Inhibition of signal termination-related kinases by membrane-permeant bitter and sweet tastants: potential role in taste signal termination

Meirav Zubare-Samuelov, Merav E. Shaul, Irena Peri, Alexander Aliluiko, Oren Tirosh, and Michael Naim

Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel

Submitted 10 November 2004 ; accepted in final form 21 March 2005


    ABSTRACT
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
Sweet and bitter taste sensations are believed to be initiated by the tastant-stimulated T1R and T2R G protein-coupled receptor (GPCR) subfamilies, respectively, which occur in taste cells. Although such tastants, with their significantly diverse chemical structures (e.g., sugar and nonsugar sweeteners), may share the same or similar T1Rs, some nonsugar sweeteners and many bitter tastants are amphipathic and produce a significant delay in taste termination (lingering aftertaste). We report that such tastants may permeate rat taste bud cells rapidly in vivo and inhibit known signal termination-related kinases in vitro, such as GPCR kinase (GRK)2, GRK5, and PKA. GRK5 and perhaps GRK2 and GRK6 are present in taste cells. A new hypothesis is proposed in which membrane-permeant tastants not only interact with taste GPCRs but also interact intracellularly with the receptors' downstream shutoff components to inhibit signal termination.

amphipathic tastants; tastant permeation; desensitization; lingering aftertaste


TASTE STIMULATION IS CHARACTERIZED by onset in less than a second and termination within a few seconds. However, some nonsugar sweeteners and many bitter tastants (most of them amphipathic, i.e., containing hydrophobic and hydrophilic domains) exhibit a significant delay in the extinction of taste sensation, which may last minutes, and is termed "lingering aftertaste" (6, 27). Although this particular delay in taste termination significantly affects the taste perception of many dietary constituents, its molecular basis is unknown.

The occurrence of G protein-coupled receptors (GPCRs) for both sweet and bitter tastes, as well as for the umami taste, is now incontestable (1, 3, 79, 22, 23, 26). In contrast, the nature of the cellular signals, essential components of taste transduction, is under considerable debate. A heterotrimeric G protein consisting of {alpha}-gustducin, G{beta}3, and G{gamma}13 is involved in the cellular response to bitter stimuli, of which the G{beta}3{gamma}13 moiety activates PLC{beta}2 (21); moreover, changes in second messengers such as cAMP, cGMP and D-myo-inositol 1,4,5-trisphosphate (IP3) in taste cells (5, 35, 40) in response to sweet and bitter taste stimulation have been proposed. Recent studies have indicated that mammalian transient receptor potential (TRPM)5, a PLC-dependent cationic channel that is believed to regulate Ca2+ entry into the cell, and PLC{beta}2 are present in taste cells (30). Interestingly, TRPM5- or PLC{beta}2-knockout mice almost completely lose their sensitivity to sugar, nonsugar sweetener, bitter, and umami taste stimulation, as evidenced by behavioral and electrophysiological experiments (46).

A few hypotheses may be proposed to explain the delay in taste signal termination induced by amphipathic tastants. Importantly, electrophysiological recordings of taste nerves at the periphery (16) have indicated a slow rise (onset) and a longer delay (lingering) in the sweet sensation response to stimulation by nonsugar sweeteners compared with that by sucrose. Biochemical time course measurements have shown that the transient onset and termination of IP3 release in taste tissue after stimulation by some bitter tastants known to possess lingering aftertaste is delayed >500 ms (38). These results suggest that the delay in taste signal termination induced by amphipathic tastants is at the taste cell level, at the periphery. Although sugar and nonsugar sweeteners appear to stimulate similar or identical sweet GPCRs, the lingering taste sensations are uniquely produced by nonsugar sweeteners (as well as bitter tastants). Because such tastants are direct activators of G proteins in vitro (25) and, unlike sucrose, rapidly permeate isolated taste bud cells in situ (31), they may interact with downstream transduction components that affect GPCR signal termination, in addition to their extracellular interaction with GPCRs.

GPCR signaling desensitizes rapidly as a consequence of receptor phosphorylation. GPCR phosphorylation is mediated by two families of protein kinases: the second messenger-dependent protein kinases (e.g., PKA) and the GPCR kinases (GRKs). In contrast to the former, GRKs specifically phosphorylate the agonist-activated form of GPCRs and promote the binding of arrestin proteins that further uncouple the receptors from G proteins (32). Thus, when ligands are involved, the phosphorylation of GPCRs by GRKs appears essential: deletion of GRK phosphorylation sites in the receptor results in stronger signaling responses to agonist stimulation in transfection studies (24). Inhibition of this phosphorylation can delay GPCR signal termination, as shown in vision (37) and other systems (39).

We previously found (31) that amphipathic tastants can translocate through multilamellar liposomes and rapidly permeate isolated circumvallate (CV) taste bud cells; intracellular concentrations of such tastants after 30 s of stimulation could reach millimolar levels. In line with previous conclusions (12), we hypothesize that such tastants, after permeation into the cells, have access to downstream transduction components that may be related to the delay in taste termination produced by such tastants. The present study was designed to further investigate whether amphipathic tastants can permeate taste cells under physiological conditions and interfere with GPCR signal termination, e.g., via inhibition of signal termination-related kinases.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
Animals and Tastants

Wistar female rats weighing 150–175 g were purchased from Harlan Laboratories (Jerusalem, Israel) and were kept in individual cages for a few days and supplied with rat laboratory chow and water ad libitum. The protocol for animal care and other procedures was approved by The Hebrew University of Jerusalem Animal Care Committee (NIH approval number: OPRR-A01-5011). The sweeteners sodium saccharin, neohesperidin dihydrochalcone (NHD), sodium cyclamate, D-tryptophan, and acesulfame K (all from Sigma, St. Louis, MO) and the bitter stimuli cyclo(Leu-Trp) (Bachem, Bubendorf, Switzerland), caffeine, quinine-HCl, L-tryptophan, limonin, and naringin (all from Sigma) were used.

Other Chemicals

Collagenase types D and A were purchased from Roche Diagnostics (Mannheim, Germany), and soybean trypsin inhibitor (type 1) and propidium iodide were from Sigma. Agarose was from Life Technologies (Paisley, UK), and ethidium bromide from Amresco (Solon, OH). Primary polyclonal antibodies for GRK2 (sc-562), GRK3 (sc-563), GRK5 (sc-565), and GRK6 (sc-566) and their respective blocking peptides were purchased from Santa Cruz International (Santa Cruz, CA). Fluorescein (FITC)-conjugated and peroxidase-conjugated AffiniPure goat anti-rabbit IgG (H+L) secondary antibody (111-095-144 and 111-035-003, respectively) were from Jackson ImmunoResearch Laboratories (West Grove, PA). DMSO was from Fluka (Buchs, Germany), HEPES, EGTA, tricine, and paraformaldehyde were from Sigma, KH2PO4 from Merck (Darmstadt, Germany), KCl and NaCl from Frutarom (Haifa, Israel), isopentane from J. T. Baker (Deventer, The Netherlands), and 20% (wt/vol) pentobarbital from J. M. Loveridge (Southampton, UK). [{gamma}-32P]ATP (~6,000 Ci/mmol) was provided by PerkinElmer Life Sciences (Boston, MA). The outer segment membranes of rod cells containing rhodopsin (kindly provided by R. J. Lefkowitz, Duke University, Durham, NC) were prepared and purified from dark-adapted bovine retinas and stripped with urea (17, 28). Bovine milk {beta}-casein and PKA (catalytic subunit from bovine heart), PKA inhibitor (fragment 6–22), and heparin were purchased from Sigma. GRK2 and GRK5, kindly provided by R. J. Lefkowitz, were purified from Sf9 insect cells (33). The three kinases were kept in 50% (vol/vol) glycerol in 2 mM EDTA, 20 mM Tris·HCl pH 7.4 buffer and stored at –12°C until use.

Permeation of Amphipathic Tastants into Taste Cells

Imaging of in situ permeation of tastants into CV taste papilla cells with confocal microscopy. During the experiments, animals were anesthetized with ether and then killed by decapitation. The CV papilla was identified under a dissection microscope (x10–12 magnification) illuminated with a Schott KL 1500 lamp. Intact CV papillae (3 x 3 x 2 mm) were removed with Vannas scissors and fine forceps. A slide containing a single CV papilla in 7.5 µl of modified Tyrode solution (in mM: 140 NaCl, 10 KCl, 1 MgCl2, 1 CaCl2, 10 glucose, 10 pyruvic acid, and 10 HEPES, adjusted to pH 7.4 with Tris base) was viewed under the Plan Nofluar x10/1.00 numerical aperture (NA) lens of a confocal laser-scanning microscope (Zeiss LSM 410 attached to a Zeiss Axiovert 135M inverted microscope). Tastant autofluorescence was used to monitor their permeation (31). At time 0, tastant solution (in Tyrode) was added, and the fluorescence intensity was set to background level. Images of the dorsal view of the CV papilla were taken in the focal plane in which the inner trench containing the taste bud area around the center of the papilla was visible (see Fig. 1, arrows). The increased fluorescence after the application of sodium saccharin (10 mM), D-tryptophan (10 mM), naringin (1 mM), cyclo(Leu-Trp) (1 mM), quinine (1 mM), and caffeine (10 mM) was monitored every minute for 6 min. To verify that the increased fluorescence with time was indeed due to tastant permeation and not to adsorption on the cell's extracellular surface, we applied, at the end of the 6-min period, Tyrode solution containing 500 mM KI, which is an efficient fluorescence quencher but an essentially membrane-impermeant salt (19, 31), for an additional 2 min. Propidium iodide (50 µg/ml), which is a fluorescent membrane-impermeant compound that is commonly used to identify dead cells and to stain nucleic acids (11), served as a positive control. KI's ability to quench the tastants and propidium iodide fluorescence were predetermined in Tyrode solution with a Varian Carry Eclipse fluorometer. Respective excitation and emission values (nm) were as follows: saccharin (340 and 407), naringin (324 and 460), caffeine (294 and 358), D-tryptophan (280 and 348), cyclo(Leu-Trp) (280 and 346), quinine (338 and 388), and propidium iodide (538 and 603). In these experiments, the degree of quenching of saccharin, naringin, caffeine, D-tryptophan, cyclo(Leu-Trp), quinine, and propidium iodide fluorescence by 500 mM KI was 93, 80, 75, 82, 60, 95, and 52%, respectively. Control CV papillae were treated in the same way, by application of the modified Tyrode solution without tastants. The resultant images were processed with Image-Pro Plus Media Cybernetics and Adobe Photoshop 6.0 software.



View larger version (74K):
[in this window]
[in a new window]
 
Fig. 1. In situ measurement of the dynamic intake of fluorescent amphipathic tastants by cells located in the inner trench area of an intact circumvallate (CV) papilla with confocal microscopy. Intact CV papillae were removed from rat tongues immediately after the animals were killed, laid on a slide, and viewed under the microscope. Images of the dorsal view were taken in the focal plane at which the inner trench containing the taste bud area around the center of the papilla is visible. Arrows indicate the inner trench circle, where most of the CV taste buds are located. The diameter of the center papilla, including the inner trench circle, is ~0.6 mm. Scale bars are 100 µm. Pictures were taken after the application of solutions of 10 mM saccharin (SAC), 1 mM cyclo(Leu-Trp) (CLT), 10 mM caffeine (CAF), 50 µg/ml propidium iodide (PI), and a control (Tyrode, no added tastant) (CON, bottom; 0, 2, and 8 min are shown). At time 0, immediately after solution was applied, fluorescence intensity was set to background and then monitored every minute for 6 min (0, 2, and 6 min are shown). At the end of 6 min of incubation, 500 mM KI (a membrane-impermeant fluorescence quencher) was added and imaging readings were continued for an additional 2 min. PI (fluorescent impermeant compound) was used as a positive control. Each image is of a representative experiment, replicated at least 4 times. Note that permeation of tastants also occurs in the surrounding nonsensory epithelial cells and that the addition of KI significantly reduced PI fluorescence without affecting tastant fluorescence.

 
Quantitative estimation of in vivo permeation of tastants into CV taste papilla cells with HPLC. Rats were anesthetized by injection of pentobarbital (60 mg/kg body wt). They were then laid on a 45°-angled stand in such a way that the oral cavity could be opened with a forceps and tastant solutions (in Tyrode) could be easily administered on and off into the oral cavity with a Pasteur pipette for a period of 90 s and could spill out from the oral cavity. Oral stimulation was conducted by administering 10 ml of 30 mM D-tryptophan, 2 mM quinine, 2 mM cyclo(Leu-Trp), or 10 mM caffeine. Because of the efflux of tastants during the subsequent collagenase treatment (see below), we selected tastants that are either fluorescent or strongly UV absorbing for HPLC-sensitive detection. Oral stimulation of control rats was carried out with solutions lacking the taste stimuli. The anesthetized animals were then immediately killed by decapitation. Tongues were rapidly removed and put into a beaker containing ice-cold Tyrode solution (40). Under a dissection microscope, a solution containing collagenase D (6 mg/ml) and trypsin inhibitor (4 mg/ml) in Tyrode was then injected submucosally at four or five locations around and under the single CV papilla. The whole tongue was then incubated in 20 ml of air-bubbled Tyrode solution at 30°C for 15 min. CV taste bud sheets were separated from the tongues under a dissection microscope as previously described (31, 40). The length of the entire process, from tongue removal, was ~25 min. Cells in taste bud sheets were then permeabilized by four freeze-thaw (–70°C) cycles in deionized water, and membranes were removed by centrifugation (30 min, 28,000 g, 4°C).

The intracellular content of various tastants was determined by HPLC equipped with UV-visible diode-array and fluorescent detectors according to Ref. 31 with some modifications. The UV-visible diode-array detector was set at 270 nm for saccharin and caffeine determinations, and separation was conducted with a LiChrospher 100 RP-18 column (5 µm, 250 mm, 4 mm; Merck) with an RP-18 precolumn. The mobile phase (isocratic separation) for the analysis of saccharin consisted of 1.5% (vol/vol) acetic acid in H2O and methanol (80:20 vol/vol) at a 0.45 ml/min flow rate. For caffeine analysis it consisted of H2O and methanol (50:50 vol/vol) at a 0.85 ml/min flow rate. The fluorescence detector was used for quinine, cyclo(Leu-Trp), and D-Trp determinations, and separation was conducted with a Purospher 55 RP-18 column (3 µm, 55 mm, 4 mm; Merck) with an RP-18 precolumn. The mobile phase (isocratic separation) for the analysis of quinine consisted of a mixture of acetonitrile and aqueous buffer containing (in mM) 10 KH2PO4, 25 SDS, and 3 tetrabutylammonium bromide, adjusted to pH 2.3 with orthophosphoric acid (47) (50:50 vol/vol) at a 0.9 ml/min flow rate. Acetonitrile with H2O (40:60 vol/vol) at a 0.7 ml/min flow rate was used for cyclo(Leu-Trp). The mobile phase for the analysis of D-tryptophan (linear gradient) was as for saccharin, except that the methanol was increased from 5% to 25% during the first 0–4 min and then returned to 5% methanol for 4–8 min, at a 1 ml/min flow rate.

Excitation at 338 nm and emission at 388 nm were used for quinine, whereas 280 and 348 nm were used for D-tryptophan and cyclo(Leu-Trp). Peaks of the tastants were identified and quantitated with known markers [Retention Time = 6.7, 5, 7.5, 4.3, and 3 min for saccharin, caffeine, quinine, D-tryptophan, and cyclo(Leu-Trp), respectively].

One might assume that during the 25-min collagenase treatments (after the 90-s taste stimulation), at least some of the tastants would be likely to leave the cells, possibly via multidrug resistance P-glycoprotein (MDR1; Ref. 18) or by simple diffusion. To estimate the amount of efflux of each tastant during the collagenase treatment, CV taste bud sheets were prepared from additional rats by collagenase separation. One-half of the CV tissue isolated from a single rat was then incubated with a tastant solution (1 of the above 4 stimuli at the same concentrations) for 30 s at 30°C. The second half of the same CV tissue was incubated with the same tastant for 30 s under the same experimental conditions. However, in this case, taste bud sheets were washed five times and left in Tyrode (at 30°C) for an additional 25 min. Taste bud sheets were then washed with Tyrode five times, cells were permeabilized (as already described), and the intracellular content of each tastant was determined by HPLC. The difference in the content of each tastant between the first half of the CV (initial intracellular concentration) and the second (final intracellular concentration) was used to estimate the efflux (calculated as percentage of the initial intracellular concentration) of tastants during the collagenase treatment. We assumed that the efflux of tastants from cells derived from isolated CV taste bud sheets is similar to that from taste cells of the CV before or during the collagenase treatment.

Experiments Designed to Identify GRK1, GRK2, GRK3, GRK5, GRK6, T2R4, and T1R3 in Taste Bud Cells

RT-PCR. CV taste bud and nonsensory epithelial sheets were prepared from rat tongues by subepithelial collagenase (type A) treatment (31, 40). Total RNA was isolated from pools of seven CV and nonsensory sheets with an RNeasy Protect Starter kit (Qiagen, Hilden, Germany) according to the manufacturer's instructions. cDNA was synthesized from 0.5 µg of RNA using a Reverse-iT first-strand synthesis kit (ABgene, Epsom, UK) according to the manufacturer's instructions. The cDNA was then amplified by PCR with a ReddyMix PCR Master Mix kit (ABgene), using specific primers for GRK1, GRK2, GRK3, GRK5, or GRK6 [believed to account for the regulation of most of the GPCRs found throughout the body (15)] and T2R4 or T1R3 taste receptors (all synthesized by the Oligonucleotide Synthesis Unit at the Weizmann Institute of Science, Rehovot, Israel). Specific primers for GAPDH (Metabion, Martinsried, Germany) were used as a control. GRK7 [apparently not present in the rat genome (44)] and GRK4 (thought to be expressed mainly in testes) were not analyzed.

The GRK1 primers were 5'-TTTCCTGCGGTTTCTTCAGT-3' (forward) and 5'-CACGTTCTCTGGCTTGAGGT-3' (reverse); the expected size of the PCR product was 460 bp. The GRK2 primers were 5'-CTTTGACATTGGCTCCTTTGA-3' (forward) and 5'-GTGGCTTGTTCTTCATCTTGG-3' (reverse); the expected size of the PCR product was 559 bp. The GRK3 primers were 5'-ACTGGCTTTGAACGAGAGGA-3' (forward) and 5'-GTCGATGCCCTTGAAGAAGA-3' (reverse); the expected size of the PCR product was 670 bp. The GRK5 primers were 5'-CTGGCTTTGAGGAAGAACGA-3' (forward) and 5'-GTTTTGATGCTGACGCCTGA-3' (reverse); the expected size of the PCR product was 841 bp. The GRK6 primers were 5'-AGGTGACCCCAGATGAGAAA-3' (forward) and 5'-TCTCGGCAGCATAGAAGACA-3' (reverse); the expected size of the PCR product was 612 bp.

The T2R4 primers were 5'-CCACACATACTTACCTTTGGA-3' (forward) and 5'-GAGTAGGTTGTTTTGTGGCAG-3' (reverse); the expected size of the PCR product was 560 bp. The T1R3 primers were 5'-CATGACCTCTCACCCAAGGTA-3' (forward) and 5'-CATAGCAGCAGGAATGAAAGC-3' (reverse); the expected size of the PCR product was 705 bp. The GAPDH primers were 5'-TCCGCCCCTTCCGCTGATG-3' (forward) and 5'-CACGGAAGGCCATGCCAGTGA-3' (reverse); the expected size of the PCR product was 350 bp.

PCR was performed with one cycle at 94°C for 5 min, then 30 cycles of 94°C for 1 min, 52°C for 1 min, 72°C for 1 min, followed by one cycle at 72°C for 10 min. cDNA was electrophoresed on a 1.5% (wt/vol) agarose gel in the presence of ethidium bromide. In control experiments, in which PCR was carried out with GRK2 primers on RNA without including the reverse transcription step, no PCR product was observed, confirming that the total RNA was not contaminated with genomic DNA. The PCR products were purified with a PCR purification kit (Qiagen) and sequenced at the Sequencing Unit of the Weizmann Institute of Science.

Immunoblot analysis for antibody specificity. Rat brains were homogenized in RIPA buffer (20 mM Tris, 137 mM NaCl, 10% glycerol, 0.1% wt/vol SDS, 0.5% wt/vol deoxycholate, 1% vol/vol Triton X-100, 2 mM EDTA, 1 mM PMSF, 20 µM leupeptin) and centrifuged at 20,000 g for 15 min at 4°C. The supernatant was removed and incubated with sample buffer at 95°C for 5 min after a sample was taken for protein concentration. Total protein from the brain extract was loaded on a 12% SDS-PAGE (20 µg/lane) and transferred to a nitrocellulose membrane. The membrane was incubated with blocking solution [2% wt/vol BSA in Tris-buffered saline Tween (TBST), and each lane was incubated at room temperature for 1 h with the appropriate antibody [GRK2 (1:5,000), GRK3 (1:500), GRK5 (1:1,000), or GRK6 (1:1,000)], with or without its blocking peptide (1 µg/ml). Peroxidase-conjugated AffiniPure goat anti-rabbit IgG (H+L) was used as a secondary antibody.

Immunohistochemistry. Immunohistochemistry was performed essentially according to Ref. 10. Rats were deeply anesthetized with pentobarbital (60 mg/kg body wt) and perfused through the left ventricle, first with Hanks’ buffer (in mM: 137 NaCl, 5.3 KCl, 0.3 Na2HPO4, 0.8 MgSO4, 1 MgCl2, 5 HEPES, 6.7 tricine, 28 glucose, 0.3 NaHCO3, and 0.96 EGTA, pH 7.3) and then with a fixative containing 3% (wt/vol) paraformaldehyde in a 0.1 M phosphate buffer (pH 7.3) for 10 min. After perfusion, tongues were removed and cryoprotected with 20% (wt/vol) sucrose in 0.1 M phosphate buffer overnight at 4°C. The tongues were then frozen in dry ice-cooled isopentane. Cryostat sections (10 µm) from CV papillae (visualized under a light microscope) were collected sequentially onto SuperFrost Plus slides (Menzel-Glazer, Braunschweig, Germany), air dried for 30 min, and kept at –20°C until use. Sections were distributed to different slides to ensure that controls and treated sections represented the same area of the CV papilla. Sections were incubated for 1 h at room temperature in a blocking solution containing 10% (vol/vol) goat serum (to reduce nonspecific binding) and 0.3% Triton X-100 in 0.1 M phosphate buffer (pH 7.3). Sections were then incubated in primary antibodies specific for GRK2, GRK5, or GRK6 (1:100 in blocking solution) in a closed, moist chamber for 24 h at 4°C. After being washed (3 times) with 1x PBS, the tissues were exposed to secondary FITC-conjugated goat anti-rabbit IgG (1:200 in PBS) for 1 h at room temperature. Sections were washed, mounted, and viewed under a Zeiss confocal fluorescent microscope with a Plan Nofluar x40/1.3 NA lens. Control samples were treated similarly with modifications that included omission of the primary antisera or addition of 25 µg/ml blocking peptide to the primary antiserum solution. The resultant images were processed with Image-Pro Plus Media Cybernetics and Adobe Photoshop 6.0 software.

Phosphorylation Assays

GRK2 (1.5–1.8 pmol/tube), GRK5 (20 pmol/tube) or PKA (5 U/tube) were preincubated with and without tastants (at the concentrations indicated in Figs. 4 and 5) in assay buffer (in mM: 20 Tris·HCl pH 7.4, 10 MgCl2, 2 EDTA, and 1 DTT) for 15 min at 30°C. Samples containing cyclo(Leu-Trp) and limonin also contained 0.1% (vol/vol) DMSO. When tastants contained additional cations (e.g., sodium saccharin and sodium cyclamate) or potassium (acesulfame K), the same concentrations of either NaCl or KCl were used as specific controls. The reaction (15 min at 30°C) under light (when rhodopsin was used) in the same buffer was started by adding substrates (rod outer membrane segments containing rhodopsin to a final concentration of 0.5 µM or 20 µg casein) and 100 µM [{gamma}-32P]ATP (1–2 µCi). After incubation, 12 µl of sample loading buffer (100 mM Tris·HCl pH 7.2, 4% SDS, 200 mM DTT, 0.25% wt/vol pyronin Y, and 20% glycerol) were added to terminate the 25-µl reaction. Samples were then subjected to SDS-PAGE. The gels were dried and exposed (14 h) to a PhosphorImager screen (Fuji Photo Film, Nakagawa, Japan). Phosphorylation was quantitated with Image J software and the mean of at least three independent image experiments, performed with and without tastants, which expressed the phosphorylation level as percentage of controls performed without any added tastants. Heparin was the positive control for inhibition of GRK2 and GRK5, and the PKA inhibitor amide, fragment 6–22, was the control for PKA.



View larger version (41K):
[in this window]
[in a new window]
 
Fig. 4. Amphipathic tastants inhibit GRK2- and GRK5-induced phosphorylation of rhodopsin and PKA-induced phosphorylation of casein. GRK2 (1.5–1.8 pmol/tube) or GRK5 (20 pmol/tube) with 0.5 µM rhodopsin or PKA (5 U/tube) with 20 µg casein was incubated with 100 µM [{gamma}-32P]ATP (1–2 µCi) for 15 min with the following tastants: caffeine (Caf, 20 mM), quinine (Quin, 1 mM), L-tryptophan (L-Trp, 20 mM), D-tryptophan (D-Trp, 20 mM), sodium cyclamate (Cyc, 20 mM), sodium saccharin (Sac, 20 mM), acesulfame K (Ac-K, 3.5 mM), limonin (Lim, 100 µM), cyclo(Leu-Trp) (CLT, 0.5 mM), neohesperidin dihydrochalcone (NHD, 1.25 mM), and naringin (Nar, 1 mM). Results (% of kinase activity obtained during incubation without tastants) are the means ± SE of 4 independent experiments. *P < 0.05, **P < 0.01, lower values than controls incubated without tastants.

 


View larger version (28K):
[in this window]
[in a new window]
 
Fig. 5. Tastants inhibit GRK2 (A and B)- and GRK5 (C and D)-induced phosphorylation of rhodopsin in a concentration-dependent manner. GRK2 and GRK5 were incubated with rhodopsin as indicated in Fig. 4 with the indicated concentrations of NHD, saccharin, caffeine, or naringin. Note that for saccharin phosphorylation, each concentration shows 2 bands, 1 for NaCl (control for sodium) and 1 for sodium saccharin. CON, phosphorylation level when tastants were not present. Results are means ± SE of 3 independent experiments.

 
Rhodopsin and ATP Kinetics

Rhodopsin and ATP concentration effects on NHD- and D-tryptophan-inhibited phosphorylation produced by GRK2 were analyzed with two concentrations of each tastant. ATP concentration ranged from 0 to 1.5 mM. Rhodopsin concentration ranged from 0 to 2.1 µM. The increase in ATP concentration came from the addition of cold ATP, the level of labeled ATP being kept constant (3–4 µCi). To keep specific activity constant in the final quantitative determination, the phosphorylation results for each ATP concentration were multiplied by the dilution factor.

Data Analysis

Mean tests using JMP statistical software (SAS Institute, Cary, NC) were performed on the quantitative estimation values derived from the tastant permeation data, Michaelis-Menten kinetics data were calculated with SigmaPlot 8.0 (SPSS, Chicago, IL), and ANOVA (JMP) was performed for the phosphorylation inhibition and kinetics data. Post hoc comparisons to determine the differences between the means were performed by either Tukey-Kramer honestly significant difference and Student's t-tests (phosphorylation data) or nonoverlapping confidence intervals (kinetics).


    RESULTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
Permeation of Amphipathic Tastants into Taste Bud Cells

Exposure of intact CV papillae to selected amphipathic tastants revealed the dynamics of tastant permeation in situ under confocal microscopy (Fig. 1) . A dorsal view shows the increased fluorescence of tastants with time, located around the circular inner trench (Fig. 1, arrows) where the taste buds are found. Tastant fluorescence was also observed in the surrounding nonsensory epithelium. With this technique, an increase in fluorescence after tastant application was already visible after 1 min; however, the 0- to 6-min exposure range was selected to show the continuing increase in fluorescence intensity. Application of the membrane-impermeant fluorescence quencher KI had no effect on the fluorescence intensity contributed by saccharin, caffeine, or cyclo(Leu-Trp) (Fig. 1) or that contributed by D-tryptophan, naringin, or quinine (data not shown). This indicated that tastant fluorescence was protected from quenching by KI because it occurred inside the CV cells. Such tastant permeation also occurred in epithelial tissue that did not contain taste buds. In contrast, fluorescence of the membrane-impermeant propidium iodide (used as a positive control) applied to CV taste cells was significantly quenched by KI. Together, these results indicate that tastants permeate deeply into the cytosolic side of the plasma membrane or the cytosol.

Moreover, a 90-s oral stimulation of anesthetized rats by similar tastants clearly indicated tastant permeation into the cytosol of the CV taste bud cells (or into the cytosolic side of the cell membrane), as evidenced by HPLC determinations (Table 1). Tastant permeation was also evident in epithelial cells lacking taste buds (data not shown).


View this table:
[in this window]
[in a new window]
 
Table 1. Estimated permeation of amphipathic tastants into CV taste bud cells via apical side

 
After the 90-s incubation period, the intracellular concentration of quinine was almost threefold that present in the extracellular medium, indicating permeation against a concentration gradient. The efflux (or the rate of tastant disappearance inside the taste cells) was significant for all tastants during the 25-min collagenase treatment, ranging between 64% and 86% (Table 1).

Presence of GRK2, GRK3, GRK5, GRK6, T2R4, and T1R3 in Taste Bud Cells

GRK5 has been shown to be present in CV papillae (33); however, expression of the other GRKs in taste bud cells was never verified. RT-PCR (Fig. 2) indicated that the signal termination-related kinases GRK2, GRK3, GRK5, and GRK6 (but not GRK1) are present in the CV taste bud sheet together with the bitter (T2R4) and sweet (T1R3) taste receptors. Specific primers for these genes were used for RT-PCR with RNA isolated from CV tissue, and agarose gel electrophoresis yielded PCR products of the expected sizes in all six tested proteins. Parallel PCR of RNA without the reverse transcriptase step did not show any bands, indicating no genomic DNA contamination. PCR products were sequenced and found identical to these four kinase genes in rats with the BLAST program [GenBank, National Center for Biotechnology Information]. Similar RT-PCR experiments indicated the presence of GRK2, GRK3, GRK5, and GRK6 in the nonsensory epithelial sheets but, as expected, no bands for the T2R4 and T1R3 receptors.



View larger version (58K):
[in this window]
[in a new window]
 
Fig. 2. RT-PCR analysis of G protein-coupled receptor kinase (GRK)1, GRK2, GRK3, GRK5, GRK6, T2R4, and T1R3 mRNA in CV taste bud sheets and nonsensory epithelium (EP). cDNA was synthesized from CV and EP RNA and then amplified by PCR using specific primers for GRK1, GRK2, GRK3, GRK5, GRK6, T2R4, or T1R3; GAPDH was used as an internal reference gene. CON, parallel PCR omitting the RT step and using GRK2 primers. PCR products were separated on a 1.5% agarose gel stained with ethidium bromide. The expected PCR product sizes were 460 bp for GRK1, 559 bp for GRK2, 670 bp for GRK3, 841 bp for GRK5, 612 bp for GRK6, 560 bp for T2R4, 705 bp for T1R3, and 350 bp for GAPDH. M, 100-bp DNA marker ladder (Bioneer, Daejeon, Korea).

 
To further explore the presence of GRKs in CV taste cells, we examined the distribution of GRK2, GRK3, GRK5, and GRK6 antibodies in 10-µm sections prepared from CV papillae in an area where CV taste buds are clearly visible (Fig. 3A). Very weak immunostaining of GRK3 was noted (data not shown). However, strong immunostaining of GRK2, GRK5, and GRK6 was found: GRK5 was particularly noticeable in the taste bud cells, whereas GRK2 and GRK6 were essentially stained in the surrounding epithelium, and their presence in taste bud cells remains questionable.



View larger version (93K):
[in this window]
[in a new window]
 
Fig. 3. A: confocal microscopy showing the immunofluorescent staining pattern for GRK2-, GRK5-, and GRK6-like immunoreactivity in a rat CV papilla. CV cryostat sections (10 µm) were incubated in a blocking solution containing 10% goat serum and 0.3% Triton X-100 in 0.1 M phosphate buffer (pH 7.3). Sections were then incubated with primary antibodies specific for GRK2, GRK5, or GRK6 for 24 h at 4°C and washed with 1x PBS, and tissues were exposed to secondary FITC-conjugated goat anti-rabbit antibody. Mounted sections were viewed by confocal microscopy. Control (CON), sections treated with secondary but no primary antibody; Ab, sections treated with primary and secondary antisera; Ab+BP, sections treated for GRK2, GRK5, or GRK6, with the appropriate blocking peptides (BP) (25 µg/ml) added to the primary antisera. Arrows indicate taste-bud cells in the CV papilla. Scale bars are 20 µm. B: immunoblot analysis for GRK2, GRK5, and GRK6 antibody specificity. Total protein from brain homogenate (20 µg) was subjected to 12% SDS-PAGE and incubated with the appropriate antibody with or without blocking peptide (1 µg/ml). Note that in the presence of blocking peptide, GRK bands either disappear or are significantly reduced. Dilutions of primary antibodies (1st Ab) were 1:5,000 for GRK2 and 1:1,000 for GRK5 and GRK6.

 
To verify the specificity of each of the antibodies used for immunostaining, a Western blot was performed, using protein extract from brain homogenate. As shown in Fig. 3B, GRK2, GRK5, and GRK6 antibodies each showed one main band at the expected weight (GRK2 at ~80 kDa , GRK5 and GRK6 at ~65 kDa). These bands either disappeared or were significantly reduced when incubated with their respective blocking peptides. Thus the antibodies were specific enough to be used for immunostaining.

Inhibition of GRK2- and GRK5-Phosphorylated Rhodopsin and PKA-Phosphorylated Casein in Vitro

Initial experiments were run to determine the enzyme concentrations needed to carry out enzymatic activities in the linear range. Depending on the kinase tested, the various amphipathic nonsugar sweeteners cyclamate, saccharin, NHD, and D-tryptophan and the bitter tastants caffeine, quinine, limonin, and naringin were found to inhibit GRK2- and GRK5-phosphorylated rhodopsin (a GPCR) and PKA-phosphorylated casein in vitro (Figs. 4 and 5). Caffeine was most active in the inhibition of the three kinases (Fig. 4). Together with caffeine, L-tryptophan, cyclamate, and saccharin were very efficient at inhibiting GRK2, whereas caffeine, quinine, L-tryptophan, saccharin, NHD, and naringin were efficient inhibitors of GRK5. Caffeine, cyclamate, limonin, and NHD had the most efficient inhibitory effect on PKA. The sweetener acesulfame K and the bitter peptide cyclo(Leu-Trp) appeared to be the least effective kinase inhibitors in this experimental system. A concentration dependence, tested for NHD, saccharin, caffeine, and naringin, was evident (Fig. 5), and depending on solubility, 90–100% inhibition could be shown. Inhibition could already be observed at relatively low concentrations, e.g., 2.5 mM caffeine, 600 µM NHD, 125 µM naringin (Fig. 5), and 100 µM limonin (Fig. 4). We tried two disaccharides, sucrose (sweet to humans) and melibiose (essentially tasteless to humans): neither affected GRK activity when applied at concentrations of up to 200 mM. About 20% inhibition was found when higher concentrations of the two sugars were applied (data not shown).

Kinetics of Phosphorylation

Inhibition of GRK2-phosphorylated rhodopsin by NHD and D-tryptophan produced saturation curves (R2 = 0.97) when either rhodopsin (Fig. 6, A and B) or ATP (Fig. 6, C and D) concentrations were increased. NHD and D-tryptophan did not significantly modify the apparent Km values of GRK2 for rhodopsin (Fig. 6, A and B). In the NHD system, Km values (µM) were 0.71 ± 0.23, 0.79 ± 0.18, and 0.73 ± 0.22 for 0, 0.63, and 1.25 mM NHD, respectively. In the D-tryptophan system, Km values (µM) were 0.37 ± 0.08, 0.35 ± 0.05, and 0.43 ± 0.15 for 0, 5, and 15 mM D-tryptophan, respectively. On the other hand, both NHD and D-tryptophan reduced the Vmax values. In the NHD system, Vmax values (relative to controls monitoring phosphorylation level without the presence of a tastant) were 0.76 ± 0.08 and 0.35 ± 0.05 (P < 0.05) for 0.63 and 1.25 mM, respectively. In the D-tryptophan system, Vmax values were 0.73 ± 0.04 and 0.49 ± 0.06 (P < 0.05) for 5 and 15 mM, respectively.



View larger version (31K):
[in this window]
[in a new window]
 
Fig. 6. Michaelis-Menten equation curves: Km and relative Vmax values for the effect of rhodopsin (A and B) and ATP (C and D) concentrations on NHD and D-tryptophan inhibition of GRK2-phosphorylated rhodopsin. A and B: GRK2 was incubated with rhodopsin (from 0 to 2.1 µM) and NHD or D-tryptophan as indicated in MATERIALS AND METHODS. A: {bullet}, control (no tastant added); {circ}, 0.63 mM NHD; {blacktriangledown}, 1.25 mM NHD. B: {bullet}, control; {circ}, 5 mM D-tryptophan; {blacktriangledown}, 15 mM D-tryptophan. C and D: GRK2 was incubated with ATP (0–1.5 mM) and NHD or D-tryptophan as indicated in MATERIALS AND METHODS. C: {bullet}, control (no tastant added); {circ}, 0.83 mM NHD; {blacktriangledown}, 1.25 mM NHD. D: {bullet}, control; {circ}, 3 mM D-tryptophan; {blacktriangledown}, 6 mM D-tryptophan. Values are the means of 3 independent experiments for each data point.

 
Similarly, NHD and D-tryptophan did not significantly modify the apparent Km values of GRK2 for ATP (Fig. 6, C and D). In the NHD system, Km values (mM) were 0.30 ± 0.03, 0.11 ± 0.05, and 0.37 ± 0.18 for 0, 0.825, and 1.25 mM NHD, respectively (Fig. 6C). In the D-tryptophan system, Km values (mM) were 0.31 ± 0.03, 0.31 ± 0.12, and 0.35 ± 0.08 for 0, 3, and 6 mM D-tryptophan, respectively (Fig. 6D). Again, both NHD and D-tryptophan reduced the Vmax values. In the NHD system, Vmax values were 0.58 ± 0.06 and 0.48 ± 0.08 (P < 0.05) for 0.825 and 1.25 mM, respectively. In the D-tryptophan system, Vmax values were 0.60 ± 0.08 and 0.47 ± 0.03 (P < 0.05) for 3 and 6 mM, respectively.


    DISCUSSION
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
Desensitization of GPCRs can be initiated when their cytosolic residues, containing either serine or threonine, are phosphorylated by selected kinases (32). Therefore, the hypothesis that amphipathic tastants interact with GRKs to affect signal termination requires that such tastants be able to permeate the cells under physiological conditions. In our previous experiments (31), we showed that amphipathic tastants can permeate taste cells rapidly and against a concentration gradient when isolated taste bud preparations after a collagenase treatment are used. In that procedure, the basolateral side of the taste buds, not only their apical side (the route by which tastants interact with sensory taste cells), may have been exposed. In the present study, we took advantage of the autofluorescence of some of these tastants in two procedures designed to explore whether these tastants can permeate taste cells only through the apical side of the taste buds (taste pore). First, in situ imaging after the application of six amphipathic tastants to an intact CV papilla surgically removed without collagenase treatment indicated evident dynamics of tastant permeation into the CV taste bud cells located around the circular inner trench (Fig. 1, arrows). To observe the autofluorescence of these tastants more clearly (some are weak fluorescent compounds) under a confocal microscope, longer exposure times (i.e., up to 6 min) were used. Most important, the membrane-impermeant quencher KI had no effect on tastant fluorescence, indicating the presence of the tastant on the cytosolic side or in the cytosol of the taste bud cells.

In the second procedure, we stimulated the oral cavity of anesthetized rats with tastant solutions for 90 s, followed by immediate animal death and an HPLC determination of the tastants' intracellular concentrations: millimolar levels of these tastants were found inside the taste cells. After oral stimulation, the entire separation process removed any tastant present on the extracellular surface of the cells. Analysis indicated only traces of tastant in the membranes: most was present in the cytosol. This indicates that tastants indeed permeated the membrane through to the cytosol and/or were released from the cytosolic side of the membrane during centrifugation. In either case, one can assume their potential access to GRKs or other membrane signal transduction components. Permeation of tastants into nonsensory epithelial cells was also observed (data not shown), as already reported when isolated taste bud sheets were used (31). The high sensitivity of the HPLC UV-visible diode-array and fluorescent detectors allowed us to detect the nanogram intracellular concentrations of these tastants in the ~25-µg dry weight of a single CV taste bud sheet (40). The use of such a procedure was therefore appropriate for membrane-permeant tastants that show strong UV absorbance or fluorescence. Furthermore, one would assume that during the 25-min collagenase treatment after the oral stimulation, applied to separate the CV taste bud sheets from other epithelial, connective, and muscle tissues (40), tastants would leave the cells to the extracellular media, e.g., via MDR1 (18) or by simple diffusion. Indeed, parallel experiments (see MATERIALS AND METHODS) indicated significant (64–86%) efflux, which was used to correct the estimated intracellular level of each tastant during the 90-s oral stimulation (Table 1). Together the results presented in Fig. 1 and Table 1 provide evidence that these and perhaps additional (though not necessarily all) amphipathic tastants rapidly permeate taste cells under physiological conditions, either through the taste bud pore or through the tight junction, and that such permeation also occurs in nonsensory lingual epithelial cells.

An additional requirement for the hypothesized interaction between membrane-permeant tastants and GRKs is the presence of GRKs in the same cells. GRK5 and potentially GRK2 have been previously identified in CV papilla (33), but there does not seem to be any evidence of their presence in taste cells. The presence of the bitter T2R (1) and the sweet (e.g., T1R3 and T1R2) (22, 26) taste receptors in subpopulations of cells present in rodent CV and other taste papillae has been demonstrated. Here, RT-PCR experiments (Fig. 2) suggested the presence of T2R4 and T1R3 along with GRK2, GRK3, GRK5, and GRK6 in isolated taste bud sheets of rat CV papillae. The same GRKs were found to be expressed in nonsensory epithelium, but, as expected, no PCR products for the T2R4 and T1R3 receptors were observed in nonsensory epithelial cells.

Furthermore, antibodies against GRK2, GRK5, and GRK6 (with weak staining of GRK3) yielded clear, positive immunostaining in the 10-µm frozen sections of the CV papilla (Fig. 3). However, only GRK5 was clearly stained in the CV taste bud cells; GRK2 and GRK6 were notably stained in the surrounding epithelium, but their presence in the taste cells remains questionable. These immunostaining experiments cannot provide evidence of T2R4 and T1R3 receptor expression in the subpopulation of taste cells containing the GRKs. However, because GRK5 appeared to be expressed in almost all taste bud cells, it is likely that it coexpresses in cells expressing T2R4 and T1R3.

Subsequently, the effect of tastants on GRK2- and GRK5-phosphorylated rhodopsin, a well-studied model for GPCR phosphorylation (32), was investigated (Figs. 46). The amphipathic nonsugar sweeteners cyclamate, saccharin, NHD, and D-tryptophan and the bitter tastants caffeine, quinine, limonin, and naringin, with diverse chemical structures, were found to inhibit GRK2- and GRK5-phosphorylated rhodopsin (a GPCR) and PKA-phosphorylated casein in vitro. Their effects depended on the kinase being tested. The nonamphipathic compounds sucrose and melibiose did not affect GRK activity when applied at concentrations of up to 200 mM. It should be noted that ~20% inhibition was found with these latter compounds at higher concentrations that are not relevant to intact mammalian cells; this minor inhibition was probably related to nonspecific osmotic factors.

Naringin (as well as some other flavonoids) has been found to inhibit phosphorylation, e.g., naringin inhibits AMP-activated protein kinase-activating kinase (20). Caffeine and theophylline have been recently found to be phosphoinositide 3-kinase inhibitors (14). However, nothing has been reported in relation to GRKs or PKA. In the present study, although inhibition could already be observed at relatively low concentrations, e.g., 2.5 mM caffeine, 600 µM NHD, 125 µM naringin (Fig. 5), and 100 µM limonin (Fig. 4), the tastant concentrations needed for kinase inhibition under the experimental conditions were at the millimolar level, higher than the micromolar levels usually used with other kinase inhibitors in vitro and in vivo medically (36) or experimentally in nontaste systems (43). Moreover, these tastants do not appear to be very specific because they inhibited PKA as well as the GRKs and may very well inhibit additional kinases. This suggests low tastant potency and raises the question of whether such inhibition has any physiological relevance. Nevertheless, the range of tastant concentrations applied here matches that used in taste stimulation during sensory and biochemical studies (16, 26, 35, 41, 45, 46), and the tastants were found inside the cells within seconds of their extracellular application (Table 1 and Ref. 31). The same tastants were also demonstrated recently to permeate other epithelial cells in situ, e.g., Xenopus laevis melanophores (48). Therefore, although these relatively high concentrations of amphipathic tastants suggest low potency, their physiological significance as kinase inhibitors in taste cells may result from their access to the cytosolic side of the taste cells and thus to GRK2 and GRK5, which are present in taste tissue (Figs. 2 and 3), and to PKA, which has been indirectly shown to be active in taste cells (42).

The mechanism(s) by which the tastants inhibit GRK-phosphorylated rhodopsin and PKA-phosphorylated casein is unclear. The inhibition of GRK2-phosphorylated rhodopsin by NHD and D-tryptophan produced a saturation curve with increasing concentrations of both rhodopsin and ATP (Fig. 6). NHD and D-tryptophan did not modify the apparent Km values of GRK2 for either substrate (rhodopsin or ATP), but the relative Vmax values were significantly reduced in the presence of both NHD and D-tryptophan. Therefore, the inhibition of GRK2 activity by both tastants appears to be noncompetitive, i.e., neither the ATP pocket nor the binding site for rhodopsin in the GRK2 molecule seems to be involved in the mechanism(s) of phosphorylation inhibition. Because tastants inhibited PKA when its catalytic unit was used (Fig. 4), such inhibition may be related to this part of the enzyme. Competitive, noncompetitive and uncompetitive types of inhibition have been found for various kinase inhibitors, and in many cases, the molecular events are unknown. When competitive inhibition is observed, it is usually related to interaction of the inhibitor with the ATP-binding pocket in the kinase and is noncompetitive with respect to the protein substrate (13, 29). Some inhibitors, depending on the kinase system, may act via two different mechanisms. The flavonoid genistein, for example, a known competitive inhibitor of ATP in some kinase systems (2, 43), can also induce both an increase in apparent Km and a decrease in Vmax values of adenylate kinase, suggesting uncompetitive inhibition (34). The slight variation in Km values for the controls in the NHD vs. D-tryptophan experiments (Fig. 6) was probably related to the different lots of GRK2 and rhodopsin used in the two experiments. The differences between the apparent Km values observed here for GRK2-rhodopsin and GRK2-ATP and those originally published (6 and 35 µM for rhodopsin and ATP, respectively; Ref. 4) may be related, in part, to the presence of glycerol (0.2%, 27 mM) in our experiments and its absence in the original studies (J. L. Benovic, personal communication). For example, we tested this level of glycerol in the PKA-casein incubation system and obtained ~30% inhibition of PKA activity. Therefore, although the presence of glycerol is important for kinase stability during storage, it may affect enzyme activity and perhaps its affinity for substrates.

In summary, a new hypothesis for the delayed taste termination caused by bitter tastants and nonsugar sweeteners is proposed. This hypothesis is based on results indicating that 1) such tastants permeate taste cells under physiological conditions, 2) GRKs are present in taste cells, and 3) such tastants inhibit the GRK- and PKA-induced phosphorylation of a GPCR in vitro. Importantly, tastant concentrations were compatible with those used in taste stimulation and those permeating taste cells. Additional studies using the newly discovered taste GPCRs are needed to show their interaction with GRKs and possibly with other kinases, such as in intact cells in vivo, before anything can be unequivocally stated. The novelty of the proposed mechanism of signal termination may lie in the fact that the ligands themselves not only interact extracellularly with GPCRs to initiate the transduction chain, but may concomitantly interact intracellularly with downstream shutoff components to affect signal termination. The fact that tastants inhibit PKA and not just GRKs suggests that they inhibit other kinases as well. Because these tastants are components of our daily diets and may access other tissues along the gastrointestinal tract, these results may have implications for cellular signaling in tissues other than those involved in taste. Finally, the studied tastants, although not as potent as other kinase inhibitors, are membrane permeant, making them desirable for research purposes.


    GRANTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
This study was supported by Grant IS-3366 from The US-Israel Binational Agricultural Research and Development Fund (BARD) and, in part, by Grant 2003015 from the US-Israel Binational Science Foundation (BSF).


    ACKNOWLEDGMENTS
 
We thank Dr. R. J. Lefkowitz of Duke University for the generous supply of purified GRKs and rhodopsin and D. Capel for technical advice. We also thank Prof. R. Seger, Dr. M. Schonfeld, Dr. N. Moran, Prof. A. I. Spielman, and Dr. L. Huang for helpful comments and Dr. M. Tarshish for confocal microscopy.

Preliminary results of this study were presented at the European Signal Transduction Conference (abstract proceedings, p. 56, Luxemburg, January 2004), at the Experimental Biology 2004 Meeting (FASEB J 18: A313, 2004), and at the XVI ECRO Congress (abstract proceedings, p. 65, Dijon, France, September 2004).


    FOOTNOTES
 

Address for correspondence: M. Naim, Inst. of Biochemistry, Food Science and Nutrition, The Hebrew Univ. of Jerusalem, PO Box 12, Rehovot 76-100, Israel (e-mail: naim{at}agri.huji.ac.il)

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.


    REFERENCES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 GRANTS
 REFERENCES
 
1. Adler E, Hoon MA, Mueller KL, Chandrashekar J, Ryba NJP, and Zuker CS. A novel family of mammalian taste receptors. Cell 100: 693–702, 2000.[CrossRef][ISI][Medline]

2. Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, Shibuya M, and Fukami Y. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem 262: 5592–5595, 1987.[Abstract/Free Full Text]

3. Bachmanov AA, Li X, Reed DR, Ohmen JD, Li S, Chen Z, Tordoff MG, de Jong PJ, Wu C, West DB, Chatterjee A, Ross DA, and Beauchamp GK. Positional cloning of the mouse saccharin preference (Sac) locus. Chem Senses 26: 925–933, 2001.[Abstract/Free Full Text]

4. Benovic JL, Mayor F Jr, Staniszewski C, Lefkowitz RJ, and Caron MG. Purification and characterization of {beta}-adrenergic receptor kinase. J Biol Chem 262: 9026–9032, 1987.[Abstract/Free Full Text]

5. Bernhardt SJ, Naim M, Zehavi U, and Lindemann B. Changes in IP3 and cytosolic Ca2+ in response to sugars and non-sugar sweeteners in transduction of sweet taste in the rat. J Physiol 490: 325–336, 1996.[Abstract]

6. Birch GG, Latymer Z, and Hollaway M. Intensity/time relationships in sweetness: evidence for a queue hypothesis in taste chemoreception. Chem Senses 5: 63–78, 1980.[ISI]

7. Bufe B, Hofmann T, Krautwurst D, Raguse JD, and Meyerhof W. The human TAS2R16 receptor mediates bitter taste in response to {beta}-glucopyranosides. Nat Genet 32: 397–401, 2002.[CrossRef][ISI][Medline]

8. Chandrashekar J, Mueller KL, Hoon MA, Adler E, Feng L, Guo W, Zuker CS, and Ryba NJP. T2Rs function as bitter taste receptors. Cell 100: 703–711, 2000.[CrossRef][ISI][Medline]

9. Chaudhari N, Landin AM, and Roper SD. A metabotropic glutamate receptor variant functions as a taste receptor. Nat Neurosci 3: 113–119, 2000.[CrossRef][ISI][Medline]

10. Clapp TR, Yang R, Stoick CL, Kinnamon SC, and Kinammon JC. Morphologic characterization of rat taste receptor cells that express components of the phospholipase C signaling pathway. J Comp Neurol 468: 311–321, 2004.[CrossRef][ISI][Medline]

11. Darzynkiewicz Z, Bruno S, Del Bino G, Gorczyca W, Hotz MA, Lassota P, and Traganos F. Features of apoptotic cells measured by flow cytometry. Cytometry 13: 795–808, 1992.[ISI][Medline]

12. DeSimone JA. Focus on "Rapid entry of bitter and sweet tastants into liposomes and taste cells: implications for signal transduction". Am J Physiol Cell Physiol 278: C13–C16, 2000.[Free Full Text]

13. Fabbro D and Garcia-Echeverria C. Targeting protein kinases in cancer therapy. Curr Opin Drug Discov Devel 5: 701–712, 2002.[ISI][Medline]

14. Foukas LC, Daniele N, Ktori C, Anderson KE, Jensen J, and Shepherd PR. Direct effects of caffeine and theophylline on p110{delta} and other phosphoinositide 3-kinases. Differential effects on lipid kinase and protein kinase activities. J Biol Chem 277: 37124–37130, 2002.[Abstract/Free Full Text]

15. Gainetdinov RR, Premont RT, Bohn LM, Lefkowitz RJ, and Caron MG. Desensitization of G protein-coupled receptors and neuronal functions. Annu Rev Neurosci 27: 107–144, 2004.[CrossRef][ISI][Medline]

16. Hellekant G. The physiology of thaumatin. In: Thaumatin, edited by Witty M and Higginbotham JD. London: CRC, 1994, p. 99–113.

17. Inglese J, Koch WJ, Caron MG, and Lefkowitz RJ. Isoprenylation in regulation of signal transduction by G-protein-coupled receptor kinases. Nature 359: 147–150, 1992.[CrossRef][ISI][Medline]

18. Jakob I, Hauser IA, Thevenod F, and Lindemann B. MDR1 in taste buds of rat vallate papilla: functional, immunohistochemical, and biochemical evidence. Am J Physiol Cell Physiol 274: C182–C191, 1998.[Abstract/Free Full Text]

19. Lange R, Anzenbacher P, Muller S, Maurin L, and Balny C. Interaction of tryptophan residues of cytochrome P450scc with a highly specific fluorescence quencher, a substrate analogue, compared to acrylamide and iodide. Eur J Biochem 226: 963–970, 1994.[Abstract]

20. Larsen AKR, Moller MTN, Blankson H, Samari HR, Holden L, and Seglen PO. Naringin-sensitive phosphorylation of plectin, a cytoskeletal cross-linking protein, in isolated rat hepatocytes. J Biol Chem 277: 34826–34835, 2002.[Abstract/Free Full Text]

21. Margolskee RF. Molecular mechanisms of bitter and sweet taste transduction. J Biol Chem 277: 1–4, 2002.[Free Full Text]

22. Max M, Shanker YG, Huang L, Rong M, Liu Z, Campagne F, Weinstein H, Damak S, and Margolskee RF. Tas1r3, encoding a new candidate taste receptor, is allelic to the sweet responsiveness locus Sac. Nat Genet 28: 58–63, 2001.[CrossRef][ISI][Medline]

23. Montmayeur JP, Liberles SD, Matsunami H, and Buck LB. A candidate taste receptor gene near a sweet locus. Nat Neurosci 4: 492–498, 2001.[ISI][Medline]

24. Mueller SG, White JR, Schraw WP, Lam V, and Richmond A. Ligand-induced desensitization of the human CXC chemokine receptor-2 is modulated by multiple serine residues in the carboxyl-terminal domain of the receptor. J Biol Chem 272: 8207–8214, 1997.[Abstract/Free Full Text]

25. Naim M, Seifert R, Nürnberg B, Grünbaum L, and Schultz G. Some taste substances are direct activators of G-proteins. Biochem J 297: 451–454, 1994.[ISI][Medline]

26. Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJP, and Zuker CS. Mammalian sweet taste receptors. Cell 106: 381–390, 2001.[CrossRef][ISI][Medline]

27. Noble AC. Application of time-intensity procedures for the evaluation of taste and mouthfeel. Am J Enol Vitic 46: 128–133, 1995.[Abstract/Free Full Text]

28. Papermaster DS and Dreyer WJ. Rhodopsin content in the outer segment membranes of bovine and frog retinal rods. Biochemistry 13: 2438–2444, 1974.[CrossRef][ISI][Medline]

29. Parang K, Kohn JA, Saldanha SA, and Cole PA. Development of photo-crosslinking reagents for protein kinase-substrate interactions. FEBS Lett 520: 156–160, 2002.[CrossRef][ISI][Medline]

30. Perez CA, Huang L, Rong M, Kozak JA, Preuss AK, Zhang H, Max M, and Margolskee RF. A transient receptor potential channel expressed in taste receptor cells. Nat Neurosci 5: 1169–1176, 2002.[CrossRef][ISI][Medline]

31. Peri I, Mamrud-Brains H, Rodin S, Krizhanovsky V, Shai Y, Nir S, and Naim M. Rapid entry of bitter and sweet tastants into liposomes and taste cells: implications for signal transduction. Am J Physiol Cell Physiol 278: C17–C25, 2000.[Abstract/Free Full Text]

32. Pitcher JA, Freedman NJ, and Lefkowitz RJ. G protein-coupled receptor kinases. Annu Rev Biochem 67: 653–692, 1998.[CrossRef][ISI][Medline]

33. Premont RT, Koch WJ, Inglese J, and Lefkowitz RJ. Identification, purification, and characterization of GRK5, a member of the family of G protein-coupled receptor kinases. J Biol Chem 269: 6832–6841, 1994.[Abstract/Free Full Text]

34. Randak C, Auerswald EA, Assfalg-Machleidt I, Reenstra WW, and Machleidt W. Inhibition of ATPase, GTPase and adenylate kinase activities of the second nucleotide-binding fold of the cystic fibrosis transmembrane conductance regulator by genistein. Biochem J 340: 227–235, 1999.[CrossRef][ISI][Medline]

35. Rosenzweig S, Yan W, Dasso M, and Spielman AI. Possible novel mechanism for bitter taste mediated through cGMP. J Neurophysiol 81: 1661–1665, 1999.[Abstract/Free Full Text]

36. Sasaki Y, Suzuki M, and Hidaka H. The novel and specific Rho-kinase inhibitor (S)-(+)-2-methyl-1-[(4-methyl-5-isoquinoline)sulfonyl]-homopiperazine as a probing molecule for Rho-kinase-involved pathway. Pharmacol Ther 93: 225–232, 2002.[CrossRef][ISI][Medline]

37. Sitaramayya A and Liebman PA. Phosphorylation of rhodopsin and quenching of cyclic GMP phosphodiesterase activation by ATP at weak bleaches. J Biol Chem 258: 12106–12109, 1983.[Abstract/Free Full Text]

38. Spielman AI, Yan W, Krizhanovsky V, Rosenzweig S, Yamamoto T, and Naim M. Modulation of two second messengers in bitter taste transduction of agriculturally relevant compounds. In: Chemistry of Taste, Mechanisms, Behaviors, and Mimics, edited by Given P and Paredes D. Washington, DC: Am. Chem. Soc., 2002, p. 18–31.

39. Stadel JM, Nambi P, Shorr RGL, Sawyer DF, Caron MG, and Lefkowitz RJ. Catecholamine-induced desensitization of turkey erythrocyte adenylate cyclase is associated with phosphorylation of the {beta}-adrenergic receptor. Proc Natl Acad Sci USA 80: 3173–3177, 1983.[Abstract/Free Full Text]

40. Striem BJ, Naim M, and Lindemann B. Generation of cyclic AMP in taste buds of the rat circumvallate papilla in response to sucrose. Cell Physiol Biochem 1: 46–54, 1991.

41. Striem BJ, Pace U, Zehavi U, Naim M, and Lancet D. Sweet tastants stimulate adenylate cyclase coupled to GTP-binding protein in rat tongue membranes. Biochem J 260: 121–126, 1989.[ISI][Medline]

42. Varkevisser B and Kinnamon SC. Sweet taste transduction in hamster: role of protein kinases. J Neurophysiol 83: 2526–2532, 2000.[Abstract/Free Full Text]

43. Vera JC, Reyes AM, Velasquez FV, Rivas CI, Zhang RH, Strobel P, Slebe JC, Nunez-Alarcon J, and Golde DW. Direct inhibition of the hexose transporter GLUT1 by tyrosine kinase inhibitors. Biochemistry 40: 777–790, 2001.[CrossRef][ISI][Medline]

44. Weiss ER, Ducceschi MH, Horner TJ, Li A, Craft CM, and Osawa S. Species-specific differences in expression of G-protein-coupled receptor kinase (GRK) 7 and GRK1 in mammalian cone photoreceptor cells: implications for cone cell phototransduction. J Neurosci 21: 9175–9184, 2001.[Abstract/Free Full Text]

45. Wong GT, Gannon KS, and Margolskee RF. Transduction of bitter and sweet taste by gustducin. Nature 381: 796–800, 1996.[CrossRef][ISI][Medline]

46. Zhang Y, Hoon MA, Chandrashekar J, Mueller KL, Cook B, Wu D, Zuker CS, and Ryba NJP. Coding of sweet, bitter, and umami tastes: different receptor cells sharing similar signaling pathways. Cell 112: 293–301, 2003.[CrossRef][ISI][Medline]

47. Zoest AR, Wanwimolruk S, and Hung CT. Simple high-performance liquid chromatographic method for the analysis of quinine in human plasma without extraction. J Liq Chromatogr 13: 3481–3491, 1990.[ISI]

48. Zubare-Samuelov M, Peri I, Tal M, Tarshish M, Spielman AI, and Naim M. Some sweet and bitter tastants stimulate the inhibitory pathway of adenylyl cyclase via melatonin and {alpha}2-adrenergic receptors in Xenopus laevis melanophores. Am J Physiol Cell Physiol 285: C1255–C1262, 2003.[Abstract/Free Full Text]





This Article
Abstract
Full Text (PDF)
All Versions of this Article:
289/2/C483    most recent
00547.2004v1
Alert me when this article is cited
Alert me if a correction is posted
Citation Map
Services
Email this article to a friend
Similar articles in this journal
Similar articles in ISI Web of Science
Similar articles in PubMed
Alert me to new issues of the journal
Download to citation manager
Google Scholar
Articles by Zubare-Samuelov, M.
Articles by Naim, M.
Articles citing this Article
PubMed
PubMed Citation
Articles by Zubare-Samuelov, M.
Articles by Naim, M.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
Visit Other APS Journals Online
Copyright © 2005 by the American Physiological Society.