Type II cGMP‑dependent protein kinase inhibits RhoA activation in gastric cancer cells

  • Authors:
    • Ying Wang
    • Yongchang Chen
    • Yueying Li
    • Ting Lan
    • Hai Qian
  • View Affiliations

  • Published online on: February 18, 2014     https://doi.org/10.3892/mmr.2014.1960
  • Pages: 1444-1452
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Small GTPase RhoA is a key signaling component regulating cell migration and stress fiber formation. Previous studies have shown that RhoA activity is regulated by protein kinases, such as cAMP‑dependent protein kinase (PKA) and type I cGMP‑dependent protein kinase (PKGI), which phosphorylate the protein. This study was designed to investigate the effect of type II cGMP‑dependent protein kinase (PKGII) on RhoA activity. Cells of the human gastric cancer line AGS were infected with adenoviral constructs bearing the PKGII cDNA in order to increase its endogenous expression, and were treated with 8‑pCPT‑cGMP to activate the PKGII enzyme. A transwell assay was performed to measure the migratory activity of the treated cells, and immunofluorescent microscopy was used to observe the formation of stress fibers. The phosphorylation of RhoA was detected by western blotting, and the activity of RhoA was measured by a pull‑down assay. Co‑immunoprecipitation (co‑IP) was performed to detect binding of PKGII to RhoA. Glutathione S‑transferase (GST)‑fused fragments of RhoA and PKGII were expressed in Escherichia coli and used to investigate the domains required for the binding. The results showed that lysophosphatidic acid (LPA) treatment increased the migration and the formation of stress fibers in AGS cells and that this effect was RhoA‑dependent. An increase in PKGII activity, not only inhibited LPA‑induced migration and stress fiber formation, but also suppressed LPA‑induced activation of RhoA. PKGII caused serine 188 (Ser188) phosphorylation of RhoA, but not the phosphorylation of the mutant RhoA Ser188A, and therefore had no inhibitory effect on the activity of the mutant protein. Co‑IP results showed that there is direct binding of PKGII to RhoA. The GST pull‑down assay showed that the fragment containing RhoA amino acid residues 1‑44 and the N‑terminal fragment of PKGII containing amino acid residues 1‑176 are required for the binding between the two proteins. These results suggested that PKGII inhibits RhoA activity by binding to this small GTPase and causing phosphorylation at its Ser188 site.

Introduction

Rho proteins are small GTPases of the Ras family that cycle between an active and an inactive GDP-bound form and regulate diverse cell functions such as cytoskeletal organization, smooth muscle contraction, muscle and neuronal differentiation, cell cycle progression and gene expression (14). RhoA is a key member of the Rho family and is activated downstream of multiple membrane receptors, such as thrombin and lysophosphatidic acid (LPA) receptors. RhoA activity is increased by the guanine nucleotide exchange factor, which promotes the release of bound GDP and subsequent binding of GTP, and is decreased by the GTPase-activating protein, which stimulates the hydrolysis of bound GTP (5). In addition, previous studies (6) have suggested that the phosphorylation-dephosphorylation cycle controls Rho protein activity. For example, the phosphorylation and inhibition of RhoA by the cAMP-dependent protein kinase (PKA) has been highlighted as one of the direct antagonizing mechanisms for RhoA activity. In vitro experiments have indicated that RhoA phosphorylation at serine 188 (Ser188) increases the ability of the Rho GDP-dissociation inhibitor to extract RhoA from the membrane (67). Another report further elucidated the significance of PKA in regulating, through phosphorylation, RhoA activity (8).

Type II cGMP-dependent protein kinase (PKGII) is a serine/threonine kinase. It is involved in several physiological functions including intestinal secretion, bone growth, learning and memory (9). Findings of previous studies have suggested that this kinase plays an important role in regulating additional biological activities such as cell proliferation, differentiation and apoptosis, especially in tumor cells (1013). Previous results from our laboratory showed that PKGII inhibits the proliferation and migration of gastric cancer cell lines through inhibition of the EGF-induced activation of EGFR and of related signal transduction pathways (1416). Since our previous results also showed that PKGII inhibits LPA-induced cell migration (17), whether this kinase exerts an inhibitory effect on RhoA activity should be elucidated. This study was designed to investigate the potential inhibition, by PKGII, of LPA-induced RhoA activation and the underlying mechanism(s) involved.

Materials and methods

Cell lines and reagents

The human gastric cancer cell line AGS, African green monkey kidney fibroblast-like cell line COS-7 and human embryonic kidney (HEK) 293A cell line were provided by the Institute of Cell Biology (Shanghai, China). Adenoviral vectors encoding β-galactosidase (pAd-LacZ) and PKGII (pAd-PKGII), the recombinant pcDNA 3.1 vector bearing the genes encoding PKGII, wild-type RhoA and mutant RhoA Ser188A, and the expression vector pGEX-2T-RBD, encoding the cDNA of glutathione S-transferase (GST) coupled to the Rho-binding domain (RBD) of rhotekin were kind gifts from Dr Gerry Boss and Dr Renate Pilz (University of California, San Diego, CA, USA). Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were from Gibco-BRL (Grand Island, NY, USA). The antibody against PKGII was from Abgent Biotechnology (San Diego, CA, USA). Goat anti-β-actin, mouse anti-RhoA and rabbit anti-phosho (p)-RhoA (Ser188) were from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Mouse anti-FLAG was from Sigma-Aldrich (St. Louis, MO, USA). Rabbit anti-p-LIMK (Thr505/508) was from Bioworld Technology Inc. (St. Louis Park, MN, USA). Horseradish peroxidase-conjugated secondary antibodies were from Jackson ImmunoResearch Laboratories (West Grove, PA, USA). The cellular permeable cGMP analog 8-pCPT-cGMP and the ROCK inhibitor Y27632 were from Calbiochem (San Diego, CA, USA). LPA, TRITC-conjugated phalloidin and the plasmid p3xFLAG-Myc-CMV-24 were purchased from Sigma-Aldrich. The cell transfection reagent Lipofectamin™ 2000 and Escherichia coli BL21 (DE3) were from Invitrogen Life Technologies (Carlsbad, CA, USA). The plasmid pGEX-4T-1 and glutathione-sepharose beads were purchased from GE Healthcare Life Sciences (Piscataway, NJ, USA). Electrochemiluminescence (ECL) reagents were from EMD Millipore (Billerica, MA, USA). Glass bottom dishes were from Shengyou Biotechnology, Co., Ltd. (Hangzhou, China). All other reagents used were of analytical grade.

Preparation of adenoviral vectors

HEK 293A cells were transfected with pAd-LacZ and pAd-PKGII vectors and cultured for ≤10 days until cytopathic effects were observed. The cells and the culture medium were harvested and underwent three freezing-thawing cycles. The supernatants containing adenoviruses (Ad-LacZ and Ad-PKGII) were used to infect new HEK 293A cells to amplify adenoviruses. The amplified adenoviral preparations were titrated to determine the pfu/ml and kept at −80°C until use.

Cell culture, transfection and infection

AGS cells were cultured in DMEM supplemented with 10% FBS and maintained at 37°C in a humidified incubator with 95% air and 5% CO2. The medium was changed every two days and the cells were sub-cultured until they reached confluence. For transfection of gastric cancer cells with plasmids, the cells were sub-cultured the day before the process and the transfection was performed according to the manufacturer’s instructions. On the day before infection with Ad-LacZ and Ad-PKGII, the cells were cultured in fresh medium at 70–80% confluence.

Cloning and expression constructs

To generate FLAG-tagged wild-type RhoA and mutant RhoA Ser188A plasmids, cDNA fragments were amplified from the pcDNA 3.1/full-length wild-type RhoA and /RhoA mutant by PCR. The 5′ primer for amplifying wild-type and mutant RhoA was 5′-CCC AAG CTT ATG GCT GCC ATC CGG AA-3′. The 3′ primers were: for wild-type RhoA, 5′-CGC GGA TCC CAA GAC AAG GCA CCC AGA TT-3′, and for mutant RhoA, 5′-CGC GGA TCC CAA GAC AAG GCA CCC AGC TT-3′. The PCR products were cleaved by HindIII and BamHI and the target fragment was cloned into the expression vector p3xFLAG-Myc-CMV-24.

The cloning vectors for the bimolecular fluorescence complementation (BiFC) assay, pBiFC-VN155 and pBiFC-VC155, were kindly provided by Dr Chang-Deng Hu (Purdue University, West Lafayette, IN, USA). To construct pBiFC-RhoA-VC, a cDNA fragment encoding the full-length RhoA was amplified by PCR from pcDNA 3.1/wild-type RhoA, using the following oligonucleotides: 5′-GGA AGA TCT ACA TGG CTG CCA TCC GGA A-3′ (RhoA-VC-N), 5′-CGG GGT ACC CAA GAC AAG GCA CCC AGA TT-3′ (RhoA-VC-C). The resulting PCR fragment was digested with BglII and KpnI and then inserted into the pBiFC-VC155 vector. To construct pBiFC-PKGII-VN, a cDNA fragment encoding the full-length PKGII was amplified by PCR using the following oligonucleotides bearing BglII and KpnI restriction sites: 5′-GGA AGA TCT CTA TGG GAA ATG GTT CAG TGA AAC-3′ (PKGII-VN-N), 5′-CGG GGT ACC GAA GTC TTT ATC CCA GCC TGA T-3′ (PKGII-VN-C). The amplified fragment was then cloned into the pBiFC-VN155 vector.

Full-length RhoA cDNA and cDNA fragments encoding RhoA amino acids 1–44 and 1–147 were amplified by PCR from the full-length pcDNA 3.1/wild-type RhoA vector. The sequence of the 5′-primer was: 5′-GCG GGA TCC ATG GCT GCC ATC CGG AAG-3′. The sequences of the 3′-primers were: 5′-CGC GTC GAC TCA CAA GAC AAG GCA CCC AGA TTT-3′, 5′-CGC GTC GAC TCA GCC ACA TAG TTC TCA AAC AC-3′ and 5′-CGC GTC GAC TCA CAT ATC TCT GCC TTC TTC AGG-3′ for full-length, RhoA amino acids 1–44 and 1–147, respectively. The PCR products were cleaved by BamHI-EcoRI and the target fragment was cloned into pGEX-4T-1. To generate PKGII cDNA fragments (encoding amino acid residues 1–176, 1–285, 286–762, 286–452, 453–711 and 712–762), cDNA fragments were amplified from the full-length pcDNA 3.1/PKGII by PCR. The following primers were used: 5′ primer for PKGII (1–176) and PKGII (1–285), 5′-TCC CCC GGG TAT GGG AAA TGG TTC AGT G-3′; for PKGII (286–762) and PKGII (286–452), 5′-TCC CCC GGG TTT GCT GAA GAA TTT ACC TG-3′; for PKGII (453–711), 5′-TCC CCC GGG TCT TGA GAT TAT TGC AAC ACT GG-3′ and for PKGII (712–762), 5′-TCC CCC GGG TAA TGG TTT TAA TTG GGA GGG ACT G-3′. The 3′ primers were: for PKGII (1–176), 5′-CCG CTC GAG CTG CTG AGG ATC CAG TCT-3′; for PKGII (1–285), 5′-CCG CTC GAG GGA TAC ACT TCT GAG GAA GTT TCT G-3′; for PKGII (286–452), 5′-CCG CTC GAG GTT CTG GAA TGG GGA TGA TG-3′; for PKGII (453–711), 5′-CCG CTC GAG TAA CCA CCT GTG TTT CTT AAT GTC-3′ and for PKGII (712–762) and PKGII (286–762), 5′-CCG CTC GAG GAA GTC TTT ATC CCA GCC TGA TAG-3′. The PCR products were cleaved by SmaI-XhoI and the target fragment was cloned into pGEX-4T-1. The validity of all the constructs was confirmed by DNA sequencing.

Expression of GST-fusion proteins in E. coli

RhoA and PKGII fragments fused to GST were expressed in the E. coli competent cells BL21 (DE3), and transcription was induced with 0.2 or 1 mM isopropyl-β-D-thiogalactopyranoside. The expressed GST-fusion proteins were purified on glutathione-sepharose beads.

Phalloidin staining

Cells were plated on glass coverslips, serum-starved for 16 h and treated with 250 μM CPT-cGMP for 1 h prior to stimulation with 10 μM LPA for 5 min. Cells were fixed with freshly prepared 40 g/l paraformaldehyde in phosphate-buffered saline (PBS) for 15 min, permeabilized with 0.3% Triton X-100 in PBS for 10 min and blocked with 3% bovine serum albumin in PBS. The cells were subsequently incubated with TRITC-labeled phalloidin (1:100) for 1 h at room temperature, and were visualized with fluorescence microscopy using a Leica DM LB2 microscope digital camera system (Leica Microsystems, Wetzlar, Germany).

Transwell migration assays

The migratory activity of AGS cells was detected using a transwell system comprising BD BioCoat™ Control Cell Culture inserts (8.0-μm PET membrane, 24-well) purchased from BD Biosciences (Franklin Lakes, NJ, USA). After trypsinization, 5×104 cells were seeded into the upper chamber of a tissue culture incubator containing culture medium without FBS. Cell migration to the bottom side of the membrane was induced by medium containing 10% FBS in the lower chamber for 12 h at 37°C. The cells remaining in the upper chamber were carefully removed with cotton swabs. Cells that had migrated to the bottom side of the membrane were fixed in 4% paraformaldehyde solution for 30 min, stained in Giemsa solution for 10 min and then rinsed in water. The stained cells were subjected to microscopic examination under a light microscope. Migrating cells were counted in five randomly selected fields per insert and the values were averaged. Each migration condition experiment was repeated three times.

Confocal laser scanning microscopy

Cos-7 cells (~2×105 cells/dish) were grown on 35-mm dishes with a 20-mm bottom well, and were transfected with BiFC pairs. The BiFC fluorescent signals were detected under a confocal microscope (Zeiss, Oberkochen, Germany).

Western blot analysis

Protein samples were electrophoresed on an SDS-PAGE (8–12%) gel, and membrane transfer was performed following the manufacturer’s instructions (Bio-Rad, Hercules, CA, USA). The primary antibodies were incubated overnight at 4°C in Tris-buffered saline with 2% Tween-20 (TBS-T) and the matching secondary antibodies were incubated for 1 h at room temperature in TBS-T, with three washes after each incubation. ECL reagents were used to allow visualizing the positive bands on the membrane. To perform densitometry analysis, digital images of the positive bands were obtained with the ChemiDoc XRS acquisition system and analyzed using the image analysis program Quantity One (Bio-Rad). The results were expressed as the ratio of target protein/loading control protein.

In vitro pull-down assay

Cells on 100-mm culture plates at ~90% confluence were washed three times with cold PBS and harvested in lysis buffer (25 mM HEPES pH 7.5, 150 mM NaCl, 1% NP40, 10% glycerol, 25 mM NaF, 10 mM MgCl2, 0.25% sodium deoxycholate, 1 mM EDTA, 1 mM Na3VO4, 10 mg/ml aprotinin and 10 mg/ml leupeptin) 48 h after transfection or infection. The supernatant obtained by centrifugation (13,800 × g, 10 min) was then mixed with GST-fusion protein beads and incubated for 2 h at 4°C with rocking. After thorough washing with lysis buffer, the bound proteins were solubilized in 2X SDS buffer and analyzed by western blotting using antibodies against RhoA or PKGII. To assess the amount of protein used in each pull-down assay, 5% of the input lysate was loaded as a control. Beads loaded with GST alone were included as negative controls in all experiments. RhoA activity was detected by a similar method, and the supernatant was incubated with glutathione-sepharose beads and GST-RBD of rhotekin at 4°C for 1 h.

Immunoprecipitation

The cells growing on 100-mm culture plates were washed twice with cold PBS and lysed with RIPA buffer (50 mM Tris-HCl pH 7.4, 1% Triton X-100, 1 mM EDTA, 1 mM leupeptin, 1 mM phenylmethylsulfonyl fluoride, 10 mM NaF and 1 mM Na3VO4) 48 h after transfection and/or infection. The supernatant was obtained by centrifugation (13,800 × g, 10 min) and then mixed with target antibodies or matched immunoglobulin G as a negative control for 12 h at 4°C with rocking. Fresh protein G conjugated to agarose was then added, followed by a 2–3 h incubation at 4°C with rocking. Immunoprecipitates were centrifuged at 400 × g for 2 min at 4°C. The supernatant was discarded and the pellet was washed four times with binding buffer (50 mM Tris-HCl, 250 mM NaCl, 0.05% Nonidet P40, 30 mM MgCl2, pH 7.4) and then resuspended with the same volume of 2X SDS buffer. The precipitates were probed with antibodies against target proteins.

Statistical analysis

Data are expressed as means ± standard deviation (SD). Statistical significance was assessed with analysis of variance (ANOVA) analyses using the SPSS statistical software (IBM, Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

PKGII inhibits the LPA-induced migration and cytoskeletal changes in AGS cells

LPA is a potent mediator in the modulation of the cell migration process. A previous study suggested that the G12/G13-RhoA signaling pathway contributes to efficient LPA-stimulated cell migration (18). To detect the role of RhoA in LPA-induced migration of gastric cancer cells, AGS cells were treated with 10 μM Y27632 (ROCK inhibitor) for 1 h before LPA stimulation. A transwell assay was used to measure the migratory activity of the cells. The results showed that LPA stimulation resulted in ~2-fold increase in cell migration compared to that of unstimulated cells (Fig. 1A). Treatment with Y27632 inhibited the LPA-stimulated cell migration, indicating that LPA-induced cell migration is controlled by the activation of the RhoA/ROCK pathway (Fig. 1A). To investigate the effect of PKGII on LPA-induced migration, AGS cells were infected with Ad-LacZ or Ad-PKGII and then treated with 250 μM 8-pCPT-cGMP for 1 h prior to treatment with LPA. Treatment with 8-pCPT-cGMP markedly and significantly inhibited LPA-induced cell migration in the cells infected by Ad-PKGII but not in those infected by Ad-LacZ, indicating that PKGII activity is important, not only for impeding the LPA-induced migration, but also for inhibition of the LPA-induced activation of RhoA (Fig. 1B). To confirm that PKGII functions through cross-talk with the RhoA/ROCK pathway, we examined the effect of PKGII on LPA-induced stress fiber formation, which is well-known to be dependent on RhoA activation. The results showed that pre-infection with Ad-PKGII and treatment with 8-pCPT-cGMP reduces the formation of stress fibers caused by LPA stimulation (Fig. 2).

PKGII inhibits RhoA activation by phosphorylating RhoA at Ser188

We used a pull-down assay to assess whether RhoA activation is affected by PKGII. Treatment of Ad-PKGII-infected cells with 8-pCPT-cGMP marginally decreased the basal RhoA activity, but inhibited LPA-induced RhoA activation by >50% (Fig. 3). LIMK is a signaling component downstream of RhoA, participating in regulating the formation of stress fibers. We detected Thr505/508 phosphorylation/activation of LIMK and found that PKGII reduced the levels of phosphorylated LIMK which was likely to have been induced by LPA (Fig. 4). These results indicated that inhibition of LPA-induced migration and stress fiber formation by PKGII is associated with inhibition of LPA-induced RhoA activity and its downstream signaling.

It was previously shown that Ser188 is the phosphorylation site through which protein kinases exert their inhibitory effect on RhoA activity (8). We used western blotting with an antibody against RhoA phosphorylated at Ser188 to detect the PKGII-induced phosphorylation of RhoA in AGS cells. The cells were treated as described above. The results showed that the level of phosphorylated RhoA Ser188 was increased in cells infected by Ad-PKGII and treated with 8-pCPT-cGMP (Fig. 5A). In the cells transfected with the plasmid expressing wild-type RhoA, the PKGII activity was sufficient to phosphorylate exogenous RhoA at Ser188. By contrast, in the cells transfected with the plasmid expressing mutant RhoA Ser188A, the phosphorylation of exogenous RhoA at Ser188 did not increase after treatment with 8-pCPT-cGMP (Fig. 5B). These results indicated that PKGII phosphorylates RhoA at Ser188.

PKGII interacts directly with RhoA

The prerequisite for a kinase to phosphorylate its substrate protein is its ability to directly bind the substrate. To determine whether RhoA is the substrate of PKGII, we used a co-immunoprecipitation (co-IP) approach to detect the potentially direct binding of PKGII to RhoA. A binding complex between PKGII and RhoA was detected in AGS cells (Fig. 6). To confirm this result, we performed a BiFC assay, based on the formation of a fluorescent complex by two non-fluorescent fragments of the Venus protein, VN155 (VN) and VC155 (VC), brought together by the association of proteins fused with each Venus fragment (19). We found that diffuse cytoplasmic signals with a few aggregates were present in cells expressing PKGII-VN and RhoA-VC (Fig. 7). This result indicated that PKGII can bind RhoA protein in vitro.

To define the specific domains of RhoA required for binding to PKGII, different RhoA fragments were fused with the GST protein according to Kato et al (20): the RhoA switch I domain (residues 1–44), the RhoA switch I and switch II domains (residues 1–147), and the full-length RhoA (residues 1–192). The fragments were expressed in E. coli, purified and added to cell extracts to test PKGII precipitation. The results showed that all three GST-RhoA constructs, but not GST alone, precipitated PKGII, suggesting that amino acid residues 1–44 of RhoA, constituting the switch I domain, are sufficient for the interaction with PKGII (Fig. 8A). We also investigated which domains of PKGII mediate the PKGII-RhoA interaction. GST-fusion proteins containing specific domains of PKGII (Fig. 8B) were generated and incubated with cell lysate containing abundant RhoA protein. Co-IP results showed that RhoA is detected in pull-downs containing the PKGII N-terminal portion, either using the fragments containing amino acid residues 1–176 or residues 1–285 (Fig. 8C). This indicated that the fragment with amino acid residues 1–176 of PKGII is important for the interaction with RhoA.

Discussion

PKGs are serine/threonine kinases that are widely distributed in eukaryotes. Two genes (prkg1 and prkg2) code for PKGs, the type I cGMP-dependent protein kinase (PKGI) and PKGII. In mammals, PKGI is widely expressed in vascular smooth muscle cells, platelets, lung, certain endothelial cells, fibroblasts, heart and the cerebellum (21). PKGI has been identified as a tumor suppressor protein based on its profound anti-tumor effect (22,23). Unlike PKGI, PKGII is a tissue-specific enzyme that is expressed mainly in the brush border of intestinal mucosa and in specific regions of the brain (24,25). PKGII is involved in regulating electrolyte and water secretion, renin and aldosterone secretion and in the adjustment of the biological clock (9,25). In recent years, new insights on the roles of PKGII on cell proliferation, migration and apoptosis were reported. For example, Swartling et al (11) found that PKGII has anti-proliferative effects on human glioma cells. Wang et al (12) reported a pro-differentiation effect of PKGII in colon cancer cells. Results from our laboratory have shown that PKGII inhibits proliferation and MAPK-mediated signal transduction in human gastric cancer cells (14,15). Furthermore, we demonstrated that PKGII inhibits migration of gastric cancer cells by blocking EGFR activation and consequently, PLCγ1- and MAPK/ERK-mediated signal transduction pathways (16). Overall, these studies indicate that PKGII plays important roles in regulating a number of tumor cell biological activities, and that this kinase, similarly to PKGI, is a potential tumor suppressor protein.

The small G protein RhoA, a member of the Rho subfamily of the Ras superfamily, is important for actin/myosin-based cortical contractility, migration, invasion, transformation, proliferation and survival of tumor cells (14). In the GTP-bound active state, RhoA translocates to the plasma membrane, where it interacts with effectors to transduce downstream signals. It has been confirmed that RhoA activates ROCK1 and then LIMK proteins to regulate the formation of stress fibers and induce the migration of cancer cells (2628). Numerous signaling molecules may cross-talk with RhoA. Among them, protein kinases exert their effect through phosphorylation-dephosphorylation of the protein. In most cases, phosphorylation occurs at a serine residue located at the C-terminal domain of RhoA protein, and modifies its cellular localization. The phosphorylation and inhibition of RhoA by PKA is well established (6,7). It was shown that PKGI can also phosphorylate RhoA at Ser188 and thereby, inactivate RhoA signaling (29,30). However, no study thus far has provided evidence that RhoA is the direct substrate of PKGII.

In the present study, we confirmed the inhibitory effect of PKGII on RhoA and elucidated the underlying mechanism in vitro. Previous studies have shown that the G12/G13-RhoA pathway is important for LPA-induced cell migratory activity, and the majority of studies available on stress fiber signaling pathways have identified RhoA as the major regulator of stress fiber formation under most physiological conditions (18,31). To confirm this in gastric cancer cells, we used Y27632, an inhibitor of ROCK, to identify the role of the RhoA/ROCK pathway in the process. The results showed that Y27632 partially blocked LPA-induced migration and stress fiber formation in AGS cells, indicating that the RhoA/ROCK pathway is involved in these processes. To determine whether RhoA is the key target of PKGII, we examined the inhibitory effect of PKGII on RhoA activation. The results showed that PKGII inhibits the LPA-induced activation of RhoA. Since the inhibition of RhoA is associated with its phosphorylation status, we measured the phosphorylation level of RhoA in cells with high activity of PKGII. The results confirmed that PKGII phosphorylates RhoA at Ser188, and that this phosphorylation partially contributes to the decrease in RhoA activation.

To elucidate whether RhoA is the direct substrate of PKGII, we investigated and confirmed the binding between RhoA and PKGII by using co-IP and BiFC assays. Kato et al (20) found that PKGI binds directly to RhoA via a direct interaction between the amino-terminus of RhoA (residues 1–44), containing the switch I domain and the amino-terminus of PKGI (residues 1–59), which includes a leucine zipper heptad repeat motif. PKGII has a domain structure similar to PKGI, consisting of an N-terminal regulatory domain, which contains a dimerization and an auto-inhibitory region, two cGMP-binding domains and a C-terminal catalytic domain. However, the position of the high- and low-affinity cGMP-binding domains in PKGII is reversed in comparison to PKGI (32). Moreover, the two PKG iso-enzymes exhibit a different affinity towards various membrane permeable cGMP-analogs, which allows their differentiation (24). To identify the domains of PKGII and RhoA that are involved in their interaction, we expressed fusion proteins with GST and RhoA or PKGII fragments and assessed their binding ability by GST pull-down assays. These experiments demonstrated that the N-terminal of PKGII and the switch I domain of RhoA are required for PKGII-RhoA binding. Taken together, these data support that PKGII directly binds to RhoA.

Since RhoA is involved in the biological activity of tumor cells, new and more effective methods blocking the RhoA/ROCK signal transduction pathway are expected to be useful in cancer therapy. Therefore, our finding that PKGII can inhibit RhoA and downstream signal transduction molecules is of high significance. It provides the evidence that PKGII is a potential tumor inhibitor and may also provide information on the development of therapeutic strategies against cancer.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (nos. 81272755, 31040002 and 81201959) and the Specialized Research Fund for Senior Personnel Program of Jiangsu University (no. 11JDG032). We thank Dr Gerry Boss and Dr Renate Pilz at the University of California, San Diego, CA, USA for the kind gifts of adenoviral constructs.

References

1 

Vega FM and Ridley AJ: Rho GTPases in cancer cell biology. FEBS Lett. 582:2093–2101. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Jaffe AB and Hall A: RHO GTPases: biochemistry and biology. Annu Rev Cell Dev Biol. 21:247–269. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar

4 

Friedl P and Wolf K: Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 3:362–374. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Bos JL, Rehmann H and Wittinghofer A: GEFs and GAPs: critical elements in the control of small G proteins. Cell. 129:865–877. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Forget MA, Desrosiers RR, Gingras D and Beliveau R: Phophorylation states of Cdc42 and RhoA regulate their interactions with Rho GDP dissociation inhibitor and their extraction from biological membranes. Biochem J. 361:243–254. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Lang P, Gesbert F, Delespine-Carmagnat M, Stancou R, Pouchelet M and Bertoglio J: Protein kinase A phosphorylation of RhoA mediates the morphological and functional effects of cyclic AMP in cytotoxic lymphocytes. EMBO J. 15:510–519. 1996.PubMed/NCBI

8 

Ellerbroek SM, Wennerberg K and Burridge K: Serine phosphorylation negatively regulates RhoA in vivo. J Biol Chem. 278:19023–19031. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Hofmann F: The biology of cyclic GMP-dependent protein kinases. J Biol Chem. 280:1–4. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Cook AL and Haynes JM: Protein kinase G II-mediated proliferative effects in human cultured prostatic stromal cells. Cell Signal. 16:253–261. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Swartling FJ, Ferletta M, Kastemar M, Weiss WA and Westermark B: Cyclic GMP-dependent protein kinase II inhibits cell proliferation, Sox9 expression and Akt phosphorylation in human glioma cell lines. Oncogene. 28:3121–3131. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Wang R, Kwon IK, Thangaraju M, Singh N, Liu K, Jay P, Hofmann F, Ganapathy V and Browning DD: Type 2 cGMP-dependent protein kinase regulates proliferation and differentiation in the colonic mucosa. Am J Physiol Gastrointest Liver Physiol. 303:G209–G219. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Fallahian F, Karami-Tehrani F, Salami S and Aghaei M: Cyclic GMP induced apoptosis via protein kinase G in oestrogen receptor-positive and -negative breast cancer cell lines. FEBS J. 278:3360–3369. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Chen YC, Ren F, Sang JR, Tao Y and Xu WR: Type II cGMP-dependent protein kinase inhibits proliferation of the gastric cancer cell line BGC-823. Mol Med Rep. 3:361–366. 2010.PubMed/NCBI

15 

Wu Y, Chen Y, Qu R, Lan T and Sang J: Type II cGMP-dependent protein kinase inhibits EGF-triggered signal transduction of the MAPK/ERK-mediated pathway in gastric cancer cells. Oncol Rep. 27:553–558. 2012.PubMed/NCBI

16 

Jiang L, Lan T, Chen Y, Sang J, Li Y, Wu M, Tao Y, Wang Y, Qian H and Gu L: PKG II inhibits EGF/EGFR-induced migration of gastric cancer cells. PLoS One. 8:e616742013. View Article : Google Scholar : PubMed/NCBI

17 

Ren F, Gao Q, Tao Y, Sang J, Yang X, Fan S and Chen Y: Impacts of cGMP dependent protein kinase II on gastric cancer cell BGC-823 migration. J Jiangsu Univ (Med Ed). 20:113–116. 2010.

18 

Bian D, Mahanivong C, Yu J, Frisch SM, Pan ZK, Ye RD and Huang S: The G12/13-RhoA signaling pathway contributes to efficient lysophosphatidic acid-stimulated cell migration. Oncogene. 25:2234–2244. 2006.

19 

Kodama Y and Hu CD: An improved bimolecular fluorescence complementation assay with a high signal-to-noise ratio. Biotechniques. 49:793–805. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Kato M, Blanton R, Wang GR, Judson TJ, Abe Y, Myoishi M, Karas RH and Mendelsohn ME: Direct binding and regulation of RhoA protein by cyclic GMP-dependent protein kinase Iα. J Biol Chem. 287:41342–41351. 2012.

21 

Keilbach A, Ruth P and Hofmann F: Detection of cGMP dependent protein kinase isozymes by specific antibodies. Eur J Biochem. 208:467–473. 1992. View Article : Google Scholar : PubMed/NCBI

22 

Deguchi A, Thompson WJ and Weinstein IB: Activation of protein kinase G is sufficient to induce apoptosis and inhibit cell migration in colon cancer cells. Cancer Res. 64:3966–3973. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Hou Y, Gupta N, Schoenlein P, Wong E, Martindale R, Ganapathy V and Browning D: An anti-tumor role for cGMP-dependent protein kinase. Cancer Lett. 240:60–68. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Lohmann SM, Vaandrager AB, Smolenski A, Walter U and De Jonge HR: Distinct and specific functions of cGMP-dependent protein kinases. Trends Biochem Sci. 22:307–312. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Hofmann F, Feil R, Kleppisch T and Schlossmann J: Function of cGMP-dependent protein kinases as revealed by gene deletion. Physiol Rev. 86:1–23. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, Obinata T, Ohashi K, Mizuno K and Narumiya S: Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 285:895–898. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Yiu G and He Z: Glial inhibition of CNS axon regeneration. Nat Rev Neurosci. 7:617–627. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Caramel J, Quignon F and Delattre O: RhoA-dependent regulation of cell migration by the tumor suppressor hSNF5/INI1. Cancer Res. 68:6154–6161. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Sawada N, Itoh H, Yamashita J, Doi K, Inoue M, Masatsugu K, Fukunaga Y, Sakaguchi S, Sone M, Yamahara K, et al: cGMP-dependent protein kinase phosphorylates and inactivates RhoA. Biochem Biophys Res Commun. 280:798–805. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Sauzeau V, Le Jeune H, Cario-Toumaniantz C, Smolenski A, Lohmann SM, Bertoglio J, Chardin P, Pacaud P and Loirand G: Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth muscle. J Biol Chem. 275:21722–21729. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Pellegrin S and Mellor H: Actin stress fibres. J Cell Sci. 120:3491–3499. 2007. View Article : Google Scholar

32 

Vaandrager AB, Hogema BM and de Jonge HR: Molecular properties and biological functions of cGMP-dependent protein kinase II. Front Biosci. 10:2150–2164. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-April
Volume 9 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Chen Y, Li Y, Lan T and Qian H: Type II cGMP‑dependent protein kinase inhibits RhoA activation in gastric cancer cells. Mol Med Rep 9: 1444-1452, 2014
APA
Wang, Y., Chen, Y., Li, Y., Lan, T., & Qian, H. (2014). Type II cGMP‑dependent protein kinase inhibits RhoA activation in gastric cancer cells. Molecular Medicine Reports, 9, 1444-1452. https://doi.org/10.3892/mmr.2014.1960
MLA
Wang, Y., Chen, Y., Li, Y., Lan, T., Qian, H."Type II cGMP‑dependent protein kinase inhibits RhoA activation in gastric cancer cells". Molecular Medicine Reports 9.4 (2014): 1444-1452.
Chicago
Wang, Y., Chen, Y., Li, Y., Lan, T., Qian, H."Type II cGMP‑dependent protein kinase inhibits RhoA activation in gastric cancer cells". Molecular Medicine Reports 9, no. 4 (2014): 1444-1452. https://doi.org/10.3892/mmr.2014.1960