Skip to main content
Journal of Leukocyte Biology logoLink to Journal of Leukocyte Biology
. 2010 Mar 3;87(6):1041–1057. doi: 10.1189/jlb.1108708

Clostridium difficile toxin B differentially affects GPCR-stimulated Ca2+ responses in macrophages: independent roles for Rho and PLA2

Robert A Rebres *,1, Christina Moon *, Dianne DeCamp †,2, Keng-Mean Lin , Iain D Fraser ‡,3, Stephen B Milne §, Tamara I A Roach *, H Alex Brown §, William E Seaman *,∥
PMCID: PMC2872536  PMID: 20200401

Abstract

Clostridium difficile toxins cause acute colitis by disrupting the enterocyte barrier and promoting inflammation. ToxB from C. difficile inactivates Rho family GTPases and causes release of cytokines and eicosanoids by macrophages. We studied the effects of ToxB on GPCR signaling in murine RAW264.7 macrophages and found that ToxB elevated Ca2+ responses to Gαi-linked receptors, including the C5aR, but reduced responses to Gαq-linked receptors, including the UDP receptors. Other Rho inhibitors also reduced UDP Ca2+ responses, but they did not affect C5a responses, suggesting that ToxB inhibited UDP responses by inhibiting Rho but enhanced C5a responses by other mechanisms. By using PLCβ isoform-deficient BMDM, we found that ToxB inhibited Ca2+ signaling through PLCβ4 but enhanced signaling through PLCβ3. Effects of ToxB on GPCR Ca2+ responses correlated with GPCR use of PLCβ3 versus PLCβ4. ToxB inhibited UDP Ca2+ signaling without reducing InsP3 production or the sensitivity of cellular Ca2+ stores to exogenous InsP3, suggesting that ToxB impairs UDP signaling at the level of InsP3/Ca2+coupling. In contrast, ToxB elevated InsP3 production by C5a, and the enhancement of Ca2+ signaling by C5a was prevented by inhibition of PLA2 or 5-LOX but not COX, implicating LTs but not prostanoids in the mechanism. In sum, ToxB has opposing, independently regulated effects on Ca2+ signaling by different GPCR-linked PLCβ isoforms in macrophages.

Keywords: phospholipase C, lipoxygenase, complement

Introduction

Pathologic overgrowth of Clostridium difficile causes pseudomembranous colitis when normal flora are disrupted during antibiotic therapy. C. difficile is the leading cause of hospital-acquired diarrhea in the United States, causing >250,000 cases/year of diarrhea at a cost of more than $1 billion/year in added hospitalization costs [1, 2]. Disruption of intestinal function and integrity results primarily from the effects of two toxins produced by C. difficile, ToxA and ToxB. ToxA, an enterotoxin, has direct effects on enterocyte ion flux and barrier integrity, and ToxB, a cytotoxin, induces cell death via apoptosis [3]. Both toxins contribute to the virulence of C. difficile [4, 5]. A major target of both toxins is the Rho family of small G-proteins [6,7,8], which regulates cellular structure and metabolism via effects on cytoskeletal organization [9, 10]. Both toxins additionally promote inflammation by activating the production of cytokines and eicosanoids. IL-1β, TNF-α, IL-8, IL-6, and MCP-1 are generated, and the activation of PLA2 also yields AA, PGE2, and LTs [11,12,13,14,15]. The resulting influx of neutrophils and macrophages and the resulting production of inflammatory mediators cause additional tissue injury.

GPCRs are important regulators of enterocyte and leukocyte function and of the inflammation associated with C. difficile infection. Trimeric G-proteins, activated by GPCRs, stimulate PLCβ isoforms to catalyze the conversion of PIP2 to DAG and InsP3; InsP3 acts on its receptors (InsP3Rs) to release Ca2+ from intracellular stores in the ER. Ca2+ signaling requires the spatial organization of these components in close proximity, providing a level of cellular regulation by compartmentalization of cellular biochemistry. Ca2+ responses from different receptors in the same cell, however, can vary widely, and we are at an early stage of understanding the factors that determine the magnitude and pattern of Ca2+ responses [16,17,18].

Gαq- and Gαi-linked GPCRs can activate Rho family members as well as Ca2+ responses. Activation of these signaling pathways differs by receptor and cell type or status, and analysis of some systems has demonstrated Rho-dependent regulation of Ca2+ responses, suggesting interaction of these signals. Levels at which Rho family members can regulate changes in [Ca2+]i include modulation of receptor activation through control of cytoskeletal organization [19,20,21,22], assembly of Ca2+ signaling complexes on scaffolding proteins [18, 23, 24], regulation of phosphatidylinositol kinases [25,26,27,28,29,30], and direct activation of PLC [31,32,33,34,35]. In all of these mechanisms described, Rho family proteins facilitate Ca2+ responses.

In our current studies of GPCR-mediated Ca2+ signaling in macrophages, we found that ToxB differentially affects Ca2+ responses to different GPCRs based on PLCβ isoform dependence. In RAW264.7 cells, ToxB increased Ca2+ responses robustly by Gαi-linked receptors, such as C5aR, but it reduced responses by Gαq-linked receptors, such as the P2Y receptors for UDP. Studies in BMDM showed that this difference correlated with differences between these receptors in their use of PLCβ isoforms and that ToxB enhanced Ca2+ signaling through PLCβ3 but inhibited Ca2+ signaling through PLCβ4. The inhibition of UDP Ca2+ signaling by ToxB was mediated by the loss of Rho activity. In contrast, the enhancement of C5a Ca2+ signaling was independent of Rho and reflected an increase in InsP3 production, which was dependent on the activation of PLA2 and the consequent production of LTs. These studies identify for the first time a differential requirement for Rho in GPCR-mediated Ca2+ signaling by macrophages, and they identify independent regulation of PLCβ isoforms by ToxB. These features of ToxB may be important in the colitis that follows infection by C. difficile.

MATERIALS AND METHODS

Reagents

Human complement fragment C5a, UDP, UTP, LPA, PAF, and spiradoline were from Sigma-Aldrich (St. Louis, MO, USA). S1P was from Avanti Polar Lipids (Alabaster, AL, USA). C. difficile ToxA and ToxB (VPI 10463) were from Calbiochem (San Diego, CA, USA) or List Biological Laboratories (Campbell, CA, USA). Native Clostridium botulinum C3 exotoxin (C3 transferase) was from Calbiochem, and cell-permeable C3 exotoxin was from Cytoskeleton, Inc., (Denver, CO, USA). PTx was from List Biological Laboratories. Fura-2 and Fluo-3 were from Invitrogen (Carlsbad, CA, USA). Photolabile-caged InsP3 [D-2,3-O-Isopropylidene-6-O(2-nitro-4,5-dimethoxy) benzyl-myo-inositol 1,4,5-trisphosphate-hexakis(propionoxymethyl)ester] was from Axxora, LLC (San Diego, CA, USA). FTA, AFC, and AGGC were from Cayman Chemical Co. (Ann Arbor, MI, USA). Ionomycin, thapsigargin, LY294002, wortmannin, PP2, and PP3 were from Calbiochem. AACOCF3, MAFP, indomethacin, MK-886, and quinacrine were from EMD Biosciences (Gibbstown, NJ, USA). Cytochalasin D and AA861 were from Sigma-Aldrich. Jasplakinolide, latrunculin A, colchicine, and nocodazole were from EMD Biosciences. AA was from NuChek Prep (Elysian, MN, USA). Mouse IgG2a was from BD Biosciences (San Jose, CA, USA), and F(ab′)2 fragments of goat anti-mouse IgG were from Jackson ImmunoResearch Laboratories, Inc. (West Grove, PA, USA). Anti-P2Y6R was from Alomone Labs, anti-C5aR (CD88) was from BD Biosciences, anti-FLAG® was from Sigma-Aldrich, and anti-P-AKT, anti-P-ERK, anti-P-p38, anti-P-JNK, anti-P-PLCγ2, and anti-PLCγ2 were from Cell Signaling Technology (Danvers, MA, USA). Annexin V-FITC and propidium iodide staining solutions were from EMD Biosciences. Additional detailed protocols for reagents, procedures, and solutions are available on the AfCS websites (http://www.signaling-gateway.org/data/ProtocolLinks.html and www.afcs.org) and are referenced according to protocol number (e.g., PP00000226).

Cell culture

RAW264.7 cells, originally obtained from the American Type Culture Collection (Manassas, VA, USA), were grown in DMEM supplemented with 10% FBS, 20 mM HEPES, and 2 mM glutamine (Protocol PS00000636) in non-TC dishes. Cells were passed every 2–3 days by harvest using PBS with 2 mM EDTA. Further details are described on the AfCS websites (Protocol PP00000226). J774 cells were grown similarly but in TC flasks. Culture media and ToxB were assayed for endotoxin contamination by limulus lysate assay and found to contain <0.001 EU/ml at final concentrations in our assays.

Mice genetically deficient in PLCβ2, PLCβ3, or PLCβ4 were described previously [36,37,38,39,40,41,42]. PLCβ3−/− and PLCβ4−/− mice were on the C57BL/6 background, and PLCβ2−/− mice were on the 129SV background. PLCβ-deficient mice and corresponding wt strains were bred and housed at California Institute of Technology (Pasadena, CA, USA) under standard conditions. All experiments were conducted according to protocols approved by the Animal Care and Use Committees of The California Institute of Technology and the San Francisco Veterans Affairs Medical Center (San Francisco, CA, USA). To obtain BMDM, femurs and tibias were removed from sex- and age-matched mice (routinely 4–20 weeks of age, matched ±4 weeks). Cells were extracted immediately, or bones were shipped in medium on ice overnight to another laboratory location. Marrow was flushed from bones, erythrocytes were lysed, and the resulting white cells were seeded in non-TC Petri dishes for selection by growth and adhesion. GM (BMDM-GM) contained DMEM plus 10% FBS, 10% CMG14-12 supernatant (containing murine rM-CSF) [43], 2 mM glutamine, 25 mM HEPES, 100 U/ml penicillin, and 50 μM β-ME (Protocol PS00000722). After 6 days, all of the surviving cells were macrophages, as indicated by CD115 (c-FMS) and CD11b (αM integrin) expression levels, and these cells were maintained for up to 35 days in culture with replacement of medium every 2–3 days and dilution to reduce cell density as needed. Passage was by brief exposure and harvest in PBS with 2 mM EDTA and 0.5% BSA, centrifugation at 250 g, and replating for culture or assay. Cells were cultured overnight in assay plates prior to use in assays (Protocol PP00000172).

Cells were treated with 5–100 ng/ml ToxB in culture medium for 2–20 h as indicated. Cells were treated with 100 μg/ml native, nonpermeable C3 exotoxin for 20 h or with 1–8 μg/ml cell-permeable C3 exotoxin for 1–20 h. Cells were intoxicated with 50–100 ng/ml PTx by culture overnight (18–22 h). Cytoskeletal, kinase, phospholipase, and LOX inhibitors or the corresponding solvent control solutions were applied for the 30-min equilibration period prior to and during assays as described below. For AA exposure, cells were treated, as described by Rouzer et al. [44], in the presence or absence of 10 μg/ml AA for 4–20 h prior to Ca2+ assay.

Expression constructs and transfection

Tobias Meyer and co-workers [45] generously provided dominant-negative (T19N) and constitutively active (Q63L) forms of Rho family proteins as CFP-fusion constructs in the plasmid pECFP (Clontech, Palo Alto, CA, USA). CFP or GFP expressed alone were used as controls. Transient transfection of RAW264.7 cells was performed with Lipofectamine 2000, and cells were assayed 24–36 h post-transfection. Transfection efficiency was 10–40%. Assays were performed on these mixed populations to allow comparison of responses of positive to negative cells in the same field of view during single-cell Ca2+ assays. After the Fura-2 Ca2+ fluorescence time series, images of CFP or GFP fluorescence were collected to allow the gating of individual cells into positive and negative populations.

Population Ca2+ assays

Ca2+ responses were measured by monitoring the fluorescence signals of Fura-2-loaded cells in 96-well plates in a fluorescence spectrophotometric plate reader (FLEXstation, Molecular Devices, Sunnyvale, CA, USA). Cells were grown overnight in black-walled TC grade assay plates, loaded with Fura-2-AM (4 μM) for 30 min at room temperature, equilibrated for 30 min at 37°C, and then assayed at 37°C. Baseline readings were collected for 30–40 s prior to ligand addition, and responses were evaluated for 2.5 min after ligand addition. Calibration steps, including additions of a Ca2+-minimizing solution (Protocol PS00000607) and Fura-2 Ca2+-saturating solution (Protocol PS00000608), were performed at the end of each recording to allow calculation of [Ca2+]i values, according to the method of Grynkiewicz et al. [46], assuming a cytoplasmic Kd of 250 nM for Fura-2. Ca2+ signals during the response period were quantified by several features, including the peak-offset response (difference between average baseline Ca2+ level and the maximal Ca2+ level observed, reported in nM) and the integrated response (integrated Ca2+ level above the average baseline over the indicated time period, e.g., 20 s or 2.5 min, reported in nM×s). Further details of the Ca2+ assay are described in Protocol PP00000211.

Single-cell Ca2+ assays

Cells were plated in GM in chambered coverglasses (Nunc, Rochester, NY, USA; eight wells/coverglass) at 6–60 × 104 cells/well and incubated for 3–24 h at 37°C in 5% CO2. Ca2+-sensitive dyes Fura-2-AM or Fluo-3-AM were loaded by incubation at 4 μM in Ca2+ assay buffer for 30 min. Caged InsP3 was loaded as a membrane-permeable PM ester at 3 μM simultaneously with Fluo-3-AM. Cells were then equilibrated in fresh Ca2+ assay buffer for 30–45 min prior to assay. Light exposure was minimized during sample preparation. Coverslips were transferred to a microscope stage incubator (Bionomics System, 20/20 Technology, Inc., Wilmington, NC, USA) and kept at 37°C throughout the experiment. Imaging was performed on a Nikon TE-300 fluorescence microscope with a Photometrics HQ2 camera. Illumination was by a 300 W Xe lamp (Sutter Instrument, Novato, CA, USA) with filter/shutter (Lambda 10-3, Sutter Instrument) and dichroic (Conix Research Inc., Springfield, OR, USA) controllers. SimplePCI software was used to control collection parameters and to extract fluorescence intensity data for individual cells. Calibration methods were as described above and were calculated on an individual cell basis. A cytoplasmic Kd of 390 nM was used for Fluo-3 conversion of intensity data to Ca2+ concentration. Responses were calculated for each cell and, for transiently transfected samples containing mixed populations of positive and negative transfectants, average peak-offset responses were calculated for each subpopulation, and the ratio of positive/negative responses was determined.

During Fura-2 recordings, illumination was minimized to prevent photobleaching, and image pairs were collected every 2–4 s. Cells were loaded with Fluo-3 and exposed to Fluo-3 excitation wavelength light for minimum duration/image (∼7 ms). To induce photolysis, samples were exposed to DAPI excitation wavelength (360±20 nm) light for bursts of 1 s every 4 s during the photolysis period. Samples were imaged during a 40-s baseline of Fluo-only light, a 60-s photolysis period of alternated DAPI and Fluo light, and a 60-s recovery period of Fluo-only light. Control samples without caged InsP3 loading were imaged similarly to confirm that measured Fluo responses were a result of InsP3 release.

Assessment of the efficacy of cytoskeletal perturbations

The efficacy of dominant-negative forms of Rho was assessed by their effect on formation of membrane blebs in response to ATP, a P2X7R and Rho-dependent phenomenon in RAW264.7 cells [47]. Briefly, RAW264.7 cells were transiently transfected with the T19N version of CFP-labeled Rho or CFP control protein and replated into assay chambers 24 h following transfection. In parallel, cells were treated with C3 exotoxin (1 μg/ml) or control medium for 24 h and then replated into assay chambers. Cells were stimulated with ATP (3.5 mM) for 5 min, fixed with paraformaldehyde, and evaluated for the formation of membrane blebs and/or CFP expression.

The efficacy of cytoskeletal inhibitors was assessed by using F-actin or microtubule-dependent assays. Macropinocytosis is dependent on F-actin, Rac/Cdc42, PLC, and PI3K [48], and was measured by internalization of FITC-dextran as described previously [49]. Briefly, cells were grown overnight in 12-well TC plates prior to assay. The medium was replaced with HBSS with 25 mM HEPES (HBSS), 0.1% BSA, pH 7.4, and cells were treated with cytochalasin D (10 μM) or DMSO (0.1% as control) for 30 min prior to stimulation with MCSF (1.25 nM) in the presence of 1 mg/ml FITC-dextran (150 kDa MW) for 10 min. Samples were then washed and harvested at 4°C for analysis by flow cytometry. Cell morphology, adhesion, and spreading are influenced by F-actin cytoskeletal integrity, and spreading of RAW264.7 cells was assessed by phase-constrast microscopy. Cells were grown overnight in six-well TC plates prior to assay. The medium was replaced with HBSS, 0.1% BSA, pH 7.4, before the cells were treated with cytochalasin D (10 μM), latrunculin A (5 μM), jasplakinolide (5 μM), or DMSO (0.2%) for 30 min and then evaluated morphologically on a phase-contrast microscope. Cell retraction, as evidenced by the presence of rounded cells with residual filopodial extensions, was quantified. Cell-cycle progression and culture expansion are dependent on microtubule function during mitosis, so RAW264.7 culture growth and cell-cycle distribution were assessed by cell-counting and cytometric analysis of DNA content. Briefly, cells were plated into six-well TC plates in the presence or absence of colchicine (25 μM), nocodazole (33 μM), or DMSO (0.1%) and cultured for 48 h. Cells were harvested, counted, fixed with paraformaldehyde, RNase-treated, and stained with propidium iodide for cell-cycle analysis by cytometry. The distribution of cells in G1, S, and G2/M phases of the cell cycle was calculated from the resulting histograms.

SDS-PAGE and Western blot analysis of phosphoproteins

Cells were plated in 35 mm non-TC dishes at 106 cells/dish in 3 ml medium and cultured overnight prior to assay. Medium was replaced with assay medium (DMEM with 2 mM glutamine, 20 mM HEPES, 0.01% BSA) 1 h before stimulation. Ligands were added in 1/10 vol, the well contents mixed gently and cells incubated for the indicated times at 37°C. For FcγRI cross-linking, cells were incubated with 5 μg/ml mouse IgG2a for 10 min prior to addition of 44 μg/ml F(ab′)2 goat anti-mouse IgG. At the end-point, the buffer was aspirated, the cells were scraped into reducing Laemelli sample buffer, and the samples were heated for 3 min at 95°C. SDS-PAGE gel lanes were loaded with ∼20 μg protein/lane. Western blots of kinases were probed with antibody mixes containing anti-P-AKT or anti-P-ERK and anti-Rho-GDI. Western blots of PLCγ were probed with anti-PLCγ2 or anti-P-PLCγ2 (expression of PLCγ2>>PLCγ1 in macrophages). Signals from P-AKT and P-ERK were normalized to Rho-GDI signals in each lane to account further for loading variations. Levels of P-PLCγ2 were normalized to total PLCγ2 in each sample, detected in lanes blotted in parallel. Further details of SDS-PAGE and Western blotting are in AfCS Protocols PP00000168 and PP00000181.

InsP3 assay

One hour prior to assay, culture medium was replaced with HBSS, 0.5% BSA, pH 7.4. Cells were stimulated by addition of warmed ligands in 1/10 vol, and, after indicated time periods, dishes were transferred to ice, the buffer aspirated, and the cells washed with ice-cold PBS. Cells were scraped into 125 μl 5.4% perchloric acid solution and transferred to siliconized microfuge tubes on ice. After 20 min, samples were centrifuged at 14,000 g for 15 min at 4°C. Supernatant (120 μl) was neutralized with 5 N KOH containing 60 mM HEPES, and the samples were recentrifuged at 14,000 g for 15 min at 4°C. Samples were stored at −80°C prior to assay of InsP3 content using an Amersham (Piscataway, NJ, USA) InsP3 [3H] Biotrak assay kit, according to the manufacturer’s instructions. Results were reported as pmoles InsP3/100 μl cell lysate and normalized to control samples in each assay.

Phosphatidylinositol measurements

Measurement of cellular PIP content was performed by mass spectrometric analysis of samples prepared from cells by a modified, acidified Bligh/Dyer extraction method [50, 51]. One hour prior to cell assay, culture medium was replaced with HBSS, 0.5% BSA, pH 7.4. Cells were stimulated by addition of warmed ligands in 1/10 vol, and, after different time periods, dishes were transferred to ice, the buffer aspirated, and the cells harvested with ice-cold PBS with 2 mM EDTA. Cells were transferred to tubes and pelleted at 2500 g for 5 min. Pellets were extracted with 400 μl ice-cold CHCl3:CH3OH (1:1) with vortexing for 1 min, and the resulting suspension was centrifuged at 7500 g for 5 min at 4°C. The supernatant was discarded, and the pellet was dissolved in 200 μl CHCl3:CH3OH (2:1) containing 0.25% concentrated HCl with vortexing for 5 min at 4°C. The suspension was transferred to a clean microfuge tube, 40 μl 1 N HCl was added, and the sample was vortexed for 15 s. The phases were then separated by brief centrifugation, and the lower phase was evaporated and redisolved in 55 μl CHCl3:CH3OH:H2O (1:1:0.3). Samples were stored under Argon at −80°C until analysis. Just prior to analysis, 5 μL 300 mM piperidine was added. Samples were analyzed on an Applied Biosystems (Foster City, CA, USA) 4000 QTrap mass spectrometer, and PIP species were quantified relative to a di-C8 PIP2 internal standard (16:0 PIP2, Avanti Polar Lipids), as described [50]. The fatty acids are designated by two numbers—xx:y—where xx is the total number of carbon atoms, and y is the number of double bonds. PIP species are reported as 36:2 and 38:4 in mono-(PIP) and bisphosphate (PIP2) forms.

Rho activation assay

The activation state of Rho was measured by using a G-LISA™ kit from Cytoskeleton, Inc., according to the manufacturer’s protocol. This kit uses a microplate-bound Rhotekin Rho-binding domain to capture GTP-loaded Rho from cell lysates, and a HRP-coupled polyclonal antibody to RhoA, -B, and -C to detect captured protein by colorimetric assay. RAW264.7 cells were grown overnight in six-well plates, and, 1 h prior to assay, culture medium was replaced with HBSS, 0.5% BSA, pH 7.4. Cells were stimulated by addition of warmed ligands added in 1/10 vol, and, after different time periods, dishes were transferred to ice, the buffer aspirated, and the cells washed with ice-cold PBS. Lysates were prepared, protein concentrations measured, and lysate containing equal amounts of protein assayed immediately for Rho-GTP content. Similar assays were performed using 293T cells as positive controls.

Statistical analyses

The error bars in the graphs depict the sem. The statistical significance of each comparison was evaluated by performing a Student’s t-test or a one-way ANOVA, followed by Dunnett’s test or individual t-tests (with Bonferroni correction). A P value of <0.05 was considered significant.

RESULTS

The action of C. difficile ToxB differentially affects GPCR Ca2+ responses in RAW264.7 macrophages: Gαi-linked receptor activity is potentiated, and Gαq-linked receptor activity is inhibited

Our laboratory has been exploring the mechanisms of synergistic Ca2+ signaling in response to dual ligand stimulation of GPCRs on macrophages [49]. In these cells, simultaneous stimulation by C5a and UDP elicits a synergistic response. To examine mechanisms of this effect, we determined the effect of C. difficile ToxB, which is known to inhibit Rho family GTPases [6, 8, 52]. ToxB did not alter synergy between these GPCR ligands, but it had sharply distinct effects on the individual Ca2+ responses to C5a and UDP; the Ca2+ response to C5a was enhanced robustly by ToxB, and the Ca2+ response to UDP was reduced substantially (Fig. 1, A–C). The effect of the toxin was apparent after 4 h, but it increased with exposures to 24 h. A similar enhancement of C5a and depression of UDP Ca2+ responses were observed in J774 cells (Fig. 1D). There was no acute effect of the toxin alone on Ca2+ levels nor change in baseline Ca2+ after prolonged exposure (data not shown).

Figure 1.

Figure 1.

C. difficile ToxB differentially affects Ca2+ responses to C5a and UDP in RAW264.7 cells. (A–C) RAW264.7 cells were treated with ToxB (5–100 ng/ml) or control medium for 4–22 h prior to assay. Ca2+ assays were performed on Fura-2-loaded adherent cells with (A) 30 nM C5a or (B) 500 nM UDP, applied after a 40-s baseline determination. HBSS served as the buffer control. Lines show the average of n = 4–5 replicate samples from one representative assay of n = 6 with similar results. (C) Ligand specificity of ToxB (TxB) Ca2+ response phenotypes in RAW264.7 cells. RAW264.7 cells or transfectants expressing the Ro2 receptor were treated with ToxB (100 ng/ml) or control medium for 4 h prior to assay. Cells were stimulated with 500 nM UDP, 500 nM UTP, 5 nM PAF, 500 nM LPA (∼EC50), 30 nM C5a, or 500 nM spiradoline (Spir; ∼EC100). Responses were measured by the integrated area under the peak (difference of [Ca2+]i from basal) over 0–2.5 min after ligand addition and reported as the ratio of ToxB responses to control responses. The dotted line corresponds to a ratio of 1.0, i.e., no change with ToxB treatment. Values are mean ± sem from n = 3–6 assays; *, P < 0.05. (D) Ligand specificity of ToxB Ca2+ response phenotypes in J774 cells. Values are mean ± sem from n = 3 assays.

C5a signals through Gαi family G-proteins [49], and ToxB also enhanced signaling through the Gαi-linked κ opioid receptor Ro2 [53], stimulated with spiradoline (Fig. 1C). UDP signals through Gαq-coupled receptors in macrophages [49], and ToxB also depressed Ca2+ signaling by UTP, LPA, or PAF GPCRs, each of which signals predominantly through Gαq in macrophages [49] (Fig. 1C). The similar effect of ToxB on multiple receptors coupled to Gαi- versus Gαq-linked pathways suggests that it perturbs Ca2+ signaling at steps common to each respective pathway rather than in a receptor-specific manner.

Signaling through C5a invokes an early monophasic Ca2+ response in RAW264.7 cells, and signaling through UDP causes an early Ca2+ peak and a prolonged, sustained phase. For C5a, ToxB enhanced the early response and introduced a prolonged, sustained phase. In the case of UDP, ToxB inhibited both phases of the Ca2+ response, and UDP Ca2+ responses were often inhibited by ToxB to levels below those of C5a (Fig. 1, A and B), indicating that the defect is not merely a result of a reduction in general cellular Ca2+ signaling capacity. These opposing effects of ToxB on signaling by Gαi- versus Gαq-linked receptors in RAW264.7 cells led us to examine mechanistic differences in the Ca2+ signaling pathways by these different groups of GPCRs and to define how these are modulated by the pathogenic toxin.

Loss of Rho activity accounts for the inhibition of UDP Ca2+ responses by ToxB in RAW264.7 cells but not for the enhancement of C5a responses

A known principal target for ToxB is the Rho family of small GTPases, which are inhibited by glucosylation of residues Thr35 or Thr37. There are more than 20 members of the Rho family [54, 55], and ToxB inactivates several members selectively: RhoA, -B, -C, and -G, Rac1, -2, and -3, Cdc42, and TC10 but not RhoD or RhoE [8, 52]. Rho, Rac, and Cdc42 isoforms have been implicated in mechanisms of signal transduction for Ca2+ responses. In different systems, enhancement by Rho of Ca2+ responses has been attributed to the activation of PLCε [34, 56], the production and availability of PIP2 [25,26,27,28,29,30], and/or the assembly of Ca2+ signaling complexes by cytoskeletal scaffolding functions [18, 23, 24, 57,58,59]. Thus, loss of Rho activity could explain the reduced Ca2+ responses to UDP in RAW264.7 cells. To test this, we first intoxicated RAW274.7 cells with C. botulinum C3 exotoxin, which inactivates Rho specifically but not Rac or Cdc42. As with ToxB, C3 exotoxin caused a reduction in the peak and sustained levels of intracellular Ca2+ in response to UDP (Fig. 2A), but it did not alter responses to C5a (Fig. 2B). The peak and sustained Ca responses to UDP were reduced by ∼40% for all doses tested. These results are consistent with the hypothesis that ToxB reduces the Ca2+ response to UDP by inhibiting Rho, but they imply that inhibition of Rho does not enhance Ca2+ signaling in response to C5a.

Figure 2.

Figure 2.

In RAW264.7 cells, inactivation of Rho inhibits Ca2+ responses to UDP but not C5a. Rho family members were targeted with C3 exotoxin (C3 exo) and dominant-negative isoform constructs, and the Ras superfamily was broadly addressed with methyltransferase inhibitors. (A and B) RAW264.7 cells were treated with C3 exotoxin (10 μg/ml) or control medium for 20 h prior to assay. Ca2+ assays were performed on Fura-2-loaded adherent cells with 500 nM UDP (A) or 30 nM C5a (B) applied after a 40-s baseline determination. (C and D) RAW264.7 cells were treated with a cell-permeable version of C3 exotoxin (1 μg/ml) or control media for 20 h prior to assay of the Ca2+ response to UDP (C) or C5a (D). (E) RAW264.7 cells were treated with varied concentrations of cell-permeable C3 exotoxin for 4 h or 20 h prior to assay of the Ca2+ response to UDP. Values are mean ± sem of n = 5 samples from a representative assay; *, P < 0.05. (F) Ca2+ responses to 500 nM UDP, 30 nM C5a, 500 nM LPA, 5 nM PAF, or 100 nM spiradoline were assessed by peak-offset feature (normalized to control responses for each ligand). Values shown are mean ± sem from n = 4–10 assays; *, P < 0.05. (G) RAW264.7 cells were transiently transfected with dominant-negative (T19N) versions of CFP-labeled RhoA, -B, or -C versus GFP or CFP as controls. Cells were replated into assay chambers 24 h following transfection, and Ca2+ assays were performed on Fura-2-loaded adherent cells 48 h post-transfection. Ligands were 25 μM UDP or 30 nM C5a, applied after a 30-s baseline determination. Responses of transfectants (fluorescent protein-positive) were quantified by peak offset and normalized to the response of nontransfectants (fluorescent protein-negative) within the same sample. Transfection efficiencies were 10–40%. Results are shown as mean ± sem from n = 3 assays/condition; *, P < 0.05. (H) Fluorescence distribution and gating of cells in CFP-RhoA-T19N versus no-CFP (untransfected) samples. (I) RAW264.7 cells were transiently transfected with the T19N version of CFP-labeled RhoA dominant-negative (CFP-RhoA-DN) or CFP control protein or pretreated with C3 exotoxin (1 μg/ml) or control medium for 24 h and then plated into assay chambers. Cells were stimulated with ATP (3.5 mM) for 5 min, fixed with paraformaldehyde, and evaluated for presence of membrane blebs and/or CFP. Values are shown as percent cells positive for membrane blebs, indicated as mean ± sem of n = 3–10 samples in a representative assay of two with similar results; *, P < 0.001. (J) RAW264.7 cells were treated with cell-permeable C3 exotoxin (1 μg/ml) or control medium for 24 h, with or without ToxB at 50 ng/ml for the final 4 h. Ca2+ assays were then performed, and peak-offset responses to 5 nM C5a were calculated. Results are shown as mean ± sem from n = 6 assays per condition; *, P < 0.01. (K) RAW264.7 cells were treated with the methyltransferase inhibitors FTA (40 μM) and AFC (30 μM) for 4 h prior to stimulation in Ca2+ assays, which were performed on Fura-2-loaded adherent cells with 500 nM UDP or 30 nM C5a, applied after a 40-s baseline determination. The peak-offset feature of the responses was quantified and presented as the mean ± sem from four assays with four samples/assay; *, P < 0.01.

C3 exotoxin enzymatically inactivates Rho by ADP ribosylation of Asn41 residues in the switch I region [60], which reduces Rho GTP-loading, as a result of increased association with Rho-GDI in the cytosol [61], and interferes with effector activation by the GTP-bound G-protein [62]. The substrates of C3 transferase exotoxin are limited to Rho isoforms A, B, and C, and other Rho or Ras family members (such as Rac, Cdc42, Ras, or Rap) are not targeted [63,64,65]. As the permeability of the cell membrane to native C3 exotoxin is low, effective enzymatic inhibition required incubation of cells with relatively high concentrations of exotoxin for prolonged periods (e.g., 10 μg/ml overnight). Therefore, we also tested a membrane-permeable version of C3 exotoxin. As shown in Figure 2, C and D, this resulted in a similar reduction in the Ca2+ response to UDP but not to C5a, acting in as little as 4 h and with as little as 1 μg/ml exotoxin (Fig. 2E). Inhibition of the UDP response was dependent on toxin dose and on time, and the maximum inhibition observed was ∼75% (Fig. 2E). The effect of Rho inhibition on UDP signaling was not a result of a reduced maximal Ca2+ signaling capacity in toxin-treated cells, as responses to low doses of UDP, which approximate the response to C5a, were inhibited to levels well below those of C5a, which was unaffected by C3 exotoxin (Fig. 2, C and D). Thus, C3 exotoxin yielded the same pattern of inhibition as seen with ToxB; Gαq-linked ligand responses were reduced, and Gαi-linked ligands were unaffected (Fig. 2F). To confirm and to characterize the Rho involvement in Ca2+ responses to UDP but not C5a, we performed assays on RAW264.7 cells transiently transfected with dominant-negative forms of Rho. As shown in Figure 2G, CFP-tagged T19N mutants of RhoA, -B, and -C isoforms yielded reduced Ca2+ responses to UDP but not to C5a. Responses of CFP expressors were normalized to nonexpressors in the same sample for optimal sensitivity (Fig. 2H). Expressed RhoA-T19N was confirmed to inhibit membrane bleb formation in response to ATP (Fig. 2I), a phenotype demonstrated to be mediated by P2X7R in RAW264.7 cells in a Rho-dependent manner [47]. RhoA, -B, and -C have overlapping and distinct activities [66]. The dominant-negative forms of Rho act by inhibiting Rho-associated GEFs, and RhoA, -B, and -C share several GEFs [55, 67], as do additional members of the Rho family. Thus, the Rho isoform specificity, if any, is not defined by these experiments, but a factor or activity common to these isoforms is implicated. C3 exotoxin and dominant-negative Rho isoform experiments support a role for RhoA-like proteins. In contrast, transient transfection of RAW264.7 cells with dominant-negative constructs of Rac1 or Cdc42 failed to affect the UDP or C5a responses (data not shown), confirming that these members of the Rho family, unlike RhoA-like proteins, do not regulate these Ca2+ responses.

In some systems, ToxB intoxication can lead to increased expression of RhoB, and when RhoB exceeds the inhibitory capacity of the toxin, this can yield a contradictory increase in net RhoB activity [68, 69]. RhoB has been implicated in the regulation of vesicle trafficking [70, 71] and receptor surface expression in macrophages [72], and it could impact GPCR Ca2+ responses. To evaluate RhoB escape from inhibition in the enhancement of C5a Ca2+ by ToxB, we combined ToxB treatment with C3 exotoxin treatment, which will maintain inactivation of RhoB by ADP-ribosylation [69]. C3 exotoxin treatment did not prevent the up-regulation of C5a Ca2+ responses (Fig. 2J), indicating that Rho does not contribute to the elevation of C5a Ca2+ responses. Additionally, expression of constitutively active RhoB did not elevate C5a Ca2+ responses (data not shown). Thus, escape of RhoB from ToxB inhibition does not appear to contribute to the elevated C5a Ca2+ responses.

Members of the Ras superfamily of small GTPases, of which the Rho family is a subset, are isoprenylated proteins that undergo carboxy-terminal methylation for activation. Inhibitors of isoprenylation or methylation disrupt the function of small GTPases by affecting subcellular localization and effector interactions [73, 74]. We used this alternative biochemical means of inhibition of Ras superfamily proteins as an independent test of their involvement in UDP versus C5a Ca2+ responses. Our results with more directed inhibitors of Rho proteins predicted that broader inhibitors of Ras proteins would similarly inhibit Ca2+ responses to UDP, and these inhibitors would help clarify if non-Rho Ras superfamily targets of ToxB contribute to the elevated C5a Ca2+ phenotype. As predicted, methyltransferase inhibitors inhibited Ca2+ responses to UDP (Fig. 2K). Effects on Ca2+ signaling were seen after as little as 30 min inhibition (data not shown), but a 4-h treatment was needed for consistently significant results [75,76,77]. Similar results were seen with FTA plus AFC (Fig. 2K) or FTA plus AGGC (data not shown). Methyltransferase inhibitors had no effect on Ca2+ responses to C5a, however, indicating that members of the Ras superfamily are not required for C5a Ca2+ signaling, nor does their inhibition induce an elevation of C5a responses.

Studies of BMDM reveal that ToxB differentially affects PLCβ3- versus PLCβ4-dependent pathways

The differential effects of ToxB on C5a and UDP Ca2+ signaling in RAW264.7 cells could reflect differential effects of ToxB on Gαi versus Gαq trimeric G-proteins themselves or, instead, events more distal in these Ca2+ signaling pathways. Previous studies from our laboratory indicated that Ca2+ signaling by C5a in both RAW264.7 and BMDM is largely dependent on PLCβ3 but not PLCβ4 [49]. Ca2+ signaling by UDP in RAW264.7, however, is largely dependent on PLCβ4, but in BMDM, UDP signaling uses both PLCβ3 and PLCβ4 [49]. In these prior studies, Ca2+ responses to UDP by BMDM were diminished by ∼80% in the absence of PLCβ3 and by ∼20% in the absence of PLCβ4, whereas in RAW264.7 cells, UDP responses were diminished by RNAi of PLCβ4 but not RNAi of PLCβ3. PLCβ2 is expressed in RAW264.7 cells and BMDM, but it appears dispensable for normal Ca2+ responses to C5a or UDP, and PLCβ1 is not expressed in either cell type [49].

To explore possible relations between ToxB and PLCβ isoforms, we examined effects of ToxB on BMDM from mice with selective genetic deletion of different PLCβ isoforms. In BMDM from wt mice, the maximal Ca2+ responses to C5a and UDP were greater than in RAW264.7 cells, and C5a responses were more robust than those of UDP; in RAW264.7 cells, UDP gave greater responses. In BMDM, the Ca2+ responses to C5a were elevated further by exposure to ToxB (Fig. 3A), as they were in RAW264.7 cells, but in contrast to RAW264.7 cells, signaling in response to UDP and other Gαq-linked receptors was enhanced rather than inhibited by ToxB (Fig. 3B), although the enhancing effect of ToxB on signaling by Gαq-linked receptors was significantly less than its enhancement of signaling by the Gαi-linked C5aR (Fig. 3C). Thus, a distinction in ToxB effect on Gαi versus Gαq Ca2+ responses persisted in BMDMs, but instead of the opposing effects seen in RAW264.7 cells, the difference was in the magnitude of enhancement.

Figure 3.

Figure 3.

In BMDM, C. difficile ToxB enhances Ca2+ responses to C5a and UDP to different extents, and C3 exotoxin inhibits Ca2+ responses to UDP but not C5a. (A–C) BMDM were treated with ToxB (10 ng/ml) or control medium for 18 h prior to assay. Ca2+ assays were performed on Fura-2-loaded adherent cells stimulated with (A) 10 nM C5a or (B) 10 μM UDP (∼EC100 s) after a 40-s baseline determination. (C) Ligand specificity of ToxB Ca2+ response phenotypes in BMDM. Cells were stimulated with 1 μM UDP, 1 μM UTP, 2 nM PAF, 1 μM LPA, 20 nM LTD4, or 1 nM C5a (∼EC50). Values shown are mean ± sem from n = 3–8 assays. (D–F) Inhibition of Rho by C3 exotoxin inhibits UDP but not C5a Ca2+ responses in BMDM, which were treated with 1 μg/ml cell-permeable C3 exotoxin for 20 h and then stimulated with 5 nM C5a (D) or 10 μM UDP (E) in Ca2+ assays. (F) Quantitation of BMDM responses by peak-offset feature. Values are mean ± sem of n = 4 samples/condition from a representative assay of five with similar results; *, P < 0.01. (G) PLCβ3 expression is required for ToxB elevation of Ca2+ responses. BMDM cultures established from mice genetically deficient (−/−) in PLBβ2, -3, -4, or -2 and -3 (2+3) were treated with or without ToxB (10 ng/ml) for 18 h and assayed for Ca2+ responses to 10 nM C5a or 10 μM UDP (∼EC100). Responses were measured by the integrated area from 0 to 2.5 min after ligand addition, and results of ToxB-treated cells were normalized to those of the corresponding untreated cell type. C5a responses in untreated PLCβ2 + 3-deficient cells were minimal and therefore not assessed (NA) for changes with ToxB treatment. Values shown are mean ± sem from 16 replicate samples/condition from five assays; *, P < 0.05.

As in RAW264.7 cells, however, inhibition of Rho using C3 exotoxin selectively inhibited UDP responses of BMDM with no effect on C5a (Fig. 3, D–F). This result suggested that in BMDM, the Rho-dependent inhibitory effects of ToxB on UDP signaling may be offset by an enhancing effect. That is, in BMDM, enhancement of UDP Ca2+ signaling by ToxB outweighs the Rho-dependent inhibition, whereas in RAW264.7 cells, inhibition by ToxB dominates. As UDP signaling in RAW264.7 cells is largely dependent on PLCβ4, and in BMDMs, it uses PLCβ4 and PLCβ3 [49], we tested the hypothesis that ToxB differentially affects PLCβ isoform pathways to Ca2+, inhibiting Ca2+ signaling through PLCβ4 but enhancing signaling through PLCβ3.

In accord with this hypothesis, ToxB failed to enhance Ca2+ responses to C5a or UDP in BMDM from mice genetically deficient in PLCβ3 (Fig. 3G). In BMDM from mice deficient in PLCβ2 or PLCβ4, ToxB still enhanced C5a and UDP responses comparably with wt BMDM. Thus, ToxB appears to promote PLCβ3-mediated Ca2+ signaling. In contrast, Ca2+ responses to UDP were inhibited by ToxB in BMDM deficient in both PLCβ2 and PLCβ3. In these mice, Ca2+ responses are dependent solely on remaining PLCβ4 (mediated by Gαq with no PLCβ1, -2, or -3 expressed). Thus, although ToxB enhances signaling by PLCβ3, it inhibits signaling by PLCβ4. This accounts for the quantitative differences in the effects of ToxB on BMDM and RAW264.7 cells. In RAW264.7 cells, UDP signals primarily through PLCβ4, and it is inhibited by ToxB. In BMDM, UDP instead signals ∼20% through PLCβ4 but ∼80% through PLCβ3. Here, enhancement of PLCβ3 signaling by ToxB dominates inhibition of PLCβ4 signaling. When PLCβ3 and PLCβ2 are absent, however, the inhibition of PLCβ4 signaling can be seen. Because RAW264.7 cells readily reveal the differential effects of ToxB, we turned to them for further studies about the mechanisms by which ToxB affects signaling.

ToxB affects the release of Ca2+ from intracellular stores in response to UDP or C5a

Changes in Ca2+ responses could reflect changes in the initial release of Ca2+ from intracellular stores, the influx of extracellular Ca2+ through SOCs, or both. Release of intracellular Ca2+ stores is a prerequisite for the influx of extracellular Ca2+, but the rapid coupling of this response to SOCs may contribute to the magnitude of the early peak of [Ca2+]i. Thus, the observed reduction in the early [Ca2+]i peak and the sustained levels of [Ca2+]i could result from deficiency in the release from Ca2+ stores alone or in SOCs coupling. To address this, we examined the effects of ToxB on Ca2+ signaling in the absence of extracellular Ca2+ so that changes in [Ca2+]i reflected only release of Ca2+ from intracellular stores. Under these conditions, ToxB again reduced the rise in [Ca2+]i elicited by UDP and increased the response to C5a (Fig. 4, A and B). ToxB did not affect the content of intracellular stores, as reflected by similar Ca2+ peaks with release of intracellular stores using thapsigargin/ionomycin (data not shown). A change in Ca2+ stores, moreover, would not alone explain the dichotomous effect of ToxB on UDP versus C5a. We conclude that ToxB differentially affects signaling pathways that alter the release of intracellular Ca2+ stores by UDP versus C5a. This indicates that ToxB affects proximal signaling steps, although it does not preclude additional effects on Ca2+ flux through SOCs.

Figure 4.

Figure 4.

ToxB affects the release of Ca2+ from intracellular stores and does not reflect a change in C5aR expression, G-protein coupling, or Src-kinase dependence. RAW264.7 cells were treated with ToxB (10 ng/ml) or control medium for 20 h prior to assay. Ca2+ assays were performed on Fura-2-loaded, adherent cells. Buffer with or without EGTA (2 mM final) was added to samples 30 s prior to addition of 25 μM UDP (A) or 30 nM C5a (B). (C) The C5a Ca2+ response remains sensitive to PTx after enhancement by ToxB. RAW264.7 cells were treated with or without PTx (50 ng/ml) and/or ToxB (10 ng/ml) for 18 h prior to Ca2+ assay. Each line represents the mean of five replicate samples. Shown are data from a representative assay of four with similar results. The mean peak-offset values were calculated for each response from these four experiments and are presented (D). Values are mean ± sem. (E) Src activity is not required for the enhancement of C5a Ca2+ responses by ToxB. RAW264.7 cells were treated with ToxB (10 ng/ml) for 20 h, and the Src inhibitor PP2 or control PP3 (10 μM) was added for 30 min prior to and during the assay. Ca2+ assays were performed with 5 nM C5a, and peak-offset responses were calculated. Shown are mean ± sem from n = 3 assays. (F) Src activity is not required for normal Ca2+ responses to C5a or UDP. RAW264.7 cells were treated with PP2 or control PP3 for 30 min prior to assay of Ca2+ responses to 5 nM C5a, 500 nM UDP, or FcγRI cross-linking. Responses to each stimulus in the presence of PP2 were normalized to those in the presence of PP3. Shown are mean ± sem from n = 3–5 assays.

Enhanced Ca2+ responses to C5a do not reflect changes in receptor expression or Gαi dependence

The effects of ToxB on Ca2+ signaling by multiple GPCRs suggested perturbation of signaling at steps common to these receptors, rather than receptor-specific changes. We nonetheless examined C5aR surface expression to determine if it was up-regulated by ToxB in conjunction with the elevation in C5a Ca2+ responses. No change in C5aR surface expression was seen with ToxB treatment (data not shown), confirming that the ToxB perturbation was distal to receptor availability.

In macrophages, signal transduction by the C5aR is mediated principally by trimeric G-proteins that are sensitive to PTx. We have shown previously, however, that a modest PTx-insensitive component exists in the Ca2+ response to high concentrations of C5a, which may represent signaling through Gα15 [49]. We therefore assessed whether ToxB might enhance C5aR Ca2+ responses by selectively enhancing PTx-insensitive pathways. It did not. In the presence of ToxB, most C5a Ca2+ signaling remained sensitive to PTx, although at high concentrations of C5a, a minor PTx-insensitive path could again be detected (Fig. 4, C and D). PTx-sensitive and insensitive components of the C5aR responses were increased by ToxB, indicating a lack of specificity of ToxB between these two pathways. These results indicate that ToxB does not alter G-protein coupling by C5aR.

The effects of ToxB are independent of src kinases and PLCγ

In some cells, activation of GPCRs can lead to transactivation of tyrosine kinase pathways and receptors and associated activation of MAPKs as well as PLCγ-dependent Ca2+ responses [78,79,80]. In particular, activation of src kinases by GPCRs [81, 82] has been identified as a critical link for GPCR responses to epidermal growth factor or vascular endothelial growth factor receptors [78, 79, 83], as well as activation of PLCγ via GPCR kinase-interacting proteins [80, 84]. In mast cells, synergy has been seen between PLCβ and PLCγ with simultaneous stimulation of PGE2 and FcεRI receptors [85]. Thus, GPCR Ca2+ responses in macrophages could reflect the net activities of PLCβ and PLCγ, and augmented GPCR Ca2+ responses could result from alteration of transactivation signaling pathways by ToxB. To evaluate the potential contribution of src kinases and PLCγ to ToxB effects, we evaluated the effect of src inhibition on Ca2+ responses and the phosphorylation state of PLCγ with ToxB treatment. There was no effect of the src inhibitor PP2 on the elevation of C5a Ca2+ signaling by ToxB nor was there a significant effect on Ca2+ responses by C5a or UDP alone, although PP2 did block FcγRI responses as expected (Fig. 4, E and F), and as expected, cross-linking of FcγRI led to phosphorylation of PLCγ, but no phosphorylation was detected in response to ToxB, with or without C5a stimulation (data not shown). Thus, we found no evidence of a contribution of these transactivation mechanisms to the ToxB effect.

Availability of PIP2 and production of InsP3 are maintained or increased in the presence of ToxB

Release of Ca2+ from intracellular stores is mediated by the opening of Ca2+ channels in response to secondary messengers, including InsP3, cyclic ADP ribose, nicotinic adenine dinucleotide, and S1P [16, 17]. PLCβ cleaves PIP2 to yield InsP3 and DAG. The activity of PLCβ is limited by the availability of PIP2, and studies in other cell systems have demonstrated previously a role for Rho in the activation of phosphatidylinositol 4 and 5 kinases, which are responsible for production of PIP2 [25, 28, 86]. To determine if ToxB causes a loss of PIP2, which could lead to reduced Ca2+ responses, we measured the availability of PIP2 in RAW264.7 cells following treatment with ToxB. ToxB treatment did not diminish the levels of PIP2 (Fig. 5A), indicating that an adequate supply of substrate is available for PLCβ prior to receptor activation. Our studies were of total PIP2, and functionally distinct pools of PIP2 have been identified that differentially contribute to GPCR Ca2+ responses [86,87,88,89]. Selective effects of ToxB on these pools could escape detection in our assays, but this explanation was excluded by the next set of experiments, in which we assessed the production of InsP3 directly.

Figure 5.

Figure 5.

Availability of PIP2 and production of InsP3 are maintained or increased following exposure to ToxB. RAW264.7 cells were treated with ToxB (10 ng/ml; A, C, E), the cell-permeable version of C3 exotoxin (1 μg/ml; D), and/or control medium (B) for 20 h prior to assay. (A) Cells were harvested and lipids extracted and quantified by mass spectrometry. Samples were spiked with a fixed mass of lipid standard, and the amounts of each PIP lipid are reported as a fraction of the standard. Shown are mean ± sem from n = 3 samples/condition from a representative assay of three with similar results. (B) Untreated cells were stimulated with 10 μM UDP, 50 nM C5a, or buffer alone (Control Baseline), and samples were harvested at the indicated times to measure cellular InsP3 (IP3) content. Shown are results from n = 11 assays. (C–E) Cells were stimulated with ligand or buffer alone (no stim) for 3 min, and samples were processed to measure cellular InsP3 content. Amounts are reported as InsP3/cell equivalent and normalized to control cells in the same assay. Values shown are mean ± sem from n = 3–6 assays with three samples/condition/assay; *, P < 0.05. (F and G) Inhibition of Rho does not alter the Ca2+ response of RAW264.7 cells to exogenous InsP3. RAW264.7 cells, with or without C3 exotoxin exposure (1 μg/ml for 20 h), were loaded with Fluo-3 and photolysable-caged InsP3-PM. Single-cell Ca2+ recordings were performed, during which cells were flashed repeatedly with 360 nM light to release InsP3 (the period indicated as “photolysis” between 40 and 100 s). (F) Representative traces of control or C3 exotoxin-treated cells showing population mean ± sem, where n = 54–128 individual cells/population. (G) The peak-offset feature of the responses was measured and normalized to the average control value for each assay. Results shown are mean ± sem with n = 8 samples/treatment from three experiments with similar results.

To determine if ToxB treatment perturbed Ca2+ signaling mechanisms proximal or distal to InsP3 production, we measured cellular InsP3 content at baseline and after stimulation with UDP or C5a. As shown in Figure 5B, a robust increase in cellular InsP3 content, peaking at ∼3 min, was observed following stimulation with UDP, and the 3-min time-point was used for subsequent analyses. When RAW264.7 cells were stimulated with UDP in the absence of ToxB, InsP3 was increased approximately threefold, and the levels of ligand-stimulated InsP3 were not reduced by ToxB or by C3 exotoxin (Fig. 5, C and D). Although similar levels of InsP3 were achieved upon stimulation with UDP, with or without ToxB treatment, baseline levels were elevated by ToxB treatment alone, and thus, a reduced change from baseline was observed. This did not, however, explain the effects of Rho inhibition on Ca2+ signaling, as C3 exotoxin had no significant effect on baseline levels of InsP3, and, like ToxB, it did not reduce levels of InsP3 in response to UDP. Thus, the inhibition of the UDP response following inhibition of Rho lies distal to the generation of InsP3.

In the absence of ToxB, a rise in InsP3 could not be detected in RAW264.7 cells following C5a (Fig. 5E), despite the dependence of the Ca2+ response on PLCβ. We presume that the activation of Ca2+ signaling by C5a reflects efficiency of coupling InsP3 to calcium stores or rapid kinetics of InsP3 that cannot be detected by our assays. Regardless, in cells treated with ToxB, C5a stimulated a robust increase in InsP3 content, which reached or exceeded that of UDP-stimulated responses (Fig. 5E). Taken together, these results indicate that ToxB inhibits Ca2+ signaling by UDP at a point distal to the production of InP3, but it enhances C5a Ca2+ signaling, at least in part, by augmenting the production of InsP3.

Inhibition of Rho in macrophages does not reduce the release of Ca2+ stores in response to InsP3

As InsP3 generation in response to UDP is intact in the presence of ToxB or C3 exotoxin, but release of Ca2+ from intracellular stores is reduced, either the sensitivity of the Ca2+-release mechanism to InsP3 is reduced, or the InsP3 that is generated in response to UDP does not reach the InsP3Rs in a concentration or a manner that allows an adequate engagement with the receptors. We tested the former possibility by measuring Ca2+ responses to exogenous InsP3, introduced by loading RAW264.7 cells with photolyzable caged InsP3. Single-cell imaging was used to monitor changes in [Ca2+]i following release of the InsP3 by a photolytic light impulse. As shown in Figure 5F, the responses included a rapid primary peak in [Ca2+]i, followed by a sustained phase during the maintained photolytic exposure. Upon discontinuation of photolytic exposure, [Ca2+]i levels began a return to baseline. Levels of [Ca2+]i were similar in cells treated with C3 exotoxin compared with controls (Fig. 5G), suggesting no loss in sensitivity of intracellular stores to InsP3 when Rho is inhibited. As levels of InsP3 are not reduced by ToxB, and sensitivity to InsP3 is not reduced by inhibition of Rho, we conclude that the loss in Ca2+ signaling in response to UDP reflects a modification of factors that promote the local engagement of InsP3 with its receptors or a change in the intracellular distribution of released InsP3, such that it is not available to the InsP3Rs.

Cytoskeletal organization is not necessary for coupling of InsP3 to Ca2+ release in response to UDP

In several cell systems, the relative proximity of InsP3 generation sites to sites of intracellular Ca2+ store release affects the magnitude of receptor-stimulated Ca2+ responses. In particular, the organization of signaling complexes by protein–protein interactions is thought to regulate differences in receptor localization in neurons and fibroblasts [18, 23, 24]. Clustering of GPCRs, PLCβ, transient receptor potential channels, shank, and InsP3Rs, among other factors, provides efficient linkage of InsP3 to InsP3Rs on the ER. Differential formation of these Ca2+ signaling complexes (“Ca2+ signalosomes”) may thus augment Ca2+ responses in a manner that amplifies proximal signaling and InsP3 generation. Scaffolding proteins that affect these clusters include F-actin and Homer. Rho is an important regulator of the F-actin cytoskeleton, and it interacts with Homer. Thus, inhibition of Rho by ToxB could disrupt Ca2+ signalosome formation.

To evaluate possible cytoskeletal contributions to UDP signaling, we tested inhibitors of microfilament organization. Treatment of RAW264.7 cells with cytochalasin D did not alter the UDP Ca2+ response (Fig. 6A), although it inhibited macropinocytosis (Fig. 6B) and phagocytosis (data not shown) and had evident changes on cell morphology (acute reduction in cell spreading within 5 min of addition and maintained throughout the Ca2+ assay period; Fig. 6C). Similarly, other inhibitors of microfilament organization (latrunculin A, jasplakinolide) or of microtubule organization (colchicine, nocodazole) did not alter UDP Ca2+ responses (Fig. 6A), despite confirmation of their efficacy: Latrunculin A and jasplakinolide caused acute reductions in cell spreading (Fig. 6C), and colchicine and nocodazole treatment arrested cell growth at the G2/M phase of the cell cycle (Fig. 6D). These results indicate that such cytoskeletal organization is not required for maintenance of the Rho-dependent coupling of InsP3 to Ca2+ release following UDP signaling. These data indicate that some of the actin-dependent scaffolding functions identified in previous studies [58, 90,91,92,93,94,95,96] do not contribute to Ca2+ signaling mechanisms of the macrophage receptors tested in these cells.

Figure 6.

Figure 6.

Disruption of cytoskeletal organization does not affect UDP Ca2+ responses, and Rho is not activated by UDP or C5aR stimulation in RAW264.7 cells. (A) RAW264.7 cells were treated with cytochalasin D (CytoD; 10 μM), latrunculin A (LatA; 5 μM), jasplakinolide (Jaspl; 5 μM), colchicine (Colc; 25 μM), nocodazole (Noco; 33 μM), or respective control buffers (low percent DMSO or ethanol) for 30 min prior to measurement of Ca2+ responses to 500 nM UDP. Similar results were observed for cytochalasin D after treatment for 5 min or 4 h (data not shown). Results shown are the mean ± sem from n = 10–15 replicates from two to five assays/condition. Controls for these results are in panels B–D. (B) Macropinocytosis was assessed in cells treated with cytochalasin D or DMSO (0.1%) as control for 30 min prior to assay. Values shown are normalized to the uptake of FITC-dextran by control samples. (C) Cell morphology was evaluated after 30 min treatment with the inhibitors shown. Values shown are percent of cells with a retracted morphology (mean±sem). (D) Cell-cycle distribution of cells treated with inhibitors overnight. Values shown are percent of cells in phases G1/S versus G2/M (mean±sem). Cell number was reduced by ∼80% (not shown). (E) UDP does not activate Rho. Normal RAW264.7 cells were serum-starved for 1 h and then stimulated with 25 μM UDP, 30 nM C5a, 2.5 μM LPA, or 1 μM S1P in HBSS-BSA. Ligand activation of Rho was assessed at 1 min by G-LISA. Activation of Rho by UDP stimulation was evaluated further by (F) time course and (G) dose response measured at 1 min.

The requirement for Rho in UDP-induced calcium responses does not reflect activation of Rho by UDP

In our studies, the inhibition of Rho by ToxB or other agents required pretreatment of cells. Thus, the requirement for Rho in the Ca2+ response to UDP could involve activation of Rho by UDP or instead, depend only on prior or concurrent Rho activity derived from other stimuli. We examined the activation of Rho by UDP or C5a in RAW264.7 macrophages and found no detectable change in Rho activity in response to either ligand (Fig. 6E). In contrast, Rho was activated robustly by LPA or S1P, which activate Gα12/13 as well as Gαq in RAW264.7 cells [97]. To ensure that we examined a broad range of conditions for activation of Rho by UDP, we analyzed the time course and the dose response of Rho to UDP. UDP did not increase Rho activation in RAW264.7 macrophages under any circumstances tested (Fig. 6, F and G), although UDP did activate Rho in 293T cells (data not shown), as reported by others [98]. We conclude that although Rho is important for the Ca2+ response to UDP, this requirement is likely met by baseline levels of Rho activity derived from alternate stimuli rather than by UDP-mediated augmentation of Rho activity upon GPCR stimulation.

In sum, with regard to PLCβ4-dependent UDP responses, ToxB inhibition of Rho reduces calcium signaling by interrupting steps between the generation of InsP3 and the recognition of InsP3 by the InsPRs. In contrast, ToxB enhancement of calcium signaling by PLCβ3-dependent C5a involves the generation of increased InsP3, and it thus acts at a different step or steps in the signaling cascade, perhaps at PLCβ3 itself. We therefore examined further pathways in C5a signaling that are altered by ToxB.

The increase in Ca2+ responses to C5a in the presence of ToxB is dependent on PLA2 activity

ToxB is known to have effects on cellular physiology beyond inhibition of Rho family small GTPases. The Rho-inhibitory effects have been distinguished from other cytopathic effects through the use of selectively resistant cell lines and by contrasting the kinetics of different cellular responses [99, 100]. ToxB and ToxA activate PLA2, which cleaves the sn-2 bonds of phospholipids to release AA and other lysophospholipids, resulting in the production of PGE2 and LTB4 [99,100,101]. These have direct cytopathic effects on enterocytes, and they contribute to the pathogenesis of pseudomembranous colitis by signaling the release of chemotactic factors for neutrophils and macrophages [2, 14, 102]. We therefore tested the role of cPLA2 in the elevation of C5a-stimulated Ca2+ signaling by ToxB. We first tested three different inhibitors of cPLA2: AACOCF3, MAFP, and quinacrine. Inclusion of any of these compounds before and during ToxB treatment blocked the elevation of C5a-stimulated Ca2+ above that of control cells (Fig. 7, A–C, and data not shown), implicating a product of cPLA2 in the up-regulation of the Ca2+ response to C5a. Although the specificity of any inhibitor may limit interpretation of such results, subsequent experiments also supported a role for AA metabolism in the effect of ToxB on C5a Ca2+ responses.

Figure 7.

Figure 7.

Elevation of Ca2+ responses by ToxB is dependent on AA metabolism. (A and B) RAW264.7 cells were treated with 20 μM AACOCF3 (inhibitor of PLA2) or vehicle control 1 h before and during a 4-h treatment with 100 ng/ml ToxB. Ca2+ responses to 30 nM C5a were then assessed in the continued presence of the inhibitors. (A) Each line represents the mean of five replicate samples/condition. (B) Quantitation of the responses by peak-offset feature. Values are mean ± sem from n = 3 assays; *, P < 0.05. (C) Similar experiments were performed using the PLA2 inhibitor MAFP at 5 μM. Values shown are mean ± sem of n = 5 replicate samples/condition from a representative assay of six with similar results; *, P < 0.05. (D) Elevation of Ca2+ responses by ToxB is dependent on the activity of 5-LOX and not COX. RAW264.7 cells were treated with the COX inhibitor indomethacin (Indo; 50 μM) or the LOX inhibitors NDGA (33 μM), AA861 (15 μM), or MK-886 (MK; 10 μM) or vehicle control 1 h before and during a 4-h treatment with 100 ng/ml ToxB. Ca2+ responses to 30 nM C5a were then assessed in the continued presence of the inhibitors. Peak-offset responses were quantified. Shown are the means ± sem from n = 4–8 assays/condition; *, P < 0.01. (E and F) Exposure of RAW264.7 to AA elevates C5a Ca2+ responses. RAW264.7 cells were cultured in the presence or absence of 10 μg/ml AA for 20 h prior to Ca2+ assay. Cells were then loaded with Fura-2 and assayed for Ca2+ responses to 30 nM C5a. (E) Results from an individual assay, in which each line represents the mean ± sem of four replicate samples/condition. (F) Values shown are mean ± sem from n = 8 assays; *, P < 0.01. (G) Elevation of Ca2+ responses by AA or ToxB is dependent on LOX activity. RAW264.7 cells were cultured in the presence or absence of NDGA (33 μM) for 30 min prior to addition of 10 μg/ml AA for an additional 4 h prior to Ca2+ assay. Cells were then loaded with Fura-2 and assayed for Ca2+ responses to 30 nM C5a in the continued presence or absence of NDGA.

5-LOX activity is required for ToxB elevation of Ca2+ responses

AA can undergo lipoxygenation, the first step in the formation of LTs, or it may be converted by COX to PGs. LT and PG synthesis in RAW264.7 cells are stimulated by TLR ligands and certain Ca2+ agonists [103,104,105,106]. Prior studies have shown that untreated RAW264.7 cells express 5-LOX and FLAP [103]. We similarly found transcripts for 5-LOX and FLAP in our RAW264.7 cells but not transcripts for 12- or 15-LOX (data not shown). To assess the role of these pathways in the action of ToxB on Ca2+ signaling, RAW264.7 cells were treated with indomethacin to inhibit COX or with NDGA to inhibit LOX, and Ca2+ responses were then assessed. As seen in Figure 7D, indomethacin failed to inhibit the elevation of C5a Ca2+ responses, but NDGA blocked the ToxB-induced elevation of responses effectively. Two other 5-LOX-specific inhibitors, AA861 and MK-886, were similarly effective in blocking the ToxB-induced elevation of responses. Thus, the 5-LOX pathway appears necessary to augment C5a Ca2+ responses.

As AA is released by the action of PLA2, we next tested whether the addition of AA would mimic the effect of ToxB in promoting a Ca2+ response. RAW264.7 cells were loaded with AA in culture overnight [44] and then stimulated with C5a. This exposure yielded elevated C5a Ca2+ responses (Fig. 7, E and F), similar to those observed with ToxB treatment. To determine if this AA effect shared a similar requirement for LOX activity, we treated cells with NDGA prior to and during AA exposure. NDGA inhibited the elevation of C5a Ca2+ responses induced by AA exposure, comparable with its inhibition of ToxB effects. Thus, AA released by cPLA2 activity associated with ToxB treatment may be sufficient to induce the changes in GPCR Ca2+ signaling observed. In all, our results support the hypothesis that ToxB enhances Ca2+ responses to C5a in a manner that is selective for PLCβ3 and that is dependent on the generation of the LTs but not PGs.

DISCUSSION

We have demonstrated for the first time selective and opposing effects of C. difficile ToxB on different GPCR signaling pathways. The results reveal notable differences in the regulation by ToxB of PLCβ3 versus PLCβ4 pathways, and they open new areas for study in the regulation of the clinical effects of ToxB. ToxB effects have often been attributed to its known inhibition of Rho family GTPases. In our studies, ToxB effects on PLCβ4-dependent Ca2+ signaling pathways reflected a requirement for Rho, and the contribution of Rho was shown to be at the level of InsP3/Ca2+ coupling. In contrast, ToxB enhanced Ca2+ signaling by PLCβ3-dependent pathways in a manner that was independent of Rho, and it depended instead on cPLA2 and lipoxygenation.

The effects of ToxB on PLCβ4-dependent Ca2+ signaling reveal new requirements for RhoA-like proteins in this pathway. Some of the mechanisms described previously by which Rho family members may regulate Ca2+ responses are associated with a defect in proximal signaling, yielding a reduction in net InsP3 production. In some studies, this reflected a reduction in cellular PIP2 supply [25, 107, 108]. In our studies, however, PIP2 was sustained in ToxB-treated cells, as was InsP3 production, and thus, the activity of PLCβ4 itself appears unaffected. We also observed normal p38 activation by UDP (data not shown), further supporting maintenance of proximal signaling by the UDP GPCR in the presence of ToxB. The distal signaling step of InsP3-stimulated release of Ca2+ from intracellular stores was also unaffected. Thus, the contribution from Rho appears distal to InsP3 production and proximal to release of Ca2+ from intracellular stores. A reduction in exposure of the ER to generated InsP3 could explain this uncoupling of InsP3 from Ca2+. Our findings indicate that a RhoA-like protein is required for coupling of responses through PLCβ4.

Full inhibition of UDP Ca2+ signaling in RAW264.7 cells by ToxB required pretreatment for ≥4 h; shorter treatments were less effective. Coupled with the observations that the effect of ToxB on UDP Ca2+ is dependent on Rho, but UDP does not activate Rho, these results imply that Rho intrinsically maintains cellular mechanisms for coupling PLCβ4-mediated InsP3 signaling to InsP3Rs in the ER. The baseline activity of Rho derived from serum factors, adhesion signals, and other stimuli [109, 110] under normal culture conditions may thus provide for normal Ca2+ responses.

Reduced coupling of InsP3 production to Ca2+ release from intracellular stores for a distinct subset of GPCRs suggests a change in the location of a relevant component as the most likely explanation for the effects of ToxB via Rho [18, 23, 24, 92, 111]. We have attempted to identify such changes in the distribution of the sites of production of InsP3 (GPCR, Gα, PLCβ) in relation to the sites of action of InsP3 (InsP3R, ER), but our studies have not revealed distinct distributions or consistent changes so far. Some gross morphological changes occur with ToxB exposure, but these are not reproduced by inhibition of Rho through other means, which similarly perturb Ca2+ responses, so these morphologic changes appear unrelated to effects of ToxB on Ca2+ signaling. Nonetheless, as UDP-stimulated InsP3 production is normal in the presence of ToxB, and exogenous InsP3 stimulates normal Ca2+ release in the presence of ToxB, differences in the intracellular distribution of InsP3 likely account for the effects of ToxB on UDP signaling. Although the cytoskeleton plays an important role in organization of Ca2+ signaling complexes for some Gαq-linked GPCRs in neurons [24, 92], our data suggest a lesser contribution from F-actin to signaling by UDP receptors in macrophages. In sum, we have narrowed the mechanism for the effects of ToxB on UDP signaling to disruption of the delivery of InsP3 signals leading to the release of Ca2+ from the ER, but the mechanism of this disruption remains to be explored.

A contribution of RhoA-like proteins, but not Rac or Cdc42, to the ToxB Ca2+ phenotypes is supported by the selectivity of the inhibitors used. Although inhibition of Rho reproduced the UDP phenotype, inhibition of Rac, Cdc42 or the broad Ras superfamily did not reproduce the effect of ToxB on the C5a phenotype. An inverse relationship can exist between the activities of some family members, e.g., Rho and Rac [112, 113], but our methods, which did not prevent reciprocal up-regulation, failed to elevate C5a Ca2+ responses. We did not exhaustively test all members of the Rho family targeted by ToxB but can dismiss any factors reciprocally activated by inhibition of Rho, Rac, or Cdc42. Thus, although changes in Rho activity are sufficient to explain the depression of UDP Ca2+ responses, the elevation of C5a responses does not appear linked to ToxB effects on this family of G-proteins.

In contrast to the inhibitory effects of ToxB on PLCβ4-mediated Ca2+ signaling, ToxB enhanced PLCβ3-mediated Ca2+ signaling through enhanced production of InsP3. This selective effect of ToxB on PLCβ3 activation required cPLA2 activity and lipoxygenation of AA. In macrophages, C5a signals primarily through PLCβ3 [49], so ToxB enhances the Ca2+ response to C5a. Gαi-linked receptors, such as the C5aR, activate PLCβ2 and PLCβ3 through the release of Gβγ, and the selective use of PLCβ3 by the C5aR in macrophages is not yet understood nor is the selective enhancement of PLCβ3 signaling by ToxB. ToxB does not promote effects of Gβγ globally, as ToxB did not enhance PLCβ2 signaling, and it inhibited rather than enhanced the activation of PI3K, another effector of Gβγ signaling (data not shown). Our studies do not, however, exclude the possibility that ToxB selectively regulates the contribution of Gβγ to these signaling pathways. It remains unclear if the PLA2-dependent enhancement of PLCβ3 is dependent on the glucosyltransferase activity of ToxB toward unidentified members of the Rho family or if its effect on PLA2 is independent of Rho family involvement.

Several possible mechanisms for this enhancement of PLCβ3 activity were evaluated, but none was supported by our experimental results. Possible changes in subcellular distribution of PLCβ3 versus PLCβ4 were examined, but we found no discernible changes to explain the enhanced PLCβ3-dependent responses (data not shown). The sufficiency of AA exposure and requirement for 5-LOX activity focused our attention on a short list of likely LT mediators, several of which can couple to Gαq-linked GPCRs. Previous studies of C5aR synergy with Gαq-coupled GPCRs [49] showed a similar elevation of PLCβ3-mediated InsP3 production and reduced phosphorylation of AKT, and LT-stimulated GPCRs could provide synergistic elevation of PLCβ3-mediated responses to C5a. Thus, we evaluated the effect of pre-exposure and simultaneous exposure to the LTs B4, D4 (same receptor as C4), 5-HETE, and 5-oxo-HETE at varied doses and times on C5a responses but observed no significant enhancement of C5a responses. 5-LOX activity may be necessary but not sufficient for the ToxB effect, and other AA metabolites may contribute to provide for enhanced PLCβ3 responses.

The dichotomous effects of ToxB on GPRC-mediated Ca2+ signaling in macrophages provide new insights into the mechanism of Ca2+ signaling in these cells and may have consequences for the pathogenicity of C. difficile. The Rho-mediated effects of ToxB on UDP Ca2+ signaling may be seen as part of a broad attack by C. difficile on macrophage function. In macrophages, the Rho family influences cell adhesion, cell migration, phagocytosis, macropinocytosis, immunoreceptor activation, and the production of reactive oxygen species [9, 10, 54]. Inhibition of these processes in macrophages (and neutrophils) may provide clear advantages to C. difficile toward immune evasion. Similarly, reduction in Rho-dependent Ca2+ signals from some GPCRs may impede the host response. The enhancing effects of ToxB on C5a Ca2+ signaling, however, present a less-obvious advantage to the pathogen and could instead reflect host responses to C. difficile or a combination of these competing goals. Thus, the generation of AA metabolites may potentiate the inflammatory response to facilitate pathogen clearance even as it exacerbates tissue injury. cPLA2 activity and AA can confer resistance to apoptotic stimuli in macrophages [114,115,116], and PLCβ3-deficient macrophages are more susceptible to apoptosis [117], suggesting that PLCβ3 imparts antiapoptotic effects in these cells. Thus, generation of AA may improve macrophage survival and facilitate host defense. Through other mechanisms, however, AA can also contribute to apoptotic responses [118, 119], so the net effect is uncertain. It would be interesting to assess the resistance of PLCβ3-deficient mice to C. difficile infection to identify if these elevated responses benefit host or pathogen.

In sum, our studies of macrophage signaling reveal selective effects of ToxB and of Rho on different GPCR signaling pathways, demonstrating that ToxB, by inhibiting Rho, selectively inhibits Ca2+ responses mediated by PLCβ4 but not PLCβ3. The requirement for Rho in PLCβ4 activity is shown to lie at the level of Ca2+ release by InsP3, implying that PLCβ4 differs mechanistically from PLCβ3 in promoting this step. In contrast to its inhibition of PLCβ4, ToxB promotes the activation of PLCβ3 by the Gαi-coupled C5aR, through a pathway that is dependent on the metabolism of AA to LTs. These differential effects of ToxB on PLCβ3 versus PLCβ4 pathways elucidate new regulatory pathways for signal transduction in macrophages, and they may lead to new targets for blocking the clinical effects of ToxB.

AUTHORSHIP

Robert A. Rebres: project design, statistical analyses, authorship, supervision, cell culture, expression constructs and transfections, population and single-cell Ca2+ assays, Western blots, Rho assays, and cytoskeletal assessments; Christina Moon: cell culture, transfections, population Ca2+ assays, Western blots, InsP3 analyses, and phosphatidylinositol analyses; Dianne DeCamp: InsP3 analyses; Keng-Mean Lin: expression constructs and InsP3 analyses; Iain D. Fraser: project design, cell culture, and expression constructs; Stephen B. Milne: phosphatidylinositol analyses; Tamara I. A. Roach: authorship and supervision; H. Alex Brown: project design and supervision; and William E. Seaman: project design, authorship, and supervision.

ACKNOWLEDGMENTS

This work was supported by National Institutes of Health grant GM 62114. We thank Amanda Norton (San Francisco Veterans Affairs Medical Center) and Jamie Liu, Leah Santat, and Lucas Cheadle (Caltech) for excellent technical assistance.

Footnotes

Abbreviations: 5-HETE=5(S)-hydroxy-6,8,11,14-eicosatetraenoic acid, 5-LOX=5-lipoxygenase, AA=arachidonic acid, AA861=2-(12-hydroxydodeca-5,10-diynyl)-3,5,6-trimethyl-p-benzoquinone, AACOCF3=arachidonyltrifluoromethyl ketone, AFC=N-acetyl-S-farnesyl-L-cysteine, AfCS=Alliance for Cellular Signaling, AGGC=N-acetyl-S-geranylgeranyl-L-cysteine, AM=acetoxymethyl ester, BMDM=bone marrow-derived macrophage(s), C5a=complement 5a, [Ca2+]I=intracellular-free calcium, CFP=cyan fluorescent protein, COX=cyclooxygenase, cPLA2=cytosolic PLA2, DAG= diacylglycerol, DAPI=4′,6-diamidino-2-phenylindole, ER=endoplasmic reticulum, FLAP=5-LOX-activating protein, FTA=S-farnesylthioacetic acid, GDI=GDP-dissociation inhibitor, GEF=guanine nucleotide exchange factor, GM=growth medium, GPCR=G-protein-coupled receptor, InsP3=inositol-1,4,5-trisphosphate, LPA=lysophosphatidic acid, LT=leukotriene, MAFP=methyl arachidonyl fluorophosphonate, MK-886=3-[1-(p-chlorobenzyl)-5-(isopropyl)-3-t-butylthioindol-2-yl]-2,2-dimethylpropanoic acid, NDGA=nordihydroguaiaretic acid, P=phospho, PAF=platelet-activating factor, PIP2=phosphatidylinositol-4,5-bisphosphate, PLA2/C=phospholipase A2/C, PM=propionoxymethyl, PP2=4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine, PP3=4-amino-7-phenylpyrazol[3,4-d]pyrimidine, PTx=pertussis toxin, RNAi=RNA interference, S1P=sphingosine-1-phosphate, SOC=store-operated channel, TC=tissue culture, ToxA/B=toxin A/B from Clostridium difficile, wt=wild-type

References

  1. Durai R. Epidemiology, pathogenesis, and management of Clostridium difficile infection. Dig Dis Sci. 2007;52:2958–2962. doi: 10.1007/s10620-006-9626-y. [DOI] [PubMed] [Google Scholar]
  2. Poxton I R, McCoubrey J, Blair G. The pathogenicity of Clostridium difficile. Clin Microbiol Infect. 2001;7:421–427. doi: 10.1046/j.1198-743x.2001.00287.x. [DOI] [PubMed] [Google Scholar]
  3. Voth D E, Ballard J D. Clostridium difficile toxins: mechanism of action and role in disease. Clin Microbiol Rev. 2005;18:247–263. doi: 10.1128/CMR.18.2.247-263.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Lyerly D M, Saum K E, MacDonald D K, Wilkins T D. Effects of Clostridium difficile toxins given intragastrically to animals. Infect Immun. 1985;47:349–352. doi: 10.1128/iai.47.2.349-352.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Lyras D, O'Connor J R, Howarth P M, Sambol S P, Carter G P, Phumoonna T, Poon R, Adams V, Vedantam G, Johnson S, Gerding D N, Rood J I. Toxin B is essential for virulence of Clostridium difficile. Nature. 2009;458:1176–1179. doi: 10.1038/nature07822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Just I, Fritz G, Aktories K, Giry M, Popoff M R, Boquet P, Hegenbarth S, von Eichel-Streiber C. Clostridium difficile toxin B acts on the GTP-binding protein Rho. J Biol Chem. 1994;269:10706–10712. [PubMed] [Google Scholar]
  7. Just I, Selzer J, von Eichel-Streiber C, Aktories K. The low molecular mass GTP-binding protein Rho is affected by toxin A from Clostridium difficile. J Clin Invest. 1995;95:1026–1031. doi: 10.1172/JCI117747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Jank T, Giesemann T, Aktories K. Rho-glucosylating Clostridium difficile toxins A and B: new insights into structure and function. Glycobiology. 2007;17:15R–22R. doi: 10.1093/glycob/cwm004. [DOI] [PubMed] [Google Scholar]
  9. Wennerberg K, Der C J. Rho-family GTPases: it’s not only Rac and Rho (and I like it) J Cell Sci. 2004;117:1301–1312. doi: 10.1242/jcs.01118. [DOI] [PubMed] [Google Scholar]
  10. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420:629–635. doi: 10.1038/nature01148. [DOI] [PubMed] [Google Scholar]
  11. Flegel W A, Muller F, Daubener W, Fischer H G, Hadding U, Northoff H. Cytokine response by human monocytes to Clostridium difficile toxin A and toxin B. Infect Immun. 1991;59:3659–3666. doi: 10.1128/iai.59.10.3659-3666.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Rocha M F, Maia M E, Bezerra L R, Lyerly D M, Guerrant R L, Ribeiro R A, Lima A A. Clostridium difficile toxin A induces the release of neutrophil chemotactic factors from rat peritoneal macrophages: role of interleukin-1β, tumor necrosis factor α, and leukotrienes. Infect Immun. 1997;65:2740–2746. doi: 10.1128/iai.65.7.2740-2746.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Linevsky J K, Pothoulakis C, Keates S, Warny M, Keates A C, Lamont J T, Kelly C P. IL-8 release and neutrophil activation by Clostridium difficile toxin-exposed human monocytes. Am J Physiol. 1997;273:G1333–G1340. doi: 10.1152/ajpgi.1997.273.6.G1333. [DOI] [PubMed] [Google Scholar]
  14. Triadafilopoulos G, Pothoulakis C, Weiss R, Giampaolo C, Lamont J T. Comparative study of Clostridium difficile toxin A and cholera toxin in rabbit ileum. Gastroenterology. 1989;97:1186–1192. doi: 10.1016/0016-5085(89)91689-2. [DOI] [PubMed] [Google Scholar]
  15. Souza M H, Melo-Filho A A, Rocha M F, Lyerly D M, Cunha F Q, Lima A A, Ribeiro R A. The involvement of macrophage-derived tumor necrosis factor and lipoxygenase products on the neutrophil recruitment induced by Clostridium difficile toxin B. Immunology. 1997;91:281–288. doi: 10.1046/j.1365-2567.1997.00243.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Berridge M J, Bootman M D, Roderick H L. Calcium signaling: dynamics, homeostasis and remodeling. Nat Rev Mol Cell Biol. 2003;4:517–529. doi: 10.1038/nrm1155. [DOI] [PubMed] [Google Scholar]
  17. Berridge M J. Calcium microdomains: organization and function. Cell Calcium. 2006;40:405–412. doi: 10.1016/j.ceca.2006.09.002. [DOI] [PubMed] [Google Scholar]
  18. Delmas P, Crest M, Brown D A. Functional organization of PLC signaling microdomains in neurons. Trends Neurosci. 2004;27:41–47. doi: 10.1016/j.tins.2003.10.013. [DOI] [PubMed] [Google Scholar]
  19. Hackam D J, Rotstein O D, Schreiber A, Zhang W, Grinstein S. Rho is required for the initiation of calcium signaling and phagocytosis by Fcγ receptors in macrophages. J Exp Med. 1997;186:955–966. doi: 10.1084/jem.186.6.955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Arrieumerlou C, Randriamampita C, Bismuth G, Trautmann A. Rac is involved in early TCR signaling. J Immunol. 2000;165:3182–3189. doi: 10.4049/jimmunol.165.6.3182. [DOI] [PubMed] [Google Scholar]
  21. Greenberg S. Modular components of phagocytosis. J Leukoc Biol. 1999;66:712–717. doi: 10.1002/jlb.66.5.712. [DOI] [PubMed] [Google Scholar]
  22. Massol P, Montcourrier P, Guillemot J C, Chavrier P. Fc receptor-mediated phagocytosis requires CDC42 and Rac1. EMBO J. 1998;17:6219–6229. doi: 10.1093/emboj/17.21.6219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Shin D M, Zhao X S, Zeng W, Mozhayeva M, Muallem S. The mammalian Sec6/8 complex interacts with Ca(2+) signaling complexes and regulates their activity. J Cell Biol. 2000;150:1101–1112. doi: 10.1083/jcb.150.5.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Duncan R S, Hwang S Y, Koulen P. Effects of Vesl/Homer proteins on intracellular signaling. Exp Biol Med (Maywood) 2005;230:527–535. doi: 10.1177/153537020523000803. [DOI] [PubMed] [Google Scholar]
  25. Chong L D, Traynor-Kaplan A, Bokoch G M, Schwartz M A. The small GTP-binding protein Rho regulates a phosphatidylinositol 4-phosphate 5-kinase in mammalian cells. Cell. 1994;79:507–513. doi: 10.1016/0092-8674(94)90259-3. [DOI] [PubMed] [Google Scholar]
  26. Schmidt M, Bienek C, Rumenapp U, Zhang C, Lummen G, Jakobs K H, Just I, Aktories K, Moos M, von Eichel-Streiber C. A role for Rho in receptor- and G protein-stimulated phospholipase C. Reduction in phosphatidylinositol 4,5-bisphosphate by Clostridium difficile toxin B. Naunyn Schmiedebergs Arch Pharmacol. 1996;354:87–94. doi: 10.1007/BF00178707. [DOI] [PubMed] [Google Scholar]
  27. Schmidt M, Rumenapp U, Keller J, Lohmann B, Jakobs K H. Regulation of phospholipase C and D activities by small molecular weight G proteins and muscarinic receptors. Life Sci. 1997;60:1093–1100. doi: 10.1016/s0024-3205(97)00052-0. [DOI] [PubMed] [Google Scholar]
  28. Zhang C, Schmidt M, von Eichei-Streiber C, Jakobs K H. Inhibition by toxin B of inositol phosphate formation induced by G protein-coupled and tyrosine kinase receptors in N1E-115 neuroblastoma cells: involvement of Rho proteins. Mol Pharmacol. 1996;50:864–869. [PubMed] [Google Scholar]
  29. Weernink P A, Guo Y, Zhang C, Schmidt M, Von Eichel-Streiber C, Jakobs K H. Control of cellular phosphatidylinositol 4,5-bisphosphate levels by adhesion signals and rho GTPases in NIH 3T3 fibroblasts involvement of both phosphatidylinositol-4-phosphate 5-kinase and phospholipase C. Eur J Biochem. 2000;267:5237–5246. doi: 10.1046/j.1432-1327.2000.01599.x. [DOI] [PubMed] [Google Scholar]
  30. McNamee H P, Ingber D E, Schwartz M A. Adhesion to fibronectin stimulates inositol lipid synthesis and enhances PDGF-induced inositol lipid breakdown. J Cell Biol. 1993;121:673–678. doi: 10.1083/jcb.121.3.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Illenberger D, Schwald F, Pimmer D, Binder W, Maier G, Dietrich A, Gierschik P. Stimulation of phospholipase C-β2 by the Rho GTPases Cdc42Hs and Rac1. EMBO J. 1998;17:6241–6249. doi: 10.1093/emboj/17.21.6241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Snyder J T, Singer A U, Wing M R, Harden T K, Sondek J. The pleckstrin homology domain of phospholipase C-β2 as an effector site for Rac. J Biol Chem. 2003;278:21099–21104. doi: 10.1074/jbc.M301418200. [DOI] [PubMed] [Google Scholar]
  33. Rhee S G. Regulation of phosphoinositide-specific phospholipase C. Annu Rev Biochem. 2001;70:281–312. doi: 10.1146/annurev.biochem.70.1.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Wing M R, Snyder J T, Sondek J, Harden T K. Direct activation of phospholipase C-ε by Rho. J Biol Chem. 2003;278:41253–41258. doi: 10.1074/jbc.M306904200. [DOI] [PubMed] [Google Scholar]
  35. Bunney T D, Katan M. Phospholipase C ε: linking second messengers and small GTPases. Trends Cell Biol. 2006;16:640–648. doi: 10.1016/j.tcb.2006.10.007. [DOI] [PubMed] [Google Scholar]
  36. Hashimoto K, Watanabe M, Kurihara H, Offermanns S, Jiang H, Wu Y, Jun K, Shin H S, Inoue Y, Wu D, Simon M I, Kano M. Climbing fiber synapse elimination during postnatal cerebellar development requires signal transduction involving G α q and phospholipase C β 4. Prog Brain Res. 2000;124:31–48. doi: 10.1016/S0079-6123(00)24006-5. [DOI] [PubMed] [Google Scholar]
  37. Offermanns S, Hashimoto K, Watanabe M, Sun W, Kurihara H, Thompson R F, Inoue Y, Kano M, Simon M I. Impaired motor coordination and persistent multiple climbing fiber innervation of cerebellar Purkinje cells in mice lacking Gαq. Proc Natl Acad Sci USA. 1997;94:14089–14094. doi: 10.1073/pnas.94.25.14089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Offermanns S, Zhao L P, Gohla A, Sarosi I, Simon M I, Wilkie T M. Embryonic cardiomyocyte hypoplasia and craniofacial defects in G α q/G α 11-mutant mice. EMBO J. 1998;17:4304–4312. doi: 10.1093/emboj/17.15.4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Xie W, Samoriski G M, McLaughlin J P, Romoser V A, Smrcka A, Hinkle P M, Bidlack J M, Gross R A, Jiang H, Wu D. Genetic alteration of phospholipase C β3 expression modulates behavioral and cellular responses to μ opioids. Proc Natl Acad Sci USA. 1999;96:10385–10390. doi: 10.1073/pnas.96.18.10385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Jiang H, Lyubarsky A, Dodd R, Vardi N, Pugh E, Baylor D, Simon M I, Wu D. Phospholipase C β 4 is involved in modulating the visual response in mice. Proc Natl Acad Sci USA. 1996;93:14598–14601. doi: 10.1073/pnas.93.25.14598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kano M, Hashimoto K, Watanabe M, Kurihara H, Offermanns S, Jiang H, Wu Y, Jun K, Shin H S, Inoue Y, Simon M I, Wu D. Phospholipase cβ4 is specifically involved in climbing fiber synapse elimination in the developing cerebellum. Proc Natl Acad Sci USA. 1998;95:15724–15729. doi: 10.1073/pnas.95.26.15724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Jiang H, Kuang Y, Wu Y, Xie W, Simon M I, Wu D. Roles of phospholipase C β2 in chemoattractant-elicited responses. Proc Natl Acad Sci USA. 1997;94:7971–7975. doi: 10.1073/pnas.94.15.7971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Takeshita S, Kaji K, Kudo A. Identification and characterization of the new osteoclast progenitor with macrophage phenotypes being able to differentiate into mature osteoclasts. J Bone Miner Res. 2000;15:1477–1488. doi: 10.1359/jbmr.2000.15.8.1477. [DOI] [PubMed] [Google Scholar]
  44. Rouzer C A, Ivanova P T, Byrne M O, Milne S B, Marnett L J, Brown H A. Lipid profiling reveals arachidonate deficiency in RAW264.7 cells: structural and functional implications. Biochemistry. 2006;45:14795–14808. doi: 10.1021/bi061723j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Heo W D, Inoue T, Park W S, Kim M L, Park B O, Wandless T J, Meyer T. PI(3,4,5)P3 and PI(4,5)P2 lipids target proteins with polybasic clusters to the plasma membrane. Science. 2006;314:1458–1461. doi: 10.1126/science.1134389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Grynkiewicz G, Poenie M, Tsien R Y. A new generation of Ca2+ indicators with greatly improved fluorescence properties. J Biol Chem. 1985;260:3440–3450. [PubMed] [Google Scholar]
  47. Pfeiffer Z A, Aga M, Prabhu U, Watters J J, Hall D J, Bertics P J. The nucleotide receptor P2X7 mediates actin reorganization and membrane blebbing in RAW 264.7 macrophages via p38 MAP kinase and Rho. J Leukoc Biol. 2004;75:1173–1182. doi: 10.1189/jlb.1203648. [DOI] [PubMed] [Google Scholar]
  48. Conner S D, Schmid S L. Regulated portals of entry into the cell. Nature. 2003;422:37–44. doi: 10.1038/nature01451. [DOI] [PubMed] [Google Scholar]
  49. Roach T I, Rebres R A, Fraser I D, Decamp D L, Mean-Lin K, Sternweis P C, Simon M I, Seaman W E. Signaling and cross-talk by C5a and UDP in macrophages selectively use PLCβ3 to regulate intracellular free calcium. J Biol Chem. 2008;283:17351–17361. doi: 10.1074/jbc.M800907200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Milne S B, Ivanova P T, DeCamp D, Hsueh R C, Brown H A. A targeted mass spectrometric analysis of phosphatidylinositol phosphate species. J Lipid Res. 2005;46:1796–1802. doi: 10.1194/jlr.D500010-JLR200. [DOI] [PubMed] [Google Scholar]
  51. Ivanova P T, Milne S B, Byrne M O, Xiang Y, Brown H A. Glycerophospholipid identification and quantitation by electrospray ionization mass spectrometry. Methods Enzymol. 2007;432:21–57. doi: 10.1016/S0076-6879(07)32002-8. [DOI] [PubMed] [Google Scholar]
  52. Just I, Selzer J, Wilm M, von Eichel-Streiber C, Mann M, Aktories K. Glucosylation of Rho proteins by Clostridium difficile toxin B. Nature. 1995;375:500–503. doi: 10.1038/375500a0. [DOI] [PubMed] [Google Scholar]
  53. Coward P, Wada H G, Falk M S, Chan S D, Meng F, Akil H, Conklin B R. Controlling signaling with a specifically designed Gi-coupled receptor. Proc Natl Acad Sci USA. 1998;95:352–357. doi: 10.1073/pnas.95.1.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hall A. Rho GTPases and the control of cell behavior. Biochem Soc Trans. 2005;33:891–895. doi: 10.1042/BST20050891. [DOI] [PubMed] [Google Scholar]
  55. Rossman K L, Der C J, Sondek J. GEF means go: turning on Rho GTPases with guanine nucleotide-exchange factors. Nat Rev Mol Cell Biol. 2005;6:167–180. doi: 10.1038/nrm1587. [DOI] [PubMed] [Google Scholar]
  56. Wing M R, Bourdon D M, Harden T K. PLC-ε: a shared effector protein in Ras-, Rho-, and G α β γ-mediated signaling. Mol Interv. 2003;3:273–280. doi: 10.1124/mi.3.5.273. [DOI] [PubMed] [Google Scholar]
  57. Shiraishi Y, Mizutani A, Bito H, Fujisawa K, Narumiya S, Mikoshiba K, Furuichi T. Cupidin, an isoform of Homer/Vesl, interacts with the actin cytoskeleton and activated rho family small GTPases and is expressed in developing mouse cerebellar granule cells. J Neurosci. 1999;19:8389–8400. doi: 10.1523/JNEUROSCI.19-19-08389.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hwang J I, Kim H S, Lee J R, Kim E, Ryu S H, Suh P G. The interaction of phospholipase C-β3 with Shank2 regulates mGluR-mediated calcium signal. J Biol Chem. 2005;280:12467–12473. doi: 10.1074/jbc.M410740200. [DOI] [PubMed] [Google Scholar]
  59. Ehrengruber M U, Kato A, Inokuchi K, Hennou S. Homer/Vesl proteins and their roles in CNS neurons. Mol Neurobiol. 2004;29:213–227. doi: 10.1385/MN:29:3:213. [DOI] [PubMed] [Google Scholar]
  60. Sekine A, Fujiwara M, Narumiya S. Asparagine residue in the rho gene product is the modification site for botulinum ADP-ribosyltransferase. J Biol Chem. 1989;264:8602–8605. [PubMed] [Google Scholar]
  61. Genth H, Gerhard R, Maeda A, Amano M, Kaibuchi K, Aktories K, Just I. Entrapment of Rho ADP-ribosylated by Clostridium botulinum C3 exoenzyme in the Rho-guanine nucleotide dissociation inhibitor-1 complex. J Biol Chem. 2003;278:28523–28527. doi: 10.1074/jbc.M301915200. [DOI] [PubMed] [Google Scholar]
  62. Genth H, Schmidt M, Gerhard R, Aktories K, Just I. Activation of phospholipase D1 by ADP-ribosylated RhoA. Biochem Biophys Res Commun. 2003;302:127–132. doi: 10.1016/s0006-291x(03)00112-8. [DOI] [PubMed] [Google Scholar]
  63. Wilde C, Genth H, Aktories K, Just I. Recognition of RhoA by Clostridium botulinum C3 exoenzyme. J Biol Chem. 2000;275:16478–16483. doi: 10.1074/jbc.M910362199. [DOI] [PubMed] [Google Scholar]
  64. Just I, Mohr C, Schallehn G, Menard L, Didsbury J R, Vandekerckhove J, van Damme J, Aktories K. Purification and characterization of an ADP-ribosyltransferase produced by Clostridium limosum. J Biol Chem. 1992;267:10274–10280. [PubMed] [Google Scholar]
  65. Aktories K, Braun U, Rosener S, Just I, Hall A. The rho gene product expressed in E. coli is a substrate of botulinum ADP-ribosyltransferase C3. Biochem Biophys Res Commun. 1989;158:209–213. doi: 10.1016/s0006-291x(89)80199-8. [DOI] [PubMed] [Google Scholar]
  66. Wheeler A P, Ridley A J. Why three Rho proteins? RhoA, RhoB, RhoC, and cell motility. Exp Cell Res. 2004;301:43–49. doi: 10.1016/j.yexcr.2004.08.012. [DOI] [PubMed] [Google Scholar]
  67. Bishop A L, Hall A. Rho GTPases and their effector proteins. Biochem J. 2000;348:241–255. [PMC free article] [PubMed] [Google Scholar]
  68. Gerhard R, Tatge H, Genth H, Thum T, Borlak J, Fritz G, Just I. Clostridium difficile toxin A induces expression of the stress-induced early gene product RhoB. J Biol Chem. 2005;280:1499–1505. doi: 10.1074/jbc.M406014200. [DOI] [PubMed] [Google Scholar]
  69. Huelsenbeck J, Dreger S C, Gerhard R, Fritz G, Just I, Genth H. Upregulation of the immediate early gene product RhoB by exoenzyme C3 from Clostridium limosum and toxin B from Clostridium difficile. Biochemistry. 2007;46:4923–4931. doi: 10.1021/bi602465z. [DOI] [PubMed] [Google Scholar]
  70. Ridley A J. Rho proteins: linking signaling with membrane trafficking. Traffic. 2001;2:303–310. doi: 10.1034/j.1600-0854.2001.002005303.x. [DOI] [PubMed] [Google Scholar]
  71. Fernandez-Borja M, Janssen L, Verwoerd D, Hordijk P, Neefjes J. RhoB regulates endosome transport by promoting actin assembly on endosomal membranes through Dia1. J Cell Sci. 2005;118:2661–2670. doi: 10.1242/jcs.02384. [DOI] [PubMed] [Google Scholar]
  72. Wheeler A P, Ridley A J. RhoB affects macrophage adhesion, integrin expression and migration. Exp Cell Res. 2007;313:3505–3516. doi: 10.1016/j.yexcr.2007.07.014. [DOI] [PubMed] [Google Scholar]
  73. Pan J, Yeung S C. Recent advances in understanding the antineoplastic mechanisms of farnesyltransferase inhibitors. Cancer Res. 2005;65:9109–9112. doi: 10.1158/0008-5472.CAN-05-2635. [DOI] [PubMed] [Google Scholar]
  74. Konstantinopoulos P A, Karamouzis M V, Papavassiliou A G. Post-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targets. Nat Rev Drug Discov. 2007;6:541–555. doi: 10.1038/nrd2221. [DOI] [PubMed] [Google Scholar]
  75. Kramer K, Harrington E O, Lu Q, Bellas R, Newton J, Sheahan K L, Rounds S. Isoprenylcysteine carboxyl methyltransferase activity modulates endothelial cell apoptosis. Mol Biol Cell. 2003;14:848–857. doi: 10.1091/mbc.E02-07-0390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Rosado J A, Sage S O. Regulation of plasma membrane Ca2+-ATPase by small GTPases and phosphoinositides in human platelets. J Biol Chem. 2000;275:19529–19535. doi: 10.1074/jbc.M001319200. [DOI] [PubMed] [Google Scholar]
  77. Rosado J A, Sage S O. Farnesylcysteine analogues inhibit store-regulated Ca2+ entry in human platelets: evidence for involvement of small GTP-binding proteins and actin cytoskeleton. Biochem J. 2000;347:183–192. [PMC free article] [PubMed] [Google Scholar]
  78. Della Rocca G J, Maudsley S, Daaka Y, Lefkowitz R J, Luttrell L M. Pleiotropic coupling of G protein-coupled receptors to the mitogen-activated protein kinase cascade. Role of focal adhesions and receptor tyrosine kinases. J Biol Chem. 1999;274:13978–13984. doi: 10.1074/jbc.274.20.13978. [DOI] [PubMed] [Google Scholar]
  79. Daub H, Wallasch C, Lankenau A, Herrlich A, Ullrich A. Signal characteristics of G protein-transactivated EGF receptor. EMBO J. 1997;16:7032–7044. doi: 10.1093/emboj/16.23.7032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Haendeler J, Yin G, Hojo Y, Saito Y, Melaragno M, Yan C, Sharma V K, Heller M, Aebersold R, Berk B C. GIT1 mediates Src-dependent activation of phospholipase Cγ by angiotensin II and epidermal growth factor. J Biol Chem. 2003;278:49936–49944. doi: 10.1074/jbc.M307317200. [DOI] [PubMed] [Google Scholar]
  81. McGarrigle D, Huang X Y. GPCRs signaling directly through Src-family kinases. Sci STKE. 2007;2007:pe35. doi: 10.1126/stke.3922007pe35. [DOI] [PubMed] [Google Scholar]
  82. Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213:589–602. doi: 10.1002/jcp.21246. [DOI] [PubMed] [Google Scholar]
  83. Andreev J, Galisteo M L, Kranenburg O, Logan S K, Chiu E S, Okigaki M, Cary L A, Moolenaar W H, Schlessinger J. Src and Pyk2 mediate G-protein-coupled receptor activation of epidermal growth factor receptor (EGFR) but are not required for coupling to the mitogen-activated protein (MAP) kinase signaling cascade. J Biol Chem. 2001;276:20130–20135. doi: 10.1074/jbc.M102307200. [DOI] [PubMed] [Google Scholar]
  84. Hoefen R J, Berk B C. The multifunctional GIT family of proteins. J Cell Sci. 2006;119:1469–1475. doi: 10.1242/jcs.02925. [DOI] [PubMed] [Google Scholar]
  85. Kuehn H S, Beaven M A, Ma H T, Kim M S, Metcalfe D D, Gilfillan A M. Synergistic activation of phospholipases Cγ and Cβ: a novel mechanism for PI3K-independent enhancement of FcεRI-induced mast cell mediator release. Cell Signal. 2008;20:625–636. doi: 10.1016/j.cellsig.2007.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Wang Y J, Li W H, Wang J, Xu K, Dong P, Luo X, Yin H L. Critical role of PIP5KI{γ}87 in InsP3-mediated Ca(2+) signaling. J Cell Biol. 2004;167:1005–1010. doi: 10.1083/jcb.200408008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Doughman R L, Firestone A J, Anderson R A. Phosphatidylinositol phosphate kinases put PI4,5P(2) in its place. J Membr Biol. 2003;194:77–89. doi: 10.1007/s00232-003-2027-7. [DOI] [PubMed] [Google Scholar]
  88. Niggli V. Regulation of protein activities by phosphoinositide phosphates. Annu Rev Cell Dev Biol. 2005;21:57–79. doi: 10.1146/annurev.cellbio.21.021704.102317. [DOI] [PubMed] [Google Scholar]
  89. Koreh K, Monaco M E. The relationship of hormone-sensitive and hormone-insensitive phosphatidylinositol to phosphatidylinositol 4,5-bisphosphate in the WRK-1 cell. J Biol Chem. 1986;261:88–91. [PubMed] [Google Scholar]
  90. Rosado J A, Sage S O. The actin cytoskeleton in store-mediated calcium entry. J Physiol. 2000;526:221–229. doi: 10.1111/j.1469-7793.2000.t01-2-00221.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Bozem M, Kuhlmann S, Blum R, Feick P, Schulz I. Hormone-stimulated calcium release is inhibited by cytoskeleton-disrupting toxins in AR4–2J cells. Cell Calcium. 2000;28:73–82. doi: 10.1054/ceca.2000.0133. [DOI] [PubMed] [Google Scholar]
  92. Delmas P, Wanaverbecq N, Abogadie F C, Mistry M, Brown D A. Signaling microdomains define the specificity of receptor-mediated InsP(3) pathways in neurons. Neuron. 2002;34:209–220. doi: 10.1016/s0896-6273(02)00641-4. [DOI] [PubMed] [Google Scholar]
  93. Ribeiro C M, Reece J, Putney J W., Jr Role of the cytoskeleton in calcium signaling in NIH 3T3 cells. An intact cytoskeleton is required for agonist-induced [Ca2+]i signaling, but not for capacitative calcium entry. J Biol Chem. 1997;272:26555–26561. doi: 10.1074/jbc.272.42.26555. [DOI] [PubMed] [Google Scholar]
  94. Gregory R B, Wilcox R A, Berven L A, van Straten N C, van der Marel G A, van Boom J H, Barritt G J. Evidence for the involvement of a small subregion of the endoplasmic reticulum in the inositol trisphosphate receptor-induced activation of Ca2+ inflow in rat hepatocytes. Biochem J. 1999;341:401–408. [PMC free article] [PubMed] [Google Scholar]
  95. Bloch W, Fan Y, Han J, Xue S, Schoneberg T, Ji G, Lu Z J, Walther M, Fassler R, Hescheler J, Addicks K, Fleischmann B K. Disruption of cytoskeletal integrity impairs Gi-mediated signaling due to displacement of Gi proteins. J Cell Biol. 2001;154:753–761. doi: 10.1083/jcb.200103011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mahon M J, Segre G V. Stimulation by parathyroid hormone of a NHERF-1-assembled complex consisting of the parathyroid hormone I receptor, phospholipase Cβ, and actin increases intracellular calcium in opossum kidney cells. J Biol Chem. 2004;279:23550–23558. doi: 10.1074/jbc.M313229200. [DOI] [PubMed] [Google Scholar]
  97. Jiang L I, Collins J, Davis R, Lin K M, DeCamp D, Roach T, Hsueh R, Rebres R A, Ross E M, Taussig R, Fraser I, Sternweis P C. Use of a cAMP BRET sensor to characterize a novel regulation of cAMP by the sphingosine 1-phosphate/G13 pathway. J Biol Chem. 2007;282:10576–10584. doi: 10.1074/jbc.M609695200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Chikumi H, Vazquez-Prado J, Servitja J M, Miyazaki H, Gutkind J S. Potent activation of RhoA by Gα q and Gq-coupled receptors. J Biol Chem. 2002;277:27130–27134. doi: 10.1074/jbc.M204715200. [DOI] [PubMed] [Google Scholar]
  99. Shoshan M C, Florin I, Thelestam M. Activation of cellular phospholipase A2 by Clostridium difficile toxin B. J Cell Biochem. 1993;52:116–124. doi: 10.1002/jcb.240520115. [DOI] [PubMed] [Google Scholar]
  100. Shoshan M C, Fiorentini C, Thelestam M. Signal transduction pathways and cellular intoxication with Clostridium difficile toxins. J Cell Biochem. 1993;52:107–115. doi: 10.1002/jcb.240520114. [DOI] [PubMed] [Google Scholar]
  101. Stratton M D, Chandel B, Deshpande Y, Kaminski D L, Li A P, Vernava A M, Longo W E. The effect of Clostridium difficile toxin on colonocyte prostanoid activity. Prostaglandins. 1994;48:367–375. doi: 10.1016/0090-6980(94)90003-5. [DOI] [PubMed] [Google Scholar]
  102. Rocha M F, Soares A M, Flores C A, Steiner T S, Lyerly D M, Guerrant R L, Ribeiro R A, Lima A A. Intestinal secretory factor released by macrophages stimulated with Clostridium difficile toxin A: role of interleukin 1β. Infect Immun. 1998;66:4910–4916. doi: 10.1128/iai.66.10.4910-4916.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Buczynski M W, Stephens D L, Bowers-Gentry R C, Grkovich A, Deems R A, Dennis E A. TLR-4 and sustained calcium agonists synergistically produce eicosanoids independent of protein synthesis in RAW264.7 cells. J Biol Chem. 2007;282:22834–22847. doi: 10.1074/jbc.M701831200. [DOI] [PubMed] [Google Scholar]
  104. Hong J, Bose M, Ju J, Ryu J H, Chen X, Sang S, Lee M J, Yang C S. Modulation of arachidonic acid metabolism by curcumin and related β-diketone derivatives: effects on cytosolic phospholipase A(2), cyclooxygenases and 5-lipoxygenase. Carcinogenesis. 2004;25:1671–1679. doi: 10.1093/carcin/bgh165. [DOI] [PubMed] [Google Scholar]
  105. Kita Y, Takahashi T, Uozumi N, Shimizu T. A multiplex quantitation method for eicosanoids and platelet-activating factor using column-switching reversed-phase liquid chromatography-tandem mass spectrometry. Anal Biochem. 2005;342:134–143. doi: 10.1016/j.ab.2005.03.048. [DOI] [PubMed] [Google Scholar]
  106. Lin H I, Chu S J, Wang D, Feng N H. Pharmacological modulation of TNF production in macrophages. J Microbiol Immunol Infect. 2004;37:8–15. [PubMed] [Google Scholar]
  107. Yang S A, Carpenter C L, Abrams C S. Rho and Rho-kinase mediate thrombin-induced phosphatidylinositol 4-phosphate 5-kinase trafficking in platelets. J Biol Chem. 2004;279:42331–42336. doi: 10.1074/jbc.M404335200. [DOI] [PubMed] [Google Scholar]
  108. Weernink P A, Meletiadis K, Hommeltenberg S, Hinz M, Ishihara H, Schmidt M, Jakobs K H. Activation of type I phosphatidylinositol 4-phosphate 5-kinase isoforms by the Rho GTPases, RhoA, Rac1, and Cdc42. J Biol Chem. 2004;279:7840–7849. doi: 10.1074/jbc.M312737200. [DOI] [PubMed] [Google Scholar]
  109. Kjoller L, Hall A. Signaling to Rho GTPases. Exp Cell Res. 1999;253:166–179. doi: 10.1006/excr.1999.4674. [DOI] [PubMed] [Google Scholar]
  110. Sah V P, Seasholtz T M, Sagi S A, Brown J H. The role of Rho in G protein-coupled receptor signal transduction. Annu Rev Pharmacol Toxicol. 2000;40:459–489. doi: 10.1146/annurev.pharmtox.40.1.459. [DOI] [PubMed] [Google Scholar]
  111. Delmas P, Brown D A. Junctional signaling microdomains: bridging the gap between the neuronal cell surface and Ca2+ stores. Neuron. 2002;36:787–790. doi: 10.1016/s0896-6273(02)01097-8. [DOI] [PubMed] [Google Scholar]
  112. Sander E E, ten Klooster J P, van Delft S, van der Kammen R A, Collard J G. Rac downregulates Rho activity: reciprocal balance between both GTPases determines cellular morphology and migratory behavior. J Cell Biol. 1999;147:1009–1022. doi: 10.1083/jcb.147.5.1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Zondag G C, Evers E E, ten Klooster J P, Janssen L, van der Kammen R A, Collard J G. Oncogenic Ras downregulates Rac activity, which leads to increased Rho activity and epithelial-mesenchymal transition. J Cell Biol. 2000;149:775–782. doi: 10.1083/jcb.149.4.775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Tommasini I, Sestili P, Guidarelli A, Cantoni O. Peroxynitrite stimulates the activity of cytosolic phospholipase A2 in U937 cells: the extent of arachidonic acid formation regulates the balance between cell survival or death. Cell Death Differ. 2002;9:1368–1376. doi: 10.1038/sj.cdd.4401123. [DOI] [PubMed] [Google Scholar]
  115. Tommasini I, Guidarelli A, Palomba L, Cerioni L, Cantoni O. 5-Hydroxyeicosatetraenoic acid is a key intermediate of the arachidonate-dependent protective signaling in monocytes/macrophages exposed to peroxynitrite. J Leukoc Biol. 2006;80:929–938. doi: 10.1189/jlb.0406240. [DOI] [PubMed] [Google Scholar]
  116. Tommasini I, Guidarelli A, Cerioni L, Cantoni O. The arachidonate-dependent cytoprotective signaling evoked by peroxynitrite is a general response of the monocyte/macrophage lineage. Biochem Biophys Res Commun. 2004;316:1191–1195. doi: 10.1016/j.bbrc.2004.02.172. [DOI] [PubMed] [Google Scholar]
  117. Wang Z, Liu B, Wang P, Dong X, Fernandez-Hernando C, Li Z, Hla T, Claffey K, Smith J D, Wu D. Phospholipase C β3 deficiency leads to macrophage hypersensitivity to apoptotic induction and reduction of atherosclerosis in mice. J Clin Invest. 2008;118:195–204. doi: 10.1172/JCI33139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Taketo M M, Sonoshita M. Phospolipase A2 and apoptosis. Biochim Biophys Acta. 2002;1585:72–76. doi: 10.1016/s1388-1981(02)00326-8. [DOI] [PubMed] [Google Scholar]
  119. Balsinde J, Perez R, Balboa M A. Calcium-independent phospholipase A2 and apoptosis. Biochim Biophys Acta. 2006;1761:1344–1350. doi: 10.1016/j.bbalip.2006.07.013. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Leukocyte Biology are provided here courtesy of The Society for Leukocyte Biology

RESOURCES