ2 (73, 77).

A positive interaction between Rac and PLCγ2 may also be (patho)physiologically relevant in platelets, where PLCγ2 is activated downstream of other immunoreceptor tyrosine-based activation motif-coupled receptors, such as the major platelet collagen receptor glycoprotein VI or CLEC-2 (78), and by integrin α2β1 (79). Inactivation of the Rac1 gene in the mouse caused defective thrombus formation on collagen under flow conditions (78), and pharmacological inhibition of Rac in human platelets led to reduced PLCγ2 activity and impaired αIIbβ3 fibrinogen receptor stimulation (79). Intriguingly, the functional consequences of Rac1 deficiency were particularly striking at low and intermediate concentrations of GPVI or CLEC-2 receptor agonists (78), suggesting that Rac may enhance the sensitivity of PLCγ2 stimulation to extracellular ligands in platelets as well. Hence, pharmacological targeting of Rac1 could be an interesting approach in the development of future antiplatelet drugs.

The regulation of B cells by BCR ligation is intertwined with their regulation by certain chemokines, such as the CXCR4 and CXCR5 agonists CXCL12 and CXCL13, respectively (5). Activated CXCR4 and CXCR5 appear to be coupled to both Btk and PLCγ2 and, through a yet unidentified RacGEF (possibly DOCK2), to activation of Rac. Recently, Rac2 has been shown to be critical for LFA-1-mediated adhesion of mouse B cells in response to CXCL12 or CXCL13 (73). It remained unclear whether Rac2 causes enhanced adhesion directly by augmenting inside-out activation of integrins or indirectly by fostering receptor-mediated activation of PLCγ2. Recently, pathogen-derived signals have been suggested to activate Rac in murine B cells via Toll-like receptor TLR4 and MyD88 (80). Activated TLR4 activation also enhanced CXCR5-mediated Rac activation. Of note, TLR4 was found in other leukocytes to be coupled to activation of PLCγ2 (81, 82), raising the possibility that Rac is of regulatory importance in that respect as well.

The enhanced BCR ligand sensitivity observed for cells expressing WT PLCγ2 versus its F897Q mutant is reminiscent of the reduced threshold for antigen receptor stimulation observed in human peripheral blood B lymphocytes upon coligation of CD19 with BCR (3). In mouse B cells, the costimulatory effect of CD19 on BCR-mediated increases in [Ca2+]i was clearly reduced in B cells from rac2−/− mice (19). Previously, these effects were mainly ascribed to the known interactions of CD19 with the RhoGEF Vav and to the positive interaction of Vav, presumably via activated Rac, with phosphatidylinositol 4-phosphate 5-kinase and/or PI3K, followed by indirect activation of PLCγ2 (6, 42). Our current results strongly suggest that Rac enhances BCR-mediated Ca2+ signaling in B cells by direct interaction with and activation of PLCγ2.

Several families have been described with members affected by homozygous mutations in the CD19 gene causing undetectable or substantially decreased levels of CD19 in B cells. In these patients, suffering from an antibody-deficiency syndrome, there were marked alterations in Ca2+ mobilization in B cells following anti-IgM treatment (83). Similar defects of Ca2+ mobilization were observed in patients with defective CD81 and CD21, which function in a complex with CD225 and CD19 and cooperate with BCR to mediate antigen recognition (84, 85). Thus, the stimulatory interaction of PLCγ2 with Rac may not only play a pivotal role in determining the sensitivity of the BCR to stimulation by antigen, but may also contribute to BCR coreceptor signaling and to functional alterations of the latter in human disease such as certain forms of monogenetic common variable immunodeficiency (86). The very recent observation of a RAC2 loss-of-function mutation in two siblings with characteristics of a common variable immunodeficiency is consistent with this view (87).

Supplementary Material

Supplemental Data

Acknowledgments

We thank Norbert Zanker and Susanne Gierschik for excellent technical assistance. We thank Dr. Tomohiro Kurosaki for providing PLCγ2-deficient DT40 cells through RIKEN BioResource Center, National Bio-Resource Project of the Ministry of Education, Culture, Sports, Science, and Technology, Japan.

*

This work was supported by Deutsche Forschungsgemeinschaft Grants SFB 497 and SFB 1074 (to the P.G. and the G. U. N. laboratories) and grants within the International Graduate School in Molecular Medicine Ulm (IGradU) to the P. G. laboratory. The authors declare that they have no conflicts of interest with the contents of this article.

This article was selected as a Paper of the Week.

Inline graphic

This article contains supplemental Video S1.

4
The abbreviations used are:
PLC
inositol-phospholipid-specific phospholipase C
PH
pleckstrin homology
spPH
split PH domain
GTPγS
guanosine 5′-3-O-(thio)triphosphate
PCI
phospholipase C inhibitor peptide
NFAT
nuclear factor of activated T cell
Rac
Ras-related C3 botulinum toxin substrate
Rho
Ras homologous
BCR
B cell receptor
DAG
diacylglycerol
InsP3
inositol 1,4,5-trisphosphate
InsP3R
InsP3 receptor
PtdInsP2
phosphatidylinositol 4,5-bisphosphate
FRAP
fluorescence recovery after photobleaching.

References

  • 1. Kadamur G., Ross E. M. (2013) Mammalian phospholipase C. Annu. Rev. Physiol. 75, 127–154 [DOI] [PubMed] [Google Scholar]
  • 2. Sun Y., Thapa N., Hedman A. C., Anderson R. A. (2013) Phosphatidylinositol 4,5-bisphosphate: targeted production and signaling. BioEssays 35, 513–522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Tedder T. F. (2010) Innate and adaptive receptors interact to balance humoral immunity. J. Immunol. 184, 2231–2232 [DOI] [PubMed] [Google Scholar]
  • 4. Rawlings D. J., Schwartz M. A., Jackson S. W., Meyer-Bahlburg A. (2012) Integration of B cell responses through Toll-like receptors and antigen receptors. Nat. Rev. Immunol. 12, 282–294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. de Gorter D. J., Beuling E. A., Kersseboom R., Middendorp S., van Gils J. M., Hendriks R. W., Pals S. T., Spaargaren M. (2007) Bruton's tyrosine kinase and phospholipase Cγ2 mediate chemokine-controlled B cell migration and homing. Immunity 26, 93–104 [DOI] [PubMed] [Google Scholar]
  • 6. Scharenberg A. M., Humphries L. A., Rawlings D. J. (2007) Calcium signalling and cell-fate choice in B cells. Nat. Rev. Immunol. 7, 778–789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Baba Y., Kurosaki T. (2011) Impact of Ca2+ signaling on B cell function. Trends Immunol. 32, 589–594 [DOI] [PubMed] [Google Scholar]
  • 8. Wang D., Feng J., Wen R., Marine J. C., Sangster M. Y., Parganas E., Hoffmeyer A., Jackson C. W., Cleveland J. L., Murray P. J., Ihle J. N. (2000) Phospholipase Cγ2 is essential in the functions of B cell and several Fc receptors. Immunity. 13, 25–35 [DOI] [PubMed] [Google Scholar]
  • 9. Hashimoto A., Takeda K., Inaba M., Sekimata M., Kaisho T., Ikehara S., Homma Y., Akira S., Kurosaki T. (2000) Essential role of phospholipase C-γ2 in B cell development and function. J. Immunol. 165, 1738–1742 [DOI] [PubMed] [Google Scholar]
  • 10. Wen R., Chen Y., Bai L., Fu G., Schuman J., Dai X., Zeng H., Yang C., Stephan R. P., Cleveland J. L., Wang D. (2006) Essential role of phospholipase C γ2 in early B cell development and Myc-mediated lymphomagenesis. Mol. Cell. Biol. 26, 9364–9376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Hikida M., Johmura S., Hashimoto A., Takezaki M., Kurosaki T. (2003) Coupling between B cell receptor and phospholipase C-γ2 is essential for mature B cell development. J. Exp. Med. 198, 581–589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Bai L., Chen Y., He Y., Dai X., Lin X., Wen R., Wang D. (2007) Phospholipase Cγ2 contributes to light-chain gene activation and receptor editing. Mol. Cell. Biol. 27, 5957–5967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Yu P., Constien R., Dear N., Katan M., Hanke P., Bunney T. D., Kunder S., Quintanilla-Martinez L., Huffstadt U., Schröder A., Jones N. P., Peters T., Fuchs H., de Angelis M. H., Nehls M., et al. (2005) Autoimmunity and inflammation due to a gain-of-function mutation in phospholipase Cγ2 that specifically increases external Ca2+ entry. Immunity 22, 451–465 [DOI] [PubMed] [Google Scholar]
  • 14. Abe K., Fuchs H., Boersma A., Hans W., Yu P., Kalaydjiev S., Klaften M., Adler T., Calzada-Wack J., Mossbrugger I., Rathkolb B., Rozman J., Prehn C., Maraslioglu M., Kametani Y., et al. (2011) A novel ENU-induced mutation in phospholipase Cγ2 causes inflammatory arthritis, metabolic defects, and in vitro male infertility in the mouse. Arthritis Rheum. 63, 1301–1311 [DOI] [PubMed] [Google Scholar]
  • 15. Ombrello M. J., Remmers E. F., Sun G., Freeman A. F., Datta S., Torabi-Parizi P., Subramanian N., Bunney T. D., Baxendale R. W., Martins M. S., Romberg N., Komarow H., Aksentijevich I., Kim H. S., Ho J., et al. (2012) Cold urticaria, immunodeficiency, and autoimmunity related to PLCG2 deletions. N. Engl. J. Med. 366, 330–338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Zhou Q., Lee G. S., Brady J., Datta S., Katan M., Sheikh A., Martins M. S., Bunney T. D., Santich B. H., Moir S., Kuhns D. B., Long Priel D. A., Ombrello A., Stone D., Ombrello M. J., et al. (2012) A hypermorphic missense mutation in PLCG2, encoding phospholipase Cγ2, causes a dominantly inherited autoinflammatory disease with immunodeficiency. Am. J. Hum. Genet. 91, 713–720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Fujikawa K., Miletic A. V., Alt F. W., Faccio R., Brown T., Hoog J., Fredericks J., Nishi S., Mildiner S., Moores S. L., Brugge J., Rosen F. S., Swat W. (2003) Vav1/2/3-null mice define an essential role for Vav family proteins in lymphocyte development and activation but a differential requirement in MAPK signaling in T and B cells. J. Exp. Med. 198, 1595–1608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Kuhne M. R., Ku G., Weiss A. (2000) A guanine nucleotide exchange factor-independent function of Vav1 in transcriptional activation. J. Biol. Chem. 275, 2185–2190 [DOI] [PubMed] [Google Scholar]
  • 19. Croker B. A., Tarlinton D. M., Cluse L. A., Tuxen A. J., Light A., Yang F. C., Williams D. A., Roberts A. W. (2002) The Rac2 guanosine triphosphatase regulates B lymphocyte antigen receptor responses and chemotaxis and is required for establishment of B-1a and marginal zone B lymphocytes. J. Immunol. 168, 3376–3386 [DOI] [PubMed] [Google Scholar]
  • 20. Walmsley M. J., Ooi S. K., Reynolds L. F., Smith S. H., Ruf S., Mathiot A., Vanes L., Williams D. A., Cancro M. P., Tybulewicz V. L. (2003) Critical roles for Rac1 and Rac2 GTPases in B cell development and signaling. Science 302, 459–462 [DOI] [PubMed] [Google Scholar]
  • 21. Tedford K., Nitschke L., Girkontaite I., Charlesworth A., Chan G., Sakk V., Barbacid M., Fischer K. D. (2001) Compensation between Vav-1 and Vav-2 in B cell development and antigen receptor signaling. Nat. Immunol. 2, 548–555 [DOI] [PubMed] [Google Scholar]
  • 22. Bae Y. S., Cantley L. G., Chen C. S., Kim S. R., Kwon K. S., Rhee S. G. (1998) Activation of phospholipase C-γ by phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 273, 4465–4469 [DOI] [PubMed] [Google Scholar]
  • 23. Piechulek T., Rehlen T., Walliser C., Vatter P., Moepps B., Gierschik P. (2005) Isozyme-specific stimulation of phospholipase C-γ2 by Rac GTPases. J. Biol. Chem. 280, 38923–38931 [DOI] [PubMed] [Google Scholar]
  • 24. Walliser C., Retlich M., Harris R., Everett K. L., Josephs M. B., Vatter P., Esposito D., Driscoll P. C., Katan M., Gierschik P., Bunney T. D. (2008) Rac regulates its effector phospholipase Cγ2 through interaction with a split pleckstrin homology domain. J. Biol. Chem. 283, 30351–30362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Bunney T. D., Opaleye O., Roe S. M., Vatter P., Baxendale R. W., Walliser C., Everett K. L., Josephs M. B., Christow C., Rodrigues-Lima F., Gierschik P., Pearl L. H., Katan M. (2009) Structural insights into formation of an active signaling complex between Rac and phospholipase Cγ2. Mol. Cell 34, 223–233 [DOI] [PubMed] [Google Scholar]
  • 26. Tybulewicz V. L., Henderson R. B. (2009) Rho family GTPases and their regulators in lymphocytes. Nat. Rev. Immunol. 9, 630–644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Schaefer B. C. (1995) Revolutions in rapid amplification of cDNA ends: new strategies for polymerase chain reaction cloning of full-length cDNA ends. Anal. Biochem. 227, 255–273 [DOI] [PubMed] [Google Scholar]
  • 28. Kredel S., Nienhaus K., Oswald F., Wolff M., Ivanchenko S., Cymer F., Jeromin A., Michel F. J., Spindler K. D., Heilker R., Nienhaus G. U., Wiedenmann J. (2008) Optimized and far-red-emitting variants of fluorescent protein eqFP611. Chem. Biol. 15, 224–233 [DOI] [PubMed] [Google Scholar]
  • 29. Arakawa H., Lodygin D., Buerstedde J. M. (2001) Mutant loxP vectors for selectable marker recycle and conditional knock-outs. BMC Biotechnol. 1, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Jiang X., Weise S., Hafner M., Röcker C., Zhang F., Parak W. J., Nienhaus G. U. (2010) Quantitative analysis of the protein corona on FePt nanoparticles formed by transferrin binding. J. R. Soc. Interface 7, S5–S13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Qin S., Chock P. B. (2001) Bruton's tyrosine kinase is essential for hydrogen peroxide-induced calcium signaling. Biochemistry 40, 8085–8091 [DOI] [PubMed] [Google Scholar]
  • 32. Axelrod D., Koppel D. E., Schlessinger J., Elson E., Webb W. W. (1976) Mobility measurement by analysis of fluorescence photobleaching recovery kinetics. Biophys. J. 16, 1055–1069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Koppel D. E., Axelrod D., Schlessinger J., Elson E. L., Webb W. W. (1976) Dynamics of fluorescence marker concentration as a probe of mobility. Biophys. J. 16, 1315–1329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Illenberger D., Walliser C., Strobel J., Gutman O., Niv H., Gaidzik V., Kloog Y., Gierschik P., Henis Y. I. (2003) Rac2 regulation of phospholipase C-β2 activity and mode of membrane interactions in intact cells. J. Biol. Chem. 278, 8645–8652 [DOI] [PubMed] [Google Scholar]
  • 35. Henis Y. I., Rotblat B., Kloog Y. (2006) FRAP beam-size analysis to measure palmitoylation-dependent membrane association dynamics and microdomain partitioning of Ras proteins. Methods 40, 183–190 [DOI] [PubMed] [Google Scholar]
  • 36. Gutman O., Walliser C., Piechulek T., Gierschik P., Henis Y. I. (2010) Differential regulation of phospholipase C-β2 activity and membrane interaction by Gαq, Gβ1γ2, and Rac2. J. Biol. Chem. 285, 3905–3915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Petersen N. O., Felder S., Elson E. L. (1986) in Handbook of Experimental Immunology (Weir D. M., Herzenberg L. A., Blackwell C. C., Herzenberg L. A., eds) pp. 24.21–24.24, Blackwell Scientific Publications, Edinburgh [Google Scholar]
  • 38. Efron B., Tibshirani R. (1993) in Introduction to Bootstrap (Cox D. R., Hinkley D. V., Reid N., Rubin D. B., Silverman B. W., eds) pp. 124–130, Chapman and Hall Ltd., London [Google Scholar]
  • 39. Spoerner M., Herrmann C., Vetter I. R., Kalbitzer H. R., Wittinghofer A. (2001) Dynamic properties of the Ras switch I region and its importance for binding to effectors. Proc. Natl. Acad. Sci. U.S.A. 98, 4944–4949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Patterson R. L., van Rossum D. B., Ford D. L., Hurt K. J., Bae S. S., Suh P. G., Kurosaki T., Snyder S. H., Gill D. L. (2002) Phospholipase C-γ is required for agonist-induced Ca2+ entry. Cell 111, 529–541 [DOI] [PubMed] [Google Scholar]
  • 41. Morita T., Tanimura A., Nezu A., Kurosaki T., Tojyo Y. (2004) Functional analysis of the green fluorescent protein-tagged inositol 1,4,5-trisphosphate receptor type 3 in Ca2+ release and entry in DT40 B lymphocytes. Biochem. J. 382, 793–801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kurosaki T. (2002) Regulation of B cell signal transduction by adaptor proteins. Nat. Rev. Immunol. 2, 354–363 [DOI] [PubMed] [Google Scholar]
  • 43. Harwood N. E., Batista F. D. (2010) Early events in B cell activation. Annu. Rev. Immunol. 28, 185–210 [DOI] [PubMed] [Google Scholar]
  • 44. Deckert M., Tartare-Deckert S., Couture C., Mustelin T., Altman A. (1996) Functional and physical interactions of Syk family kinases with the Vav proto-oncogene product. Immunity 5, 591–604 [DOI] [PubMed] [Google Scholar]
  • 45. Guinamard R., Fougereau M., Seckinger P. (1997) The SH3 domain of Bruton's tyrosine kinase interacts with Vav, Sam68 and EWS. Scand. J. Immunol. 45, 587–595 [DOI] [PubMed] [Google Scholar]
  • 46. Fu C., Turck C. W., Kurosaki T., Chan A. C. (1998) BLNK: a central linker protein in B cell activation. Immunity 9, 93–103 [DOI] [PubMed] [Google Scholar]
  • 47. Wienands J., Schweikert J., Wollscheid B., Jumaa H., Nielsen P. J., Reth M. (1998) SLP-65: a new signaling component in B lymphocytes which requires expression of the antigen receptor for phosphorylation. J. Exp. Med. 188, 791–795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Arana E., Vehlow A., Harwood N. E., Vigorito E., Henderson R., Turner M., Tybulewicz V. L., Batista F. D. (2008) Activation of the small GTPase Rac2 via the B cell receptor regulates B cell adhesion and immunological-synapse formation. Immunity 28, 88–99 [DOI] [PubMed] [Google Scholar]
  • 49. Hashimoto A., Okada H., Jiang A., Kurosaki M., Greenberg S., Clark E. A., Kurosaki T. (1998) Involvement of guanosine triphosphatases and phospholipase C-γ2 in extracellular signal-regulated kinase, c-Jun NH2-terminal kinase, and p38 mitogen-activated protein kinase activation by the B cell antigen receptor. J. Exp. Med. 188, 1287–1295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Hao S., Kurosaki T., August A. (2003) Differential regulation of NFAT and SRF by the B cell receptor via a PLCγ-Ca2+-dependent pathway. EMBO J. 22, 4166–4177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Weber M., Treanor B., Depoil D., Shinohara H., Harwood N. E., Hikida M., Kurosaki T., Batista F. D. (2008) Phospholipase C-γ2 and Vav cooperate within signaling microclusters to propagate B cell spreading in response to membrane-bound antigen. J. Exp. Med. 205, 853–868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Dupont G., Combettes L., Bird G. S., Putney J. W. (2011) Calcium oscillations. Cold Spring Harb. Perspect. Biol. 3, a004226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Berridge M. J. (2007) Inositol trisphosphate and calcium oscillations. Biochem. Soc. Symp. 74, 1–7 [DOI] [PubMed] [Google Scholar]
  • 54. Miyakawa T., Maeda A., Yamazawa T., Hirose K., Kurosaki T., Iino M. (1999) Encoding of Ca2+ signals by differential expression of IP3 receptor subtypes. EMBO J. 18, 1303–1308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Hirose K., Kadowaki S., Tanabe M., Takeshima H., Iino M. (1999) Spatiotemporal dynamics of inositol 1,4,5-trisphosphate that underlies complex Ca2+ mobilization patterns. Science 284, 1527–1530 [DOI] [PubMed] [Google Scholar]
  • 56. Nishida M., Sugimoto K., Hara Y., Mori E., Morii T., Kurosaki T., Mori Y. (2003) Amplification of receptor signalling by Ca2+ entry-mediated translocation and activation of PLCγ2 in B lymphocytes. EMBO J. 22, 4677–4688 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Kiyonaka S., Kato K., Nishida M., Mio K., Numaga T., Sawaguchi Y., Yoshida T., Wakamori M., Mori E., Numata T., Ishii M., Takemoto H., Ojida A., Watanabe K., Uemura A., et al. (2009) Selective and direct inhibition of TRPC3 channels underlies biological activities of a pyrazole compound. Proc. Natl. Acad. Sci. U.S.A. 106, 5400–5405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Kitajima N., Watanabe K., Morimoto S., Sato Y., Kiyonaka S., Hoshijima M., Ikeda Y., Nakaya M., Ide T., Mori Y., Kurose H., Nishida M. (2011) TRPC3-mediated Ca2+ influx contributes to Rac1-mediated production of reactive oxygen species in MLP-deficient mouse hearts. Biochem. Biophys. Res. Commun. 409, 108–113 [DOI] [PubMed] [Google Scholar]
  • 59. Bezzerides V. J., Ramsey I. S., Kotecha S., Greka A., Clapham D. E. (2004) Rapid vesicular translocation and insertion of TRP channels. Nat. Cell Biol. 6, 709–720 [DOI] [PubMed] [Google Scholar]
  • 60. Hajnóczky G., Thomas A. P. (1997) Minimal requirements for calcium oscillations driven by the IP3 receptor. EMBO J. 16, 3533–3543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Rossi A. M., Tovey S. C., Rahman T., Prole D. L., Taylor C. W. (2012) Analysis of IP3 receptors in and out of cells. Biochim. Biophys. Acta 1820, 1214–1227 [DOI] [PubMed] [Google Scholar]
  • 62. Cambier J., Chen Z. Z., Pasternak J., Ransom J., Sandoval V., Pickles H. (1988) Ligand-induced desensitization of B cell membrane immunoglobulin-mediated Ca2+ mobilization and protein kinase C translocation. Proc. Natl. Acad. Sci. U.S.A. 85, 6493–6497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Cambier J. C., Fisher C. L., Pickles H., Morrison D. C. (1990) Dual molecular mechanisms mediate ligand-induced membrane Ig desensitization. J. Immunol. 145, 13–19 [PubMed] [Google Scholar]
  • 64. Hoffman G. R., Nassar N., Oswald R. E., Cerione R. A. (1998) Fluoride activation of the Rho family GTP-binding protein Cdc42Hs. J. Biol. Chem. 273, 4392–4399 [DOI] [PubMed] [Google Scholar]
  • 65. Amin R. H., Chen H. Q., Veluthakal R., Silver R. B., Li J., Li G., Kowluru A. (2003) Mastoparan-induced insulin secretion from insulin-secreting βTC3 and INS-1 cells: evidence for its regulation by Rho subfamily of G proteins. Endocrinology 144, 4508–4518 [DOI] [PubMed] [Google Scholar]
  • 66. Dolmetsch R. E., Lewis R. S., Goodnow C. C., Healy J. I. (1997) Differential activation of transcription factors induced by Ca2+ response amplitude and duration. Nature 386, 855–858 [DOI] [PubMed] [Google Scholar]
  • 67. Dolmetsch R. E., Xu K., Lewis R. S. (1998) Calcium oscillations increase the efficiency and specificity of gene expression. Nature 392, 933–936 [DOI] [PubMed] [Google Scholar]
  • 68. Li W., Llopis J., Whitney M., Zlokarnik G., Tsien R. Y. (1998) Cell-permeant caged InsP3 ester shows that Ca2+ spike frequency can optimize gene expression. Nature 392, 936–941 [DOI] [PubMed] [Google Scholar]
  • 69. Tomida T., Hirose K., Takizawa A., Shibasaki F., Iino M. (2003) NFAT functions as a working memory of Ca2+ signals in decoding Ca2+ oscillation. EMBO J. 22, 3825–3832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Gwack Y., Feske S., Srikanth S., Hogan P. G., Rao A. (2007) Signalling to transcription: store-operated Ca2+ entry and NFAT activation in lymphocytes. Cell Calcium 42, 145–156 [DOI] [PubMed] [Google Scholar]
  • 71. Bourne H. R., Nicoll R. (1993) Molecular machines integrate coincident synaptic signals. Cell 72, 65–75 [DOI] [PubMed] [Google Scholar]
  • 72. Carrasco Y. R., Fleire S. J., Cameron T., Dustin M. L., Batista F. D. (2004) LFA-1/ICAM-1 interaction lowers the threshold of B cell activation by facilitating B cell adhesion and synapse formation. Immunity 20, 589–599 [DOI] [PubMed] [Google Scholar]
  • 73. Henderson R. B., Grys K., Vehlow A., de Bettignies C., Zachacz A., Henley T., Turner M., Batista F., Tybulewicz V. L. (2010) A novel Rac-dependent checkpoint in B cell development controls entry into the splenic white pulp and cell survival. J. Exp. Med. 207, 837–853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Sánchez-Martín L., Sánchez-Sánchez N., Gutiérrez-López M. D., Rojo A. I., Vicente-Manzanares M., Pérez-Alvarez M. J., Sánchez-Mateos P., Bustelo X. R., Cuadrado A., Sánchez-Madrid F., Rodríguez-Fernández J. L., Cabañas C. (2004) Signaling through the leukocyte integrin LFA-1 in T cells induces a transient activation of Rac-1 that is regulated by Vav and PI3K/Akt-1. J. Biol. Chem. 279, 16194–16205 [DOI] [PubMed] [Google Scholar]
  • 75. Nishihara H., Maeda M., Oda A., Tsuda M., Sawa H., Nagashima K., Tanaka S. (2002) DOCK2 associates with CrkL and regulates Rac1 in human leukemia cell lines. Blood 100, 3968–3974 [DOI] [PubMed] [Google Scholar]
  • 76. Hogg N., Laschinger M., Giles K., McDowall A. (2003) T-cell integrins: more than just sticking points. J. Cell Sci. 116, 4695–4705 [DOI] [PubMed] [Google Scholar]
  • 77. Spaargaren M., Beuling E. A., Rurup M. L., Meijer H. P., Klok M. D., Middendorp S., Hendriks R. W., Pals S. T. (2003) The B cell antigen receptor controls integrin activity through Btk and PLCγ2. J. Exp. Med. 198, 1539–1550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Pleines I., Elvers M., Strehl A., Pozgajova M., Varga-Szabo D., May F., Chrostek-Grashoff A., Brakebusch C., Nieswandt B. (2009) Rac1 is essential for phospholipase C-γ2 activation in platelets. Pflugers Arch. 457, 1173–1185 [DOI] [PubMed] [Google Scholar]
  • 79. Guidetti G. F., Bernardi B., Consonni A., Rizzo P., Gruppi C., Balduini C., Torti M. (2009) Integrin α2β1 induces phosphorylation-dependent and phosphorylation-independent activation of phospholipase Cγ2 in platelets: role of Src kinase and Rac GTPase. J. Thromb. Haemost. 7, 1200–1206 [DOI] [PubMed] [Google Scholar]
  • 80. Barrio L., Saez de Guinoa J., Carrasco Y. R. (2013) TLR4 signaling shapes B cell dynamics via MyD88-dependent pathways and Rac GTPases. J. Immunol. 191, 3867–3875 [DOI] [PubMed] [Google Scholar]
  • 81. Aki D., Minoda Y., Yoshida H., Watanabe S., Yoshida R., Takaesu G., Chinen T., Inaba T., Hikida M., Kurosaki T., Saeki K., Yoshimura A. (2008) Peptidoglycan and lipopolysaccharide activate PLCγ2, leading to enhanced cytokine production in macrophages and dendritic cells. Genes Cells 13, 199–208 [DOI] [PubMed] [Google Scholar]
  • 82. Chiang C. Y., Veckman V., Limmer K., David M. (2012) Phospholipase Cγ-2 and intracellular calcium are required for lipopolysaccharide-induced Toll-like receptor 4 (TLR4) endocytosis and interferon regulatory factor 3 (IRF3) activation. J. Biol. Chem. 287, 3704–3709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. van Zelm M. C., Reisli I., van der Burg M., Castaño D., van Noesel C. J., van Tol M. J., Woellner C., Grimbacher B., Patiño P. J., van Dongen J. J., Franco J. L. (2006) An antibody-deficiency syndrome due to mutations in the CD19 gene. N. Engl. J. Med. 354, 1901–1912 [DOI] [PubMed] [Google Scholar]
  • 84. van Zelm M. C., Smet J., Adams B., Mascart F., Schandené L., Janssen F., Ferster A., Kuo C. C., Levy S., van Dongen J. J., van der Burg M. (2010) CD81 gene defect in humans disrupts CD19 complex formation and leads to antibody deficiency. J. Clin. Invest. 120, 1265–1274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Thiel J., Kimmig L., Salzer U., Grudzien M., Lebrecht D., Hagena T., Draeger R., Voelxen N., Völxen N., Bergbreiter A., Jennings S., Gutenberger S., Aichem A., Illges H., Hannan J. P., et al. (2012) Genetic CD21 deficiency is associated with hypogammaglobulinemia. J. Allergy Clin. Immunol. 129, 801–810 [DOI] [PubMed] [Google Scholar]
  • 86. Salzer U., Warnatz K., Peter H. H. (2012) Common variable immunodeficiency–an update. Arthritis Res. Ther. 14, 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Alkhairy O. K., Rezaei N., Graham R. R., Abolhassani H., Borte S., Hultenby K., Wu C., Aghamohammadi A., Williams D. A., Behrens T. W., Hammarström L., Pan-Hammarström Q. (2015) RAC2 loss-of-function mutation in 2 siblings with characteristics of common variable immunodeficiency. J. Allergy Clin. Immunol. 135, 1380–1384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Homma Y., Takenawa T. (1992) Inhibitory effect of src homology (SH) 2/SH3 fragments of phospholipase C-γ on the catalytic activity of phospholipase C isoforms: identification of a novel phospholipase C inhibitor region. J. Biol. Chem. 267, 21844–21849 [PubMed] [Google Scholar]
  • 89. Takata M., Homma Y., Kurosaki T. (1995) Requirement of phospholipase C-γ2 activation in surface immunoglobulin M-induced B cell apoptosis. J. Exp. Med. 182, 907–914 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Data
Download video file (8.4MB, avi)

Articles from The Journal of Biological Chemistry are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES