G{alpha}13-mediated transformation and apoptosis are permissively dependent on basal ERK activity

Vyacheslav A. Adarichev, Rita Vaiskunaite, Jiaxin Niu, Irina V. Balyasnikova, and Tatyana A. Voyno-Yasenetskaya

Departments of Pharmacology and Anesthesiology, University of Illinois, Chicago, Illinois 60612

Submitted 25 March 2003 ; accepted in final form 1 May 2003


    ABSTRACT
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
We previously reported that the {alpha}-subunit of heterotrimeric G13 protein induces either mitogenesis and neoplastic transformation or apoptosis in a cell-dependent manner. Here, we analyzed which signaling pathways are required for G{alpha}13-induced mitogenesis or apoptosis using a novel mutant of G{alpha}13. We have identified that in human cell line LoVo, the mutation encoding substitution of Arg260 to stop codon in mRNA of G{alpha}13 subunit produced a mutant protein (G{alpha}13-T) that lacks a COOH terminus and is endogenously expressed in LoVo cells as a polypeptide of 30 kDa. We found that G{alpha}13-T lost its ability to promote proliferation and transformation but retained its ability to induce apoptosis. We found that full-length G{alpha}13 could stimulate Elk1 transcription factor, whereas truncated G{alpha}13 lost this ability. G{alpha}13-dependent stimulation of Elk1 was inhibited by dominant-negative extracellular signal-regulated kinase (MEK) but not by dominant-negative MEKK1. Similarly, MEK inhibitor PD-98059 blocked G{alpha}13-induced Elk1 stimulation, whereas JNK inhibitor SB-203580 was ineffective. In Rat-1 fibroblasts, G{alpha}13-induced cell proliferation and foci formation were also inhibited by dominant-negative MEK and PD-98059 but not by dominant-negative MEKK1 and SB-203580. Whereas G{alpha}13-T alone did not induce transformation, coexpression with constitutively active MEK partially restored its ability to transform Rat-1 cells. Importantly, full-length but not G{alpha}13-T could stimulate Src kinase activity. Moreover, G{alpha}13-dependent stimulation of Elk1, cell proliferation, and foci formation were inhibited by tyrosine kinase inhibitor, genistein, or by dominant-negative Src kinase, suggesting the involvement of a Src-dependent pathway in the G{alpha}13-mediated cell proliferation and transformation. Importantly, truncated G{alpha}13 retained its ability to stimulate apoptosis signal-regulated kinase ASK1 and c-Jun terminal kinase, JNK. Interestingly, the apoptosis induced by G{alpha}13-T was inhibited by dominant-negative ASK1 or by SB-203580.

mitogen-activated kinase; effector mutant; Src kinase


G{alpha}13 BELONGS TO THE FAMILY of heterotrimeric guanine nucleotide-binding proteins that provide a link between membrane receptors and a set of signaling molecules inside the cell. The {alpha}-subunit of heterotrimeric G13 proteins regulates multiple cell-type dependent intracellular and cellular events. G{alpha}13 induces mitogenesis and neoplastic transformation in NIH3T3 and Rat-1 cells (21, 52, 57), apoptosis in Chinese hamster ovarian (CHO), and COS-7 cells (1, 4) and regulates the ERK (51, 52) and JNK pathways (49, 51). A possible candidate, which could generate such a cell-specific response, is the extracellular signal-regulated kinases (ERK) pathway. Activation of the ERK pathway is often associated with increased cell proliferation (29). Interestingly, a number of studies have shown that activation of ERK could protect against various apoptotic stimuli in different cell types (12, 56). Although G{alpha}13 by itself is not able to stimulate ERK, it could potentiate growth factor-induced ERK activation (52). Importantly, the role of the mitogen-activated signaling pathways in G{alpha}13-mediated mitogenesis or apoptosis was not analyzed.

Investigation of the signaling properties of mutant proteins in which effector-protein binding is defective has provided a useful approach to the functional study of Rho and Ras GTPases (3, 55). Moreover, effector mutants of G{alpha}s, G{alpha}q, and G{alpha}i proteins were used intensively to analyze the activation of specific signaling pathways (5, 30). So far, effector mutants of G{alpha}13 have not been identified. While screening multiple samples obtained from human tumors and tumor cell lines for the mutations in the {alpha}-chain of G13 protein, we found a mutation-encoding substitution of Arg260 to a stop codon in G{alpha}13 mRNA obtained from the human adenocarcinoma cell line LoVo. We found that G{alpha}13-T lost its ability to promote proliferation and transformation but retained its ability to induce apoptosis. Here, we analyzed which signaling pathways are required for G{alpha}13-induced mitogenesis or apoptosis using a novel mutant of G{alpha}13 as a specific tool to dissect G{alpha}13-mediated signals.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Materials. TaqI DNA polymerase, Moloney murine leukemia virus reverse transcriptase, DMEM, calf serum, penicillin G, streptomycin, amphotericine B, LipofectAMINE 2000, and Giemsa stain were from GIBCO BRL. PfuI was from Stratagene. DNA isolation kit was obtained from QIAGEN. G{alpha}13 and c-Src antibodies were obtained from Santa Cruz Biotechnology. 12CA5 antibody was from Babco. Dominant-negative c-Src (K295M/Y527F) was from S. Gutkind. Dominant-active MEK1 (S218E/S222D) and dominant-negative MEK1 (K79M) were from S. Mansour. Dominant-negative MEKK1 (K432A) was from G. Johnson. PD-98059 and SB-203580 were from CalBiochem.

Reverse transcription. Reverse transcription (RT) was performed with 1 µg of total RNA, 10 µlof5x first-strand buffer, 10 mM dithiothreitol, 0.5 mM of each dNTP, 10 ng/µl oligo-(dT)12-18, 200 U Moloney murine leukemia virus reverse transcriptase; final volume was adjusted with water to 20 µl. After incubation for 60 min at 37°C, the reaction was stopped by heating for 3 min at 100°C. Samples were kept at -20°C. To amplify the human G{alpha}13 gene, oligonucleotide primers were designed on the basis of published sequence (22).

Polymerase chain reaction. Polymerase chain reaction (PCR) was performed with 2 µl of RT mixture, 2.5 µl of 10x PCR buffer, 1.5 mM MgCl2, 0.2 mM of each dNTP, 1 µl {alpha}[32P]dCTP 5000 Ci/mmol, 1 µM of each primer, 2.5 U TaqI, and 0.16 U PfuI. PCR was performed using DNA thermal cycler and Perkin Elmer Cetus for 35 cycles as follows: 1 min at 94°C, 1 min at 61°C, and 2 min at 72°C, followed by 10 min of elongation at 72°C. PCR products were examined by 1.0% agarose gel electrophoresis after ethidium bromide staining.

Single-strand conformation polymorphism analysis. Single-strand conformation polymorphism analysis (SSCP) of PCR products was performed essentially as described (37a). To denature DNA before electrophoresis, 3 µl of samples were mixed with 17 µl of 95% formamide, 20 mM EDTA, 0.05% bromophenol blue, and 0.05% xylene cyanol and heated at 97°C for 5 min. Then, samples were immediately chilled on ice and loaded onto 8% polyacrylamide gel with N,N'-methylene bisacrylamide/acrylamide ratio 1:29, wt/wt. Electrophoresis was performed in 90 mM Tris-borate, pH 8.3, and 4 mM EDTA at 10 V/cm for 20 h at room temperature. After electrophoresis, the gels were transferred to Whatman paper and dried on a gel dryer (Bio-Rad). Transferred products were analyzed with PhosphorImager (Bio-Rad).

Site-directed mutagenesis. To introduce Q226L activation mutation into GTPase domain of truncated and full-version human G{alpha}13, we used a PCR-based site-directed mutagenesis kit provided by Stratagene (La Jolla, CA). Two synthetic oligonucleotide primers, direct and reverse, containing the mutation that converts CAG codon for Gln226 to CTT codon for Leu226 were extended during temperature cycling with Pfu polymerase, and methylated, nonmutated parental DNA template was with DpnI. DNA fragments that incorporated the desired mutation were transfected into HB101 competent cells. Plasmids containing the mutated gene were recovered by screening with restriction endonuclease, and the presence of the mutation was determined by sequencing. Hemagglutinin (HA)-epitope tagging of G{alpha}13-T was performed with double-stranded linker containing sequences for HA epitope (YPYDVPDYAS), followed by stop codon and flanking sites for restriction endonucleases to introduce the linker into plasmid. Primary structure was confirmed by sequencing.

Cell culture, transfection, foci formation, and colony-forming efficiency. Rat-1 murine fibroblasts were grown in high-glucose DMEM supplemented with 10% calf serum, 100 IU/ml penicillin G, 100 IU/ml streptomycin, and 0.25 mg/l amphofericine B until 80% confluence and passage every 2-4 days. For foci formation assay, Rat-1 cells were transfected on 60-mm dishes using 15 µl LipofectAMINE 2000 and 5 µg of DNA constructs and maintained in growth medium reduced to 5% heat-inactivated calf serum. The appearance of foci was scored in 2-3 weeks upon cells being fixed with paraformaldehyde and being stained with Giemsa stain.

Colony-survival assay. Colony-survival assay was performed by transfection of fibroblasts as described above. After transfection, cells were reseeded into 6-cm-diameter dishes. G418 selection (0.15 mg/ml) was applied the following day, and individual colonies (comprising >100 cells) were scored after a further 2 wk.

Luciferase assays. Elk-dependent gene expression was assayed by ELK-1 "PathDetect" trans-reporter system (Stratagene). Briefly, cells at 90% confluency grown on 24-well plates were transfected with the following plasmids (per well): 200 ng pFR-Luciferase (reporter plasmid), 12.5 ng pFA2-ELK1 (fusion trans-activator plasmid) (for ELK-dependent gene expression) with 50 ng pCMV-{beta}gal (transfection efficiency control plasmid), and 50 ng G{alpha}13Q229L, balanced with pCMV vector alone. The day before stimulation, cells were incubated in either 0.2 or 0% serum to achieve serum starvation. Activation of Elk1-dependent gene expression was similar in the cells exposed to either 0 or 0.2% serum. Cells were washed twice with PBS and lysed in protein extraction reagent, and the cleared lysates were assayed for luciferase and {beta}-galactosidase activity using the corresponding assay kits (Promega, Madison, WI). In order to account for differences in transfection efficiency, the Luciferase activity of each sample was normalized to {beta}-galactosidase activity and expressed as percent of the maximal response to G{alpha}-subunit stimulation.

JNK, ERK1, and ASK1 activity assays. JNK, ERK-1, and ASK1 activities were determined as described previously (4, 19). Briefly, HA-JNK or HA-ASK1 were transfected in the presence of various cDNA constructs as described in RESULTS. The kinase activity of HA-JNK using recombinant c-Jun and the kinase activity of HA-ERK or HA-ASK1 were measured or myelin basic protein (MBP) as a substrate, respectively. Aliquots of the whole cell lysates from the same experiment were subjected to immunoblotting analysis to confirm the appropriate expression of transfected proteins. In some experiments, phosphospecific ERK1/2 antibody (Cell Signaling Technology) was used.

Src kinase assay. Src family kinase assay was performed after the immunoprecipitation of endogenous c-Src (Src2, Santa Cruz) from NIH3T3 cells transfected with G{alpha}13Q229L. The immunocomplexes were collected with protein-A-agarose (GIBCO) and washed three times with lysis buffer. Kinase activity of c-Src was evaluated by its ability to phosphorylate the acid-denatured enolase (36). The radioactivity incorporated into enolase was measured by a BioImaging System (Bio-Rad).

Cell death assays. DNA fragmentation was determined as previously described using the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL method) (Apoptag Plus, Intergen) according to manufacturer's instructions (4). In this assay, residues of digoxigenin-nucleotide are catalytically added to the DNA by TdT. The incorporated digoxigenin is then recognized by fluorescein-coupled digoxigenin antibody. For apoptosis analysis, cells were cotransfected with cDNA-encoding {beta}-galactosidase. Twenty-four hours after transfection, cells were serum starved for an additional 16 h and then fixed with 2% paraformaldehyde for 15 min. Cells were permeabilized with 0.1% Triton X-100 for 10 min, and fragmented genomic DNA was stained as described (4). {beta}-Galactosidase was stained with polyclonal {beta}-galactosidase antibody, followed by a rabbit rhodamine-conjugated IgG. The percentage of {beta}-galactosidase-positive cells with fragmented nuclei was assessed by fluorescence microscopy. In each experiment, 300-400 cells were counted.

Caspase 3/7 activity assay. Caspase 3/7 activity was determined using Apo-ONE kit (Promega) according to the manufacturer's instructions. Briefly, Rat-1 cells seeded onto 24-well plates were transfected with indicated constructs for 24 h. Cells were washed with phosphate-buffered saline (PBS). Caspase 3/7 reagent (200 µl) was added to each well and incubated at 30°C for 4 h. The fluorescence was measured using spectrofluorimeter with an excitation wavelength of 485 nm and an emission wavelength of 530 nm using CytoFluor II. In some experiments, caspase activity was blocked by pretreatment with 10 µM Z-VAD-FMK caspase inhibitor for 16 h before experiment.


    RESULTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Human adenocarcinoma LoVo cell line contains a mutation in G{alpha}13 protein. Many proto-oncogenes encode proteins that transmit signals that regulate normal cell growth (32). The finding of GTPase-inhibiting mutations in genes for the {alpha}-subunits of Gs and Gi2 in certain endocrine tumors suggests that heterotrimeric G proteins might contribute to neoplasia (32). Our published data demonstrated that G{alpha}13 can behave as a potent, dominant-acting oncogene, suggesting that the G12/G13 family of G proteins may represent a novel class of oncogenes (52, 58). We used chromosomal homology maps constructed for mouse and human genomes as an approach to predict chromosomal localization of the G{alpha}13 gene. Comprehensive information about mouse-human chromosomal homology is available at the National Center for Biotechnology Information at http://www.ncbi.nlm.nih.gov. As the flanking genetic markers of G{alpha}13 and sequences linked with these genes in mouse and in human were known, we determined that G{alpha}13 is located on mouse chromosome 11 at 49-80 cM, which is homologous to the region 17q21-25 in humans. Thereafter, we used online the Mendelian Inheritance in Men database at http://www.ncbi.nlm.nih.gov/Omim/ that collects information on different human diseases and information on their genetic background to analyze genetic disorders that are associated with the chromosomal localization of G{alpha}13. We determined that the G{alpha}13-containing chromosomal region is involved in neuroblastoma, invasive pituitary tumors, cardiac myxoma, adrenocortical nodular dysplasia, and colorectal cancer (2, 9, 28, 31). Prompted by the knowledge of pathophysiology associated with chromosomal loci-containing G{alpha}13 gene, we analyzed the area flanking the GTPase region (149-269 aa) of the G{alpha}13 gene in 70 human tumors and tumor cell lines obtained from glioblastomas and colorectal cancers for possible mutations. To search for mutations in the GTPase region of G{alpha}13 cDNA, we used SSCP analysis. Total RNA isolated from cell lines and tissue samples of different origin served as a template in RT reaction, followed by PCR amplification of target sequences. Using SSCP analysis, we found that one of the cell lines obtained from LoVo human adenocarcinoma contained PCR fragments with altered mobility (Fig. 1A).



View larger version (40K):
[in this window]
[in a new window]
 
Fig. 1. Characterization of mutant G{alpha}13 subunit from LoVo cell line. A: detection of G{alpha}13 mutation using single-strand conformation polymorphism analysis (SSCP) of human tumor cell lines. The first strand of cDNA was synthesized by reverse transcriptase reaction using total RNA isolated from human tumors as described in MATERIALS AND METHODS. All tested samples (1, H-498, adenocarcinoma, colon; 2, HT-29 colorectal adenocarcinoma; 3, LoVo colorectal adeno-carcinoma; 4, Caco2, colorectal adenocarcinoma; 5, DLD1, colorectal adenocarcinoma) contained the wild type of both DNA strands, whereas LoVo adenocarcinoma also contained mutated cDNA. Experiment was repeated from the step of first-strand cDNA synthesis with the same results, confirming that the detected mutation was located in mRNA and not spontaneously gained during multiple polymerase reactions. B: Western blot detection of full-length and truncated G{alpha}13 in NIH3T3 cells transfected with wild-type and truncated G{alpha}13 and in LoVo adenocarcinoma cells expressing both endogenous wild-type and truncated G{alpha}13. NIH3T3 cells were transiently transfected with full-length and truncated G{alpha}13 for 48 h. Total cell lysates of LoVo cells and NIH3T3 cells were used for immunoblotting with G{alpha}13 antibody. Preabsorption of the G13-specific antibody with the immunizing peptide abolished the immunostaining of LoVo cells that express both wild-type and truncated G{alpha}13 (right).

 

Thereafter, the whole coding region of G{alpha}13 from human adenocarcinoma was cloned into mammalian expression vector by PCR approach using primers flanking the gene. DNA sequencing showed mutation at codon 260 (Arg260-> Stop) that resulted in the introduction of the premature stop codon. The predicted molecular mass of the truncated protein was 30 kDa. Immunoblotting analysis using antibody against the NH2 terminus (Santa Cruz Biotechnology) of G{alpha}13 showed that both full-length and truncated copies of G{alpha}13 were expressed endogenously in LoVo cells (Fig. 1B). To control the specificity of staining, G13-specific antibody was preabsorbed with immunizing peptide; the right panel depicts the absence of staining with G13-specific antibody that was preabsorbed with immunizing peptide (Fig. 1B). Similarly, in NIH3T3 cells transfected with truncated G{alpha}13, immunoblotting analysis of total cell lysates detected a single band with an apparent molecular mass of 30 kDa (Fig. 1B).

These data suggest that G{alpha}13-T forms a stable polypeptide that both exists as endogenous protein and can be transiently expressed in the cells.

G{alpha}13-mediated transformation of Rat-1 cells is permissively dependent on basal ERK activity. We have previously shown that mutationally activated G{alpha}13 induces neoplastic transformation of Rat-1 and NIH3T3 fibroblasts (52). As truncated G{alpha}13 protein was cloned from a LoVo cell line that derived from metastatic tumor node from a patient with adenocarcinoma of the colon, we have tested whether the G{alpha}13-T could induce neoplastic transformation. Focus formation assay in Rat-1 fibroblasts was used as a test for cell transformation. Rat-1 cells were transfected with corresponding constructs and maintained after transfection in the medium supplemented with 5% calf serum for 2 wk. Only constitutively activated mutant of human full-length G{alpha}13 (G{alpha}13Q226L) induced focus formation with transformation-specific morphology as a 0.1 to 1-mm area of multilayer cell growth (Fig. 2A). These data were consistent with previously published results obtained with constitutively activated mouse G{alpha}13 (52). Both wild-type G{alpha}13 and G{alpha}13-T did not induce focus formation in Rat-1 fibroblasts (Fig. 2A). To model the presence of two distinct G{alpha}13 polypeptides in LoVo cells, we determined whether equimolar amounts of wild-type G{alpha}13 and G{alpha}13-T could induce focus formation in Rat-1 fibroblasts. G{alpha}13-T did not enhance the transformation by wild-type G{alpha}13 (Fig. 2A). Finally, G{alpha}13Q226L-T (Q226L mutation was introduced in a GTPase region of G{alpha}13-T) also did not induce focus formation in Rat-1 fibroblasts (Figure 2A). Similar data were obtained using NIH3T3 fibroblasts (data not shown). Thus G{alpha}13-T failed to induce focus formation in Rat-1 fibroblasts and probably cannot be considered as a reason for the neoplastic phenotype of LoVo adenocarcinoma.



View larger version (32K):
[in this window]
[in a new window]
 
Fig. 2. Truncated G{alpha}13 failed to induce focus formation in Rat-1 fibroblasts. Rat-1 fibroblasts were transfected with different G{alpha}13 constructs using LipofectAMINE 2000 reagent and were grown in medium supplemented with 5% calf serum. Focus formation was scored after 15 days. Cells were fixed with formaldehyde and stained with Giemsa, and regions of multilayer cell growth with distinct morphology were examined under the microscope. A: representative focus formation assay. The data shown are representative of independent experiments, with each determination representing the average number of foci from 3 dishes. The error bars indicate SD. B: clonogenic analyses showed that transfection of the G{alpha}13- and G{alpha}13Q22L-T expression plasmids caused significant inhibition of cell growth. Rat-1 cells were transfected with equimolar amounts of pcDNA.3.1 vector, G{alpha}13, or G{alpha}13 Q226L-T and selected for 14 days with G418, and then appearance of drug-resistant colonies was quantitated. Data are representative of 2 independent experiments and are the average of 4 plates.

 

Interestingly, both G{alpha}13-T and G{alpha}13Q226L-T inhibited G{alpha}13Q226L-induced foci formation by 65% (data not shown). Because transformation inhibition seen with G{alpha}13-T may not be specific but instead may be a consequence of nonspecific growth inhibition, we determined whether G{alpha}13-T could cause inhibition of cell growth by employing a colony survival assay by drug selection of transfected Rat-1 cells. As shown in Fig. 2B, both G{alpha}13-T and G{alpha}13Q226L-T significantly inhibited the number of surviving colonies. We also noted that many of the colonies generated from G{alpha}13-T or G{alpha}13Q226L-T transfected Rat-1 cells grew poorly and had a propensity to detach from the culture dish, with widespread cell death, suggesting that these two mutants may induce apoptosis in Rat-1 cells.

We have previously shown that in Rat-1 cell lines stably transfected with G{alpha}13, EGF-stimulated ERK1 activity was potentiated (52); however, significance of this phenomenon is not understood. Involvement of the ERK signaling pathway in cell transformation induced by a variety of oncogenes is now well documented (for review, see Ref. 29); however, the involvement of this pathway in G{alpha}13-dependent transformation has not been investigated. Interestingly, we could not detect G{alpha}13-dependent activation of ERK1 using kinase assay (52) or using or phospho-ERK1 antibody (data not shown).

Signaling pathways such as the MAP kinases, ERK1 and 2, p38, and JNK induce phosphorylation of the transcription factor Elk-1, resulting in its activation (46). Therefore, Elk-1-induced expression of the reporter gene was used as a readout of G{alpha}13-induced gene transcription. In transient transfection, full-length G{alpha}13Q226L stimulated Elk-1 by fivefold (Fig. 3). The constitutively active mutant form of MEK1 (S218E/S222D) (33) stimulated Elk-1 by 50-fold as compared with vector-transfected cells (Fig. 3). To determine whether G{alpha}13-induced activation of Elk-1 occurs via simulation of the MEK-ERK signaling pathway, we measured Elk-1 activation by G{alpha}13 in the presence of the dominant-negative (K79M) form of MEK1 (33) or the dominant-negative (K432A) form of MEKK1 (51). In some experiments, cells were pretreated with the pharmacological inhibitor of MEK PD-98059 or the pharmacological inhibitor of p38MAPK and JNK SB-202190. SB-202190 was initially described as an inhibitor of p38 MAPK (27); however, it was recently shown that at higher concentration, it also inhibits JNK activity (20). Because our unpublished data confirmed that SB-203580 inhibited p38 at 4 µM and inhibited JNK at 40 µM, we used SB-203580 at 40 µM, a concentration that was capable of inhibiting both pathways.



View larger version (20K):
[in this window]
[in a new window]
 
Fig. 3. MEK-ERK pathway is involved in G{alpha}13-dependent proliferation and focus formation. A: activation of Elk-1 transcription in cells transiently transfected with G{alpha}13 and its mutants. Rat-1 cells were transfected with 150 ng pFR-Luciferase (reporter plasmid), 7.5 ng pA2-Elk1 (fusion trans-activator plasmid), 92.5 ng pcDNA LacZ (transfection efficiency control plasmid), 100 ng of different G{alpha}13 mutants, 100 ng of activated MEK1 as indicated, and 200 ng of dominant-negative MEK or dominant-negative MEKK1 balanced with empty vector, as described under MATERIAL AND METHODS. Cells were serum starved overnight. In some cases, during this period cells were treated with 17 µM PD-98059 or 40 µM SB-203580. Luciferase activity was measured in the cell extracts, normalized to {beta}-galactosidase activity, and expressed as fold stimulation from the basal level. Data represent means ± SE from 1 of at least 3 experiments performed in triplicate. B: Rat-1 cells stably transfected with G{alpha}13Q226L were grown in the presence of 17 µM PD-98059. Data shown are from 3 experiments performed in triplicate. Error bars are SD. C, left: Rat-1 cells were transfected with 1 µg of G{alpha}13QL in the absence or presence of 2 µg of dominant-negative MEK, dominant-negative MEKK1, and 17 µM PD-98059 or 40 µM SB-203580 as indicated. Empty pcDNA3 vector was added to maintain the DNA concentration constant in all transfections. Growth medium supplemented with indicated pharmacological inhibitors was changed every other day. Foci of transformed cells were counted at 15 days. Data represent means ± SE from 1 of at least 3 experiments performed in triplicate. Right: Rat-1 cells were transfected with 1 µg of G{alpha}13Q22L-T in the absence or presence of 2 µg of dominant-active MEK(S218E/S222D), MEK-SS as indicated. Empty pcDNA3 vector was added to maintain the DNA concentration constant in all transfections. Foci of transformed cells were counted at 15 days.

 

Our data showed that Elk-1 activation by G{alpha}13 was suppressed by dominant-negative MEK1 but not by dominant-negative MEKK1 (Fig. 3). In addition, PD-98059 but not SB-202190 inhibited G{alpha}13-induced Elk1 activation, suggesting that G{alpha}13 can activate the ERK signaling pathway. Interestingly, G{alpha}13wt, G{alpha}13-T, and G{alpha}13Q226L-T did not stimulate Elk-1 activation (Fig. 3).

We then determined whether the MEK-ERK pathway is involved in the G{alpha}13-dependent regulation of cell growth and transformation. To characterize growth rate, Rat-1 cells stably transfected with constitutively activated G{alpha}13Q226L were plated in medium containing 5% calf serum, and cells were counted every day. In the presence of 5% calf serum, G{alpha}13Q226L-expressing cells proliferated faster than vector only-expressing cells and reached a confluent state faster, and maximal cell density was higher as compared to cells transfected with vector alone (Fig. 3B). The overexpression of G{alpha}13Q226L was about twofold when compared with endogenously expressed G{alpha}13 (Fig. 3B, inset). Proliferation of the Rat-1 cell line expressing wild-type G{alpha}13 was similar to that of the vector only-expressing cell line (data not shown). Specific MEK inhibitor PD-98059 inhibited G{alpha}13Q226L-induced cell growth (Fig. 3B), whereas inhibitor of p38MAPK and JNK SB-202190 did not affect G{alpha}13-induced proliferation (data not shown). Similarly, PD-98059 and dominant-negative MEK inhibited G{alpha}13Q226L-induced focus formation (Fig. 3C), whereas SB-202190 and dominant-negative MEKK1 did not affect the foci-forming activity of G{alpha}13Q226L. Thus, although G{alpha}13 did not activate ERK, the basal ERK activity may be essential for transformation, whereas p38MAPK and JNK pathways did not seem to be involved in G{alpha}13-induced transformation.

Finally, we determined whether activation of ERK pathway could salvage G{alpha}13-T deficiency to induce neoplastic transformation in Rat-1 cells. For these experiments, we cotransfected Rat-1 cells with a constitutively active mutant form of MEK1 (S218E/S222D) (33). Increased activity of ERK in the cells transfected with constitutively active MEK1 was confirmed by Luciferase-based determination of ERK activity (data not shown). Constitutively active MEK1 alone induced a small but significant increase of foci formation (Fig. 3C) that was consistent with previously published data (33). Cotransfection of MEK1 (S218E/S222D) with G{alpha}13Q226L-T resulted in a synergistic increase of foci formation (Fig. 3C). These results indicate that MEK-ERK pathway is required for G{alpha}13-induced transformation of Rat-1 cells. These results also indicate that failure to induce transformation by truncated G{alpha}13 may be explained by its failure to activate the MEK-ERK pathway.

To further examine the mechanism of G{alpha}13-induced cell transformation, we investigated the possible role of Src, whose involvement in the regulation of cell proliferation and transformation is well established. To determine the activity of the endogenous c-Src kinase, we used Rat-1 cells transfected with G{alpha}13Q226L (Fig. 4A). Equal quantities of endogenous Src family kinases were immunoprecipitated from Rat-1 cells, and Src kinase activity was estimated by phosphorylation of acid-denatured enolase. G{alpha}13Q226L increased the activity of endogenous Src kinase by ~5.4-fold. Pretreatment of the cells with tyrosine kinase inhibitor genistein inhibited G{alpha}-induced activation of Src kinase (Fig. 4A). Importantly, truncated G{alpha}13 did not induce Src phosphorylation (Fig. 4A).



View larger version (17K):
[in this window]
[in a new window]
 
Fig. 4. Src-kinase is involved in G{alpha}13-dependent proliferation and focus formation. A: activation of Src kinase in Rat-1 cells expressing G{alpha}13Q226L. Endogenous Src was immunoprecipitated from serum-starved Rat-1 cells transfected with 1 mg of G{alpha}13Q226L, G{alpha}13-T, or G{alpha}13Q226L-T for 48 h. Cells were treated with 50 µM genistein for 6 h before the experiment. Kinase activity of Src was determined as described in MATERIALS AND METHODS. Upper band shows phosphorylation of acid-denatured enolase. Lower panel shows that an equivalent amount of Src was immunoprecipitated from the cells. B: activation of ELK by G{alpha}13Q226L inhibited by dominant-negative Src. Rat-1 cells were transfected with pFR-Luciferase (reporter plasmid), pA2-Elk1 (fusion trans-activator plasmid), pcDNA LacZ (transfection efficiency control plasmid) G{alpha}13Q226L, and dominant-negative Src (c-Src K295M/Y527F, Src-DN). Cells were serum starved overnight and, as indicated, incubated with 50 µM genistein for 6 h before the experiment. Luciferase activity was measured in the cell extracts, normalized to {beta}-galactosidase activity, and expressed as a fold stimulation from the basal level. Data represent means ± SE from 1 out of at least 3 experiments performed in triplicate. C: inhibition of G{alpha}13-dependent proliferation by Src inhibition. Rat-1 cells stably transfected with G{alpha}13Q226L were grown in the presence of 50 µM genistein. Data shown are from 3 experiments performed in triplicate. Error bars are SD. D: inhibition of G{alpha}13-dependent focus formation by dominant-negative Src. Rat-1 cells were transfected with 1 µg of G{alpha}13Q226L and 1 µg of dominant-negative Src. Empty vector was added to maintain the DNA concentration constant in all transfections. Foci of transformed cells were counted at 15 days.

 

Finally, we determined how inhibition of Src affected G{alpha}13-induced mitogenic signaling. Data showed that both dominant-negative c-Src (c-Src K295M/Y527F, SrcDN) and genistein completely inhibited G{alpha}13 Q226L-dependent activation of Elk-1 (Fig. 4B). Similarly, genistein inhibited G{alpha}13-dependent cell growth (Fig. 4C), whereas SrcDN inhibited G{alpha}13Q226L-induced focus formation (Fig. 4D). To rule out nonspecific growth inhibition, we determined whether SrcDN could cause inhibition of cell growth by employing a colony-survival assay by drug selection of transfected Rat-1 cells. No difference was observed between the vector and dominant-negative Src with respect to the number of colonies (data not shown). Thus the inhibition of focus-forming activity that we detected could not be the result of a nonspecific growth inhibition by mutant protein. Therefore, the data suggest that activation of Src pathway is required for G{alpha}13 subunit-dependent transformation.

Mitogen-activated signaling is required for G{alpha}13-induced apoptosis in Rat-1 fibroblasts. As we have shown previously that G{alpha}13 activates the JNK pathway (51), we have now tested whether G{alpha}13-T and G{alpha}13Q226L-T could activate the JNK pathway. Consistent with previously published data, G{alpha}13Q226L-induced phosphorylation of c-Jun by JNK was inhibited by dominant-negative MEKK1 and SB-202190 (51), whereas dominant-negative MEK1 and PD-98059 did not affect JNK activity (Fig 5A). Interestingly, two truncated mutants of G{alpha}13 also induced JNK activation that was inhibited by both dominant-negative MEKK1 and SB-202190 (Fig. 5B).



View larger version (38K):
[in this window]
[in a new window]
 
Fig. 5. G{alpha}13-induced activation of JNK. Rat-1 cells were seeded onto 60-mm dishes and transfected with 1 µg of indicated constructs of G{alpha}13 and 2 µg of dominant-negative MEK or dominant-negative MEKK1 and 0.5 µg of HA-JNK. cDNA amount was balanced with pcDNA3 vector. In some cases, cells were treated with 17 µM PD-98059 or 40 µM SB-203580 for 1 h. Protein expression and kinase activity using anti-hemagglutinin (HA) to immunoprecipitate JNK-HA and c-Jun as a substrate were determined as described (4).

 

We have recently described that although G{alpha}13 induces neoplastic transformation in Rat-1 and NIH3T3 cells (52), it induces apoptosis in other cell lines such as COS-7 and CHO cells (1, 4); the reason for these cell-specific effects remains unknown. Because two truncated mutants of G{alpha}13 had decreased colony-forming efficiency (Fig. 2B), we tested whether they could induce apoptosis in Rat-1 cells using in situ detection of DNA fragmentation using the TUNEL method (4). Different {alpha}-subunit constructs were transiently transfected (at a ratio of 1 µg of cDNA:106 cells), and 24 h after transfection, cell death was estimated measuring the number of cells with fragmented DNA labeled with TdT (Apoptag Plus kit) (Fig. 6A) in cells expressing G{alpha}13 constructs. Under these conditions, 2 ± 1.1% of vector-transfected cells underwent apoptosis (Fig. 6A). Neither wild-type G{alpha}13 nor activated G{alpha}13Q226L mutant induced apoptosis in these cells; these data were consistent with our previous observations (4, 52). However, both G{alpha}13-T and G{alpha}13Q226L-T induced apoptosis in 46 ± 7% of transfected cells (Fig. 6A). Dilution of G{alpha}13-T or G{alpha}13Q226L-T cDNA 10-fold with the vector cDNA during transfection resulted in a reduced percentage of apoptotic cells (13 ± 5.5%, data not shown), suggesting that G{alpha}13-T-induced apoptosis is a function of G{alpha}13-T expression levels.



View larger version (34K):
[in this window]
[in a new window]
 
Fig. 6. Truncated G{alpha}13-induced apoptosis in Rat-1 cells. A: Rat-1 cells were transfected with {beta}-galactosidase and indicated G{alpha}13 constructs (1:10 cDNA ratio) (A and B), G{alpha}13Q226L, and G{alpha}13Q226L-T (C and D). Cells with fragmented nuclei (B and D) were counted among the cells expressing {beta}-galactosidase (A and C) as described in MATERIALS AND METHODS. Cells with fragmented nuclei were counted among the cells expressing {beta}-galactosidase. Graphic representation of apoptosis induced by G{alpha}13 constructs of 3 independent experiments. In each experiment, 300-400 cells were counted. Percentage values were expressed as average values ± SE. B: constitutively active MEK and SB-203580 inhibited apoptosis induced by a truncated G{alpha}13. Percentage of apoptosis in transfected cells was determined as described in MATERIALS AND METHODS. Cells were transfected with truncated G{alpha}13 in the presence of MEK-SS as indicated. Alternatively, cells were treated with 40 µM SB-203580 for 24 h. The data represent means ± SD from 3 independent experiments performed in duplicate. Two-hundred {beta}-galactosidase-positive cells were counted on each coverslip. C: caspase 3/7 activity was determined in Rat-1 cells transfected with different amount of G{alpha}13-T as described in MATERIALS AND METHODS.

 

Because mitogenic stimuli through the ERK cascade have been shown to have inhibitory effect on the induction of apoptosis (56), and our data showed that G{alpha}13-T could not stimulate ERK-dependent Elk-1 activation, we determined whether ERK-mediated signaling could be involved in protection of Rat-1 cells from G{alpha}13-induced apoptosis. Therefore, we determined whether activation of the ERK pathway could inhibit G{alpha}13-T-induced apoptosis. For these experiments, we cotransfected Rat-1 cells with a constitutively active mutant form of MEK1 (S218E/S222D). Under these conditions, Rat-1 cells were efficiently protected against G{alpha}13-T-induced apoptosis (Fig. 6A). Similarly, inhibition of JNK and p38 kinase by SB-202190 effectively protected Rat-1 cells from apoptosis (Fig. 6A).

Because caspase 3 and 7 are activated during the earliest phases of apoptotic induction (41), we determined the activity of caspase-3/7 using rhodamineconjugated caspase 3/7 substrate, bis-(N-CBZ-L-aspartyl-L-glutamyl-L-valyl-L-aspartic acid amide, Z-DEVDR110) according to the manufacturer's instruction (Apo-ONE Promega). Consistently, only the truncated construct of G{alpha}13 induced caspase 3/7 activation (Fig. 6B). Pretreatment of the cells with cell-permeable caspase inhibitor Z-VAD-FMK blocked G{alpha}13-T-induced caspase activation.

We have previously shown that G{alpha}13-induced apoptosis in COS-7 cells is mediated by apoptosis signal-regulating kinase (ASK1) (4). To examine whether truncated G{alpha}13 could affect the same signaling pathway, ASK-1 activity was determined in the Rat-1 cells expressing full-length and truncated G{alpha}13 and ASK1 (Fig. 7A). Consistent with previously published data, mutationally activated G{alpha}13 induced ASK1 activity (4). In addition, truncated G{alpha}13 also stimulated ASK1 (Fig. 7A). Finally, we determined whether dominant-negative ASK1 could inhibit apoptosis induced by truncated G{alpha}13 in Rat-1 cells. Data showed that cotransfection of truncated G13 with dominant-negative ASK1 resulted in significant inhibition of apoptosis (Fig. 7B). These results indicate that apoptosis induced by truncated G{alpha}13 can be mediated via ASK1. Besides, both full-length and truncated G{alpha}13 stimulated ASK1; however, only truncated G{alpha}13 induced apoptosis in Rat-1 cells, suggesting that full-length G{alpha}13 may stimulate additional signaling pathways that protect cells from apoptosis.



View larger version (26K):
[in this window]
[in a new window]
 
Fig. 7. Truncated G{alpha}13 induce apoptosis via apoptosis signal-regulating kinase 1 (ASK1) activation. A: truncated G{alpha}13 induced activation of ASK1. The indicated constructs of G{alpha}13 were expressed in Rat-1 cells, together with HA-tagged ASK1 (4). Their expression did not affect the expression level of ASK1 because it was determined by Western blotting. ASK1 was immunoprecipitated with HA antibody and assayed for kinase activity using myelin basic protein as a substrate. The experiment was performed 3 times with similar results. B: dominant-negative ASK1 inhibited G{alpha}13-T-induced apoptosis. Rat-1 cells were transfected with G{alpha}13-T or G{alpha}13Q226L-T and {beta}-galactosidase in the absence or presence of ASK1 (K709R). Cells with fragmented nuclei were counted among the cells expressing {beta}-galactosidase as described in MATERIALS AND METHODS. Experiment was performed 3 times in duplicate. Two-hundred {beta}-galactosidase-positive cells were counted on each coverslip. The values represent means + SD.

 

Figure 8 shows the model 3-D structures of truncated G{alpha}13 (Fig. 8A) and full-length protein (Fig. 8B). The very first 19 amino acids from G{alpha}13 NH2 terminus are unique and not homologous to any known sequence. Therefore, they were excluded from our 3-D model. The region of deletion, amino acids 260-377 of G{alpha}13, is shown in purple (Fig. 8B). Major putative sites for interaction with G{alpha}{beta}{gamma}-subunits, sites for lipid modification, regulators of G protein signaling (RGS)-interacting domains (shown as three blue loops), and G1-G3 sites, ensuring chelating of guanine nucleotide, are present in G{alpha}13-T. G1, G2, and G3 domains could theoretically coordinate {alpha}, {beta}, and phosphates, as well as provide coordinated molecule of water and Mg2+ in the catalytic site of the protein for GTP/GDP turnover. However, two extra sites responsible for the interaction of {alpha}-subunit with guanine nucleotide are lost. The G4 region is responsible for coordination of the guanine ring and, by this means, secures GTP/GDP interaction. The function of G5 region that is conserved among G proteins is unknown.



View larger version (43K):
[in this window]
[in a new window]
 
Fig. 8. Model ribbon diagrams of G{alpha}13. A: putative ribbon diagram for truncated G{alpha}13. The 3-D structure for the G{alpha}13 was generated with Swiss-Pdb Viewer v3.5 software using crystal structure of G{alpha}i1 in complex with GDP, AlF-4, and regulators of G protein signaling (RGS4) as a matrix. Position of deletion is marked as "260." The first 19 amino acids are not shown because there is no homology found for them in GenBank. Guanine nucleotide is in the middle of protein structure, and it can be seen that the guanine ring is not chelated by truncated polypeptide.

 

Thus, from the analysis of 3-D crystal models for G{alpha}13, we can predict that guanine nucleotide binding and GTP hydrolysis will be seriously impaired in G{alpha}13-T mutant. Interestingly, mitogenic response that required a GTP-bound form of G{alpha}13 (at least, in Rat-1 cells Ref. 52) was absent in G{alpha}13-T. However, G{alpha}13-T has maintained its ability to induce apoptosis, a response that did not seem to depend on the guanine nucleotide-binding state of the mutant protein. We conclude that this mutant protein will be a useful tool for identifying G{alpha}13-interacting protein(s) that define the ability of G{alpha}13 to induce mitogenesis or apoptosis. Yeast two-hybrid studies and DNA array displays using truncated mutant of G{alpha}13 are currently in progress.


    DISCUSSION
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Mutations in G subunits that lead to abnormal signaling are known to cause a large number of diseases (18). Increasing evidence is accumulating to implicate heterotrimeric G proteins in the growth regulation of various cancers. Many diverse ligands acting on G protein-coupled receptors are well known to cause tumor cell proliferation (25, 26, 32). Expression of mutationally activated G{alpha}i2 in Rat-1 fibroblasts caused neoplastic transformation and tumorigenicity (38). Constitutionally activating mutations in the genes for G{alpha}s (R201C and Q227L) and G{alpha}i2 (R179C and Q205L) have been found in specific subsets of human tumors (25, 26, 32). Others and we have recently reported that the mutationally activated {alpha}-subunits of G12 and G13 cause neoplastic transformation and induce proliferation when ectopically expressed in NIH3T3 fibroblasts and Rat-1 fibroblasts and induce tumor formation in nude mice (52, 57, 58). Interestingly, overexpression of wild-type G{alpha}12 in NIH3T3 cells also induced cell transformation (8), although transformation induced by mutationally activated G{alpha}12 was significantly greater (52). Because current information does not point to specific tissues in which G{alpha}12 and G{alpha}13 can mediate a proliferative stimulus, we have performed a broad-based search for mutant G{alpha}12 and G{alpha}13 in various human tumors and tumor-derived cell lines.

G{alpha}12 and G{alpha}13 are localized in chromosomal regions showing association with colon, pancreas, and brain tumors. In this study, extensive screening of tumors from different tissues did not reveal mutations that could increase mitogenic potential of G{alpha}13. Comparing this result with the level of polymorphism for G{alpha}s, it is necessary to underscore that all activated mutations of G{alpha}s were restricted to three types of tumor: pituitary adrenocorticotropic growth hormone-secreting adenomas, thyroid tumors, and adrenocortical adenomas. The probability of finding a mutated form of G{alpha}s in these tumors was from 10 to 40% (32). Mutated G{alpha}i2 is also tissue specific and can be found mainly in ovarian sex cord stromal tumors and adrenal cortical tumor with a probability of about 30% (32). An extensive search for activated mutations in G{alpha}11, G{alpha}14, and G{alpha}q did not detect mutations of GTPase domain in neoplasias from different origins (14). Because G{alpha}12 was identified as a putative oncogene in soft tissue sarcomas (8), we cannot exclude the possibility that both G{alpha}12 and G{alpha}13 could be putative oncogenes in tissues that we did not test in this study.

Here, we report the identification and characterization of the novel mutant of G{alpha}13 protein. The mutation encoding the substitution of Arg260 to stop the codon in mRNA of the G{alpha}13 subunit produced a mutant protein (G{alpha}13-T) that lacks a COOH terminus and is endogenously expressed in LoVo cells as a polypeptide of 30 kDa. The LoVo cell line was initiated in 1971 from a metastatic tumor nodule in the left ventricular region of a patient with adenocarcinoma of the colon (15, 16). The established cell line retains many features associated with malignant cells; for example, it secretes carcinoembryonic antigen and induces tumors in nude mice with 100% frequency (15, 16). Although G{alpha}13-T failed to induce focus formation in Rat-1 fibroblasts and probably cannot be considered as a reason for the neoplastic phenotype of LoVo adenocarcinoma, it may contribute to the sensitivity of this cell line to apoptosis-inducing agents (42).

Three-dimensional model structure of G{alpha}13. The crystal structure of G{alpha}13 has not been solved yet. However, 3-D structures for a number of G proteins {alpha}-subunits in a complex with {beta}{gamma}-subunits, different guanine nucleotide analogs, and RGS were successfully solved (11, 24, 44, 45). Alignment of amino acid sequence for the protein against G{alpha}s, G{alpha}t, G{alpha}i1, p21ras, and other homologous proteins reveals a high level of identity and similarity (24). Therefore, using Swiss-Pdb Viewer v. 3.5 and Geneva Biomedical Research Center homology modeling server "Swiss-Model" (39), we threaded a G{alpha}13 protein primary sequence onto a 3-D template of homologous protein (Fig. 6). Human G{alpha}13 contains several conservative amino acid regions responsible for chelating and hydrolysis of guanine nucleotide (6, 22). These regions are G1, G2, G3, G4, and G5 according to Bourne nomenclature (6).

Studies of crystal structures for p21ras,G{alpha}t, and G{alpha}i1 proteins complexed with guanine nucleotide analogs showed that the first region (A or G1) is responsible for chelating {alpha}-, {beta}-, and {gamma}-phosphates, the second (C or G2) interacts with {gamma}-phosphate, the third (G or G3) is a catalytic domain and interacts with {gamma}-phosphate, and the fourth (I or G4) is responsible for interaction with the guanine ring (6).

RGS proteins, which accelerate the rate of GTP hydrolysis, can interact with the surface of protein globule that is formed by amino acids from switch I, II, and III the Ras-like domains and do not make significant contact with the {alpha}-helical domain of G{alpha}, as was shown for RGS4 bound to AlF- 4 -activated G{alpha}i1. RGS4 could enhance catalysis by binding a molecule of water in the active site through the Asn128 residue that is in close vicinity with Gln204 and Ser206 of the G{alpha}i1 switch II domain (11, 45). Guanine nucleotide-releasing proteins (GNRP) interact with the very C-end of G{alpha} protein (6).

Figure 6 shows the model 3-D structures of truncated G{alpha}13 (Fig. 8A) and full-length protein (Fig. 8B). The very first 19 amino acids from the G{alpha}13 NH2 terminus are unique and not homologous to any known sequence. Therefore, they were excluded from our 3-D model. The region of deletion, amino acids 260-377 of G{alpha}13, is shown in purple (Fig. 8B). Major putative sites for interaction with G{beta}{gamma}-subunits, sites for lipid modification, RGS interacting domains (shown as a three blue loops), and G1-G3 sites ensuring chelating of guanine nucleotide are present in G{alpha}13-T. G1, G2, and G3 domains could theoretically coordinate {alpha}-, {beta}-, and {gamma}-phosphates, as well as provide a coordinated molecule of water and Mg2+ in the catalytic site of the protein for GTP/GDP turnover. However, two extra sites responsible for the interaction of the {alpha}-subunit with the guanine nucleotide are lost. The G4 region is responsible for coordination of the guanine ring and, by this means, secures GTP/GDP interaction. The function of the G5 region that is conserved among G proteins is unknown.

Thus, from the analysis of 3-D crystal models for G{alpha}13, we can predict that guanine nucleotide binding and GTP hydrolysis will be seriously impaired in G{alpha}13-T mutant. Interestingly, the mitogenic response that required a GTP-bound form of G{alpha}13 (at least, in Rat-1 cells, Ref. 52), was absent in G{alpha}13-T. However, G{alpha}13-T has maintained its ability to induce apoptosis, a response that did not seem to depend on the guanine nucleotide-binding state of the mutant protein. We conclude that this mutant protein will be a useful tool for identifying G{alpha}13-interacting protein(s) that define the ability of G{alpha}13 to induce mitogenesis or apoptosis. Yeast two-hybrid studies and DNA array displays using the truncated mutant of G{alpha}13 are currently in progress.

Regulation of mitogenesis and apoptosis by MAPK pathways. With the recognition that G{alpha}13 protein is functionally coupled to both mitogenic and apoptotic responses, we hypothesized that G{alpha}13 regulates multiple signaling pathways that produce bifurcating signals leading to mitogenic or apoptotic response. The variable effects of G{alpha}13 on mitogenic and apoptotic responses could reflect the use of these multiple pathways under different circumstances and in different cell types.

MAPK are a family of Ser/Thr kinases involved in the regulation of a wide range of cellular responses, including cell proliferation and survival (43). To date, several distinct MAPK have been identified that act independently on signaling pathways. These include ERK, JNK, and p38MAPK; these kinases represent the terminal stages of growth/survival factor or death receptors (54). The MAPK pathways act as key regulators of mitogenic and apoptotic pathways. Deregulation of ERK pathway leads to cell proliferation and neoplastic transformation (23, 33). The role of JNK pathway in mitogenesis and neoplastic transformation is poorly understood; however, several reports suggest that the JNK pathway also participates in neoplastic transformation (40).

The role of G{alpha}13 in ERK activation is complex and seems to be cell dependent. Several studies have not detected ERK activation or even detected ERK inhibition by G{alpha}12 and G{alpha}13 (51), whereas other studies have shown these proteins can either activate ERK (13, 35) or enhance the extent and persistence of the agonist-dependent ERK activation (52). Our data showed that basal activity of ERK (but not JNK pathway) is involved in G{alpha}13-mediated proliferation and transformation. Although we could not detect G{alpha}13-induced ERK activation using kinase assay and phospho-ERK antibody, pharmacological and genetic inhibition of MEK-ERK pathway inhibited G{alpha}13-induced proliferation and transformation. Similar inhibition of JNK-p38 MAPK pathway did not have any effect on G{alpha}13-induced proliferation and transformation. Moreover, constitutively active MEK partially salvaged the ability of G{alpha}13-T to transform Rat-1 cells. It is not clear how G{alpha}13 could modulate basal level of ERK activity. One possibility is that G{alpha}13 could affect the expression level of some of the components of ERK signaling pathway, thus modulating the basal ERK activity. This possibility is currently under investigation.

Another interesting possibility is in a recently described interaction between G{alpha}13 and E-cadherin (34). E-cadherin is a long recognized tumor suppressor (50), and inhibition of the ERK pathway induces synthesis of E-cadherin (10). It is tempting to speculate that the expression of G{alpha}13-T may result in an increased expression of E-cadherin due to inhibition of the ERK pathway, which may lead to the enhanced tumor suppressor function of E-cadherin. This hypothesis is currently under investigation.

Our data suggest that the Src pathway mediates G{alpha}13-dependent gene expression, cell proliferation, and transformation. Thus G{alpha}13-induced cell proliferation and foci formation were abolished by either tyrosine kinase inhibitor or by the dominant-negative mutant of Src. In addition, G{alpha}13Q226L could stimulate Src activity.

The MAPK pathways, in particular the JNK pathway, participate in the induction of apoptosis (56). The activation of JNK and p38 MAPK is generally associated with the promotion of apoptosis, whereas ERK activity inhibits apoptosis. Thus, in PC-12 cell during apoptosis induced by growth factor withdrawal, JNK and p38 MAPK are activated, whereas ERK is inhibited (56). In contrast, the activation of ERK has been shown to inhibit apoptosis induced by hypoxia (7), growth factor withdrawal (17), and H2O2 (53).

The striking difference between G{alpha}13 and G{alpha}13-T revealed in the present work is the ability of G{alpha}13-T to induce apoptosis in a cell line that is unaffected by full-length G{alpha}13. We propose that modulation of basal ERK activity by G{alpha}13 is one of the mechanisms that protect the cells from apoptosis on the basis of the following observations. We have shown that G{alpha}13 inhibits the ERK pathway in COS-7 cells (51); at the same time, G{alpha}13 induces apoptosis in COS-7 cells (4). However, in Rat-1 cells in which G{alpha}13 seems to potentiate the ERK pathway, it did not induce apoptosis but instead promotes proliferation and transformation. Finally, G{alpha}13-T that lacks the ability to modulate the ERK pathway also causes apoptosis in Rat-1 cells. Interestingly, the cells were partially rescued from G{alpha}13-T-induced apoptosis by stimulation of ERK with constitutively active MEK. The mechanism of G{alpha}13-induced apoptosis is not understood. Our published and unpublished data suggest that signaling proteins that can contribute to the G{alpha}13-dependent signaling interact with G{alpha}13 in both guanine nucleotide-dependent and -independent ways (37, 47, 48). Due to the low level of expression, it was not possible to address the guanine nucleotide-binding properties of G{alpha}13-T. Because both G{alpha}13-T and G{alpha}13-T harboring putative GTPase-inhibiting mutation (G{alpha}13Q226L-T) were capable of stimulating the JNK pathway and inducing apoptosis, it is conceivable that G{alpha}13-T either exists in the GTP-bound state or constitutively interacts with molecules that signal apoptosis. This important question is a target of our further investigation.

In conclusion, we have shown that 1) mitogen-activated signaling is involved in the regulation of mitogenic and apoptotic responses induced by G{alpha}13, and 2) the ability of G{alpha}13 to transform cells could be dissociated from its propensity to promote apoptotic response. We conclude that this mutant protein will be a useful tool for identifying G{alpha}13-interacting protein(s) that define the ability of G{alpha}13 to induce mitogenesis or apoptosis.


    DISCLOSURES
 
This work was supported by grants from National Institute of General Medical Sciences GM-56159 and GM-65160.


    FOOTNOTES
 

Address for reprint requests and other correspondence: T. Voyno-Yasenetskaya, Dept. of Pharmacology, College of Medicine, Univ. of Illinois, 835 South Wolcott Ave., Chicago, IL 60612 (E-mail: tvy{at}uic.edu).

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
 REFERENCES
 
1. Althoefer H, Eversole-Cire P, and Simon MI. Constitutively active G{alpha}q and G{alpha}13 trigger apoptosis through different pathways. J Biol Chem 272: 24380-24386, 1997.[Abstract/Free Full Text]

2. Alvaro V, Levy L, Dubray C, Roche A, Peillon F, Querat B, and Joubert D. Invasive human pituitary tumors express a point-mutated alpha-protein kinase C. J Clin Endocrinol Metab 77: 1125-1129, 1993.[Abstract]

3. Aspenstrom P. Effectors for the Rho GTPases. Curr Opin Cell Biol 11: 95-102, 1999.[ISI][Medline]

4. Berestetskaya YV, Faure MP, Ichijo H, and Voyno-Yasenetskaya TA. Regulation of apoptosis by alpha subunits of G12 and G13 proteins via apoptosis signal-regulated kinase, Ask1. J Biol Chem 273: 27816-27823, 1998.[Abstract/Free Full Text]

5. Berlot CH and Bourne HR. Identification of effector-activating residues of G{alpha}s. Cell 68: 911-922, 1992.[ISI][Medline]

6. Bourne HR, Sanders DA, and McCormick F. The GTPase superfamily: conserved structure and molecular mechanism. Nature 349: 117-127, 1991.[ISI][Medline]

7. Buckley S, Driscoll B, Barsky L, Weinberg K, Anderson K, and Warburton D. ERK activation protects against DNA damage and apoptosis in heperoxic rats. Am J Physiol Lung Cell Mol Physiol 277: L159-L166, 1999.[Abstract/Free Full Text]

8. Chan AML, Fleming TP, McGovern ES, Chedid M, Miki T, and Aaronson SA. Expression cDNA cloning of a transforming gene encoding the wild-type G{alpha}12 gene product. Mol Cell Biol 13: 762-768, 1993.[Abstract]

9. Chang CL, Zhu XX, Thoraval DH, Ungar D, Rawwas J, Hora N, Strahler JR, Hanash SM, and Radany E. Nm23-H1 mutation in neuroblastoma. Nature 370: 335-336, 1994.[ISI][Medline]

10. Chen Y, Lu Q, Schneeberger EE, and Goodenough DA. Restoration of tight junction structure and barrier function by down-regulation of the mitogen-activated protein kinase pathway in ras-transformed Madin-Darby canine kidney cells. Mol Biol Cell 11: 849-862, 2000.[Abstract/Free Full Text]

11. Coleman DE and Sprang SR. Structure of G{alpha}i1. GppNHp, autoinhibition in a G{alpha} protein-substrate complex. J Biol Chem 274: 16669-16672, 1999.[Abstract/Free Full Text]

12. Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind JS, and Spiegel S. Suppression of ceramide-mediated cell death by sphingosine-1-phosphate. Nature 381: 800-803, 1996.[ISI][Medline]

13. Denis-Henriot D, Mazancourt P, Morot M, and Giudicelli Y. Mutant {alpha}-subunit of the G protein G12 activates proliferation and inhibits differentiation of 3T3-F442A preadipocytes. Endocrinology 139: 2892-2899, 1998.[Abstract/Free Full Text]

14. Dong Q, Brucker-Davis F, Weintraub BD, Smallridge RC, Carr FE, Battey J, Spiegel AM, and Shenker A. Screening of candidate oncogenes in human thyrotroph tumors: absence of activating mutations of the G{alpha}q, G{alpha}11, G{alpha}s, or thyrotropin-releasing hormone receptor genes. J Clin Endocrinol Metab 81: 1134-1140, 1996.[Abstract]

15. Drewinko B, Romsdahl MM, Yang LY, Ahearn MJ, and Trujillo JM. Establishment of a human carcinoembryonic antigen-producing colon adenocarcinoma cell line. Cancer Res 36: 467-475, 1976.[Abstract]

16. Drewinko B, Yang LY, Barlogie B, Romsdahl M, Meistrich M, Malahy MA, and Giovanella B. Further biologic characteristics of a human carcinoembryonic antigen-producing colon carcinoma cell line. J Natl Cancer Inst 61: 75-83, 1978.[ISI][Medline]

17. Erhardt P, Schremser EJ, and Cooper GM. B-Raf inhibits programmed cell death downstream of cytochrome c release from mitochondria by activating MEK/ERK pathway. Mol Cell Biol 19: 308-315, 1999.

18. Farfel Z, Bourne HR, and Iiri T. Mechanisms of disease: the expanding spectrum of G protein disease. N Engl J Med 340: 1012-1020, 1999.[Free Full Text]

19. Faure M, Voyno-Yasenetskaya TA, and Bourne HR. cAMP and {beta}{gamma} subunits of heterotrimeric G proteins stimulate the mitogen-activated protein kinase pathway in COS-7 cells. J Biol Chem 269: 7851-7854, 1994.[Abstract/Free Full Text]

20. Jacinto E, Werlen G, and Karin M. Cooperation between Syk and Rac1 leads to synergistic JNK activation in T lymphocytes. Immunity 8: 31-41, 1998.[ISI][Medline]

21. Jiang H, Wu D, and Simon MI. The transforming activity of activated G{alpha}12. FEBS Lett 330: 319-322, 1993.[ISI][Medline]

22. Kabouridis PS, Waters ST, Escobar S, Stanners J, and Tsoukas CD. Expression of GTP-binding protein alpha subunits in human thymocytes. Mol Cell Biochem 144: 45-51, 1995.[ISI][Medline]

23. Khosravi-Far R, White MA, Westwick JK, Solski PA, Chrzanowska-Wodnicka M, Van Aelest L, Wigler MH, and Der CJ. Oncogenic Ras activation of Raf/mitogen-activated protein kinase-independent pathways is sufficient to cause tumorigenic transformation. Mol Cell Biol 16: 3923-3933, 1996.[Abstract]

24. Lambright DG, Noel JP, Hamm HE, and Sigler PB. The 1.8. A crystal structure of transducin {alpha}. GDP: structural determinants for activation of a heterotirmeric G protein {alpha} subunit. Nature 369: 621-628, 1994.[ISI][Medline]

25. Landis CA, Harsh G, Lyons J, Davis RL, McCormick F, and Bourne HR. Clinical characteristics of acromegalic patients whose pituitary tumors contain mutant Gs protein. J Clin Endocrinol Metab 71: 1416-1420, 1990.[Abstract]

26. Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, and Vallar L. GTPase inhibiting mutations activate the {alpha} chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340: 692-696, 1989.[ISI][Medline]

27. Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ, Heys JR, and Landvatter SW. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 372: 739-746, 1994.[ISI][Medline]

28. Lem J, Chin AC, Thayer MJ, Leach RJ, and Fournier RE. Coordinate regulation of two genes encoding gluconeogenic enzymes by the trans-dominant locus Tse-1. Proc Natl Acad Sci USA 85: 7302-7306, 1988.[Abstract]

29. Lewis TS, Shapiro PS, and Ahn NG. Signal transduction through MAP kinase cascades. Adv Cancer Res 74: 49-139, 1998.[ISI][Medline]

30. Li Y, Sternweis PM, Charnecki S, Smith TF, Gilman AG, Neer EJ, and Kozasa T. Sites for G{alpha} binding on the G protein {beta} subunit overlap with sites for regulation of phospholipase C {beta} and adenylyl cyclase. J Biol Chem 273: 16265-16272, 1998.[Abstract/Free Full Text]

31. Liu W, Dong X, Mai M, Seelan RS, Taniguchi K, Krishnadath KK, Halling KC, Cunningham JM, Qian C, Christensen E, Roches PC, Smith DI, and Thibodeau SN. Mutations in AXIN2 cause colorectal cancer with defective mismatch repair by activating beta-catenin/TCF signalling. Nat Genet 26: 146-147, 2000.[ISI][Medline]

32. Lyons J, Landis CA, Harsh G, Vallar L, Grünewald K, Feichtinger H, Duh QY, Clark OH, Kawasaki E, Bourne HR, and McCormick F. Two G protein oncogenes in human endocrine tumors. Science 249: 655-659, 1990.[ISI][Medline]

33. Mansour SJ, Matten WT, Hermann AS, Candia JM, Rong S, Fukasawa K, Vande Woude GF, and Ahn NG. Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 265: 966-970, 1994.[ISI][Medline]

34. Meigs TE, Fields TA, McKee DD, and Casey PJ. Interaction of G{alpha}12 and G{alpha}13 with the cytoplasmic domain of cadherin provides a mechanism for beta-catenin release. Proc Natl Acad Sci USA 98: 519-524, 2001.[Abstract/Free Full Text]

35. Mitsui H, Takuwa N, Kurokawa K, Exton JH, and Takuwa Y. Dependence of activated G{alpha}12-induced G1 to S phase cell cycle progression on both Ras/mitogen-activated protein kinase and Ras/Rac1/Jun N-terminal kinase cascades in NIH3T3 fibroblasts. J Biol Chem 272: 4904-4910, 1997.[Abstract/Free Full Text]

36. Nagao M, Kaziro Y, and Itoh H. The Src family tyrosine kinase is involved in Rho-dependent activation of c-Jun N-terminal kinase by G{alpha}12. Oncogene 18: 4425-4434, 1999.[ISI][Medline]

37. Niu J, Vaiskunaite R, Carr D, Kozasa T, Dulin N, and Voyno-Yasenetskaya TA. A-Kinase anchoring protein 110 (AKAP110): a novel effector molecule of G13 protein that mediates cAMP-independent PKA activation. Curr Biol 11: 1-5, 2001.[ISI][Medline]

37. Orita M, Iwahana H, Kanazawa H, Hayashi K, and Sekiya T. Detection of polymorphisms of human DNA by gel electrophoresis as single-strand conformation polymorphisms. Proc Natl Acad Sci USA 86: 2766 -2770, 1989.[Abstract]

38. Pace AM, Wong YH, and Bourne HR. A mutant {alpha} subunit of Gi2 induces neoplastic transformation of Rat-1 cells. Proc Natl Acad Sci USA 88: 7031-7035, 1991.[Abstract]

39. Peitsch MC. ProMod and Swiss-model: internet-based tools for automated comparative protein modelling. Biochem Soc Trans 24: 274-279, 1996.[ISI][Medline]

40. Rodrigues GA, Park M, and Schlessinger J. Activation of the JNK pathway is essential for transformation by the Met oncogene. EMBO J 16: 2634-2645, 1997.[Abstract/Free Full Text]

41. Shievella AR, Chen JH, Graham JR, and Lin LL. MADD, a novel death domain protein that interacts with the type 1 tumor necrosis factor receptor and activates mitogen-activated protein kinase. J Biol Chem 272: 12069-12075, 1997.[Abstract/Free Full Text]

42. Sieder S, Richter E, Becker K, Heins R, and Steinfelder HJ. Doxorubicin-resistant LoVo adenocarcinoma cells display resistance to apoptosis induction by some but not all inhibitors of ser/thr phosphatases 1 and 2A. Toxicology 134: 109-115, 1999.[ISI][Medline]

43. Su B and Karin M. Mitogen-activated protein kinase cascades and regulation of gene expression. Curr Opin Immunol 8: 402-411, 1996.[ISI][Medline]

44. Sunahara RK, Tesmer JJ, Gilman AG, and Sprang SR. Crystal structure of the adenylyl cyclase activator Gs{alpha}. Science 278: 1943-1947, 1997.[Abstract/Free Full Text]

45. Tesmer JJ, Berman DM, Gilman AG, and Sprang SR. Structure of GRS4 bound to AlF- 4 -activated Gi{alpha}1: stabilization of the transition state for GTP hydrolysis. Cell 89: 251-261, 1997.[ISI][Medline]

46. Treisman R. Ternary complex factors: growth factor regulated transcriptional activators. Curr Opin Genet Dev 4: 96-101, 1994.[Medline]

47. Vaiskunaite R, Adarichev V, Furthmayr H, Kozasa T, Gudkov A, and Voyno-Yasenetskaya TA. Conformational activation of radixin by G13 protein {alpha} subunit. J Biol Chem 275: 26206-26212, 2000.[Abstract/Free Full Text]

48. Vaiskunaite R, Kozasa T, and Voyno-Yasenetskaya TA. Interaction between G{alpha} subunit of heterotrimeric G12 protein and heat shock protein 90, Hsp90 is required for G{alpha}12 signaling. J Biol Chem 267: 46088-46093, 2001.

49. Vara Prasad MVVS, Dermott JM, Heasley LE, Johnson GL, and Dhanasekaran N. Activation of Jun kinase/stress-activated protein kinase by GTPase-deficient mutants of G{alpha}12 and G{alpha}13. J Biol Chem 270: 18655-18659, 1995.[Abstract/Free Full Text]

50. Vleminckx K, Vakaet L, Mareel M, Fiers W, and van Roy F. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell 66: 107-119, 1991.[ISI][Medline]

51. Voyno-Yasenetskaya TA, Faure M, Ahn N, and Bourne HR. G{alpha}12 and G{alpha}13 regulate ERK and JNK pathways by different mechanisms in COS-7 cells. J Biol Chem 271: 21081-21087, 1996.[Abstract/Free Full Text]

52. Voyno-Yasenetskaya TA, Pace AM, and Bourne HR. Mutant {alpha} subunits of G12 and G13 proteins induce neoplastic transformation of Rat-1 fibroblasts. Oncogene 9: 2559-2565, 1994.[ISI][Medline]

53. Wang X, Martindale JL, Liu Y, and Holbrook NJ. The cellular response to oxidative stress: influences of mitogen-activated protein kinase signaling pathways on cell survival. Biochem J 333: 291-300, 1998.[ISI][Medline]

54. Widmann C, Gibson S, and Jarpe MD. Mitogen-activated protein kinases: conservation of a three kinase module from yeast to human. Physiol Rev 79: 143-180, 1999.[Abstract/Free Full Text]

55. Wittinghofer A. Signal transduction via Ras. Biol Chem 379: 933-937, 1998.[ISI][Medline]

56. Xia Z, Dickens M, Raingeaud J, Davis RJ, and Greenberg ME. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 270: 1326-1331, 1995.[Abstract]

57. Xu N, Bradlet L, Ambdukar I, and Gutkind JS. A mutant {alpha} subunit of G12 potentiates the eicosanoid pathway and is highly oncogenic in NIH3T3 cells. Proc Natl Acad Sci USA 90: 6741-6745, 1993.[Abstract]

58. Xu N, Voyno-Yasenetskaya TA, and Gutkind JS. Potent transforming activity of the G{alpha}13 subunit defines a novel family of oncogenes. Biochem Biophys Res Commun 201: 603-609, 1994.[ISI][Medline]