RhoA mediates the expression of acidic extracellular pH-induced matrix metalloproteinase-9 mRNA through phospholipase D1 in mouse metastatic B16-BL6 melanoma cells

  • Authors:
    • Toyonobu Maeda
    • Satoshi Yuzawa
    • Atsuko Suzuki
    • Yuh Baba
    • Yukio Nishimura
    • Yasumasa Kato
  • View Affiliations

  • Published online on: January 5, 2016     https://doi.org/10.3892/ijo.2016.3322
  • Pages: 1251-1257
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Solid tumors are characterized by acidic extracellular pH (pHe). The present study examined the contribution of small GTP-binding proteins to phospholipase D (PLD) activation of acidic pHe-induced matrix metalloproteinase-9 (MMP-9) production. Acidic pHe-induced MMP-9 production was reduced by C3 exoenzyme, which inhibits the Rho family of GTPases; cytochalasin D, which inhibits actin reorganization; and simvastatin, which inhibits geranylgeranylation of Rho. Small interfering RNA (siRNA) against RhoA, but not against Rac1 or Cdc42, significantly inhibited acidic pHe induction of MMP-9. Pull-down assays showed that acidic pHe increased the activated form of RhoA. Forced expression of constitutively active RhoA induced MMP-9 production, even at neutral pHe. RhoA siRNA also reduced acidic pHe induced PLD activity. Specific inhibition of PLD1 and Pld1 gene knockout significantly reduced acidic pHe-induced MMP-9 expression. In contrast, PLD2 inhibition or knockout had no effect on MMP-9 expression. These findings suggested that RhoA-PLD1 signaling is involved in acidic pHe induction of MMP-9.

Introduction

Tumor cells generate energy through aerobic glycolysis rather than through oxidative phosphorylation in mitochondria. Thus, tumor cells are characterized by acidic extracellular pH (pHe), a phenomenon known as the Warburg effect, due to lactate production through aerobic glycolysis. H+ generated during glycolysis must be secreted to maintain a neutral or weakly alkaline intracellular pH. Several mechanisms are involved in H+ secretion, including transporters and/or exchangers such as monocarboxylate transporters (MCTs), which act as H+-lactate co-transporters, and Na+/H+ exchangers (1). Acidity is also caused by CO2 generated through the pentose phosphate pathway (2). We have reported that acidic pHe induces the expression of matrix metalloproteinase-9 (MMP-9), acting for type IV collagen break down (3,4). The ability of acidic pHe to induce tumor invasion and metastasis (510) suggests that acidic pHe is an important microenvironment supporting tumor malignancy.

The epithelial mesenchymal transition (EMT) is a critical process for tumor invasion and metastasis. EMT can be mediated by growth factors, including transforming growth factor (TGF)-β (11). Notably, we and others have reported that acidic pHe induces EMT in several cell models (10,12). Although actin cytoskeletal reorganization is involved in EMT, disruption of this pathway was reported associated with the downregulation of MMP-9 expression (13,14). Indeed, the intracellular signaling pathway that involves acidic pHe has not been completely elucidated (1525).

Phospholipase D (PLD) is an esterase that hydrolyzes the phospholipids of plasma membranes of mammalian cells. PLD has two major isozymes, PLD1 and PLD2 (26), which localize at perinuclear regions and plasma membranes, respectively (27). Activators of these enzymes include phosphatidylinositol 4,5-bisphosphate (PIP2), small GTPases and ADP-ribosylation factors. Although melanocytes possess negligible levels of PLD1 and PLD2, melanomas express much higher levels of PLD1 (28). In contrast, PLD2 plays a role in the progression of colorectal cancer (29). Thus, the association between these isozymes and malignant phenotypes is likely cell type specific. We have reported that acidic pHe induces Mmp-9 mRNA expression through intracellular pathways involving Ca2+-triggered PLD, mitogen activated protein (MAP) kinase and nuclear factor-κB (NF-κB) (15,16) and acidic sphingomyelinase and NF-κB (16). However, the mechanism underlying acidic pHe-induced PLD isozyme expression remains unclear. The present study was designed to assess the role of the RhoA-PLD1 axis in acidic pHe-induced MMP-9 production.

Materials and methods

Reagents

Dulbecco's modified Eagle's medium (DMEM), Ham's F12 medium, and High Capacity RNA-to-cDNA kits were purchased from Life Technologies (Grand Island, NY, USA). Simvastatin and C3 exoenzyme from Clostridium botulinum were from Merck Millipore (Darmstadt, Germany). Rhodamine-phalloidin and RhoA Pull-down Activation Assay Biochem kits were from Cytoskeleton, Inc. (Denver, CO, USA). Isogen total RNA extraction kits were purchased from Nippon Gene (Tokyo, Japan), and SYBR Premix Ex Taq II and Xfect Transfection reagent were from Takara Bio (Tokyo, Japan). VU0359595 and CAY10594 were from Cayman Chemical (Ann Arbor, MI, USA). Fetal bovine serum (FBS) was from HyClone Laboratories Inc. (South Logan, UT, USA). The blocking reagent N102 was from NOF Corp. (Tokyo, Japan). Immobilon-P PVDF membranes and chemiluminescence reagent were from Merck Millipore (Billerica, MA, USA). Avidin-conjugated horseradish peroxidase (HRP) was from Bio-Rad Laboratories (Hercules, CA, USA). Dual reporter assay kits were from Toyo Ink (Tokyo, Japan).

Vectors and transfection

Constitutively active RhoA vector (pRK5mycL63RhoA) and its control vector (pRK5myc) were the kind gifts of Dr Alan Hall, Memorial Sloan Kettering Cancer Center (New York, NY, USA). The luciferase reporter gene constructs driven by the 5′-flanking region of human MMP-9 (-670) were from Dr Douglas D. Boyd, MD Anderson Cancer Center (Houston, TX, USA). pSpCas9(BB)-2A-GFP (PX458) was obtained from Addgene (Cambridge, MA, USA), and the cytomegalovirus-driven Renilla luciferase reporter vector (pRL-CMV) was from Promega (Madison, WI, USA). Vectors were transfected with Xfect Transfection reagent according to the manufacturer's protocol.

Cell and cell culture

B16-BL6 cells were kindly provided by Dr Kaoru Miyazaki, Yokohama City University (Yokohama, Japan). The basal culture medium consisted of a 1:l mixture of Dulbecco's modified Eagle's medium and Ham's F-12 medium (DMEM/F12) supplemented with 4 mM phosphoric acid, 15 mM HEPES, 1.8 g/l NaHCO3, 100 units/ml penicillin G, and 0.1 mg/ml streptomycin sulfate, adjusted to the desired pH with HCl or NaOH. For serial culture, basal medium at pH 7.4 was supplemented with 10% fetal bovine serum (FBS). All cells were cultured at 37°C in a 5% CO2 incubator.

Immunofluorescence microscopy

Cells were incubated in basal medium (pH 7.4) containing 10% FBS for 1 day on glass coverslips in 6-well plates. The cells were fixed with 4.0% paraformaldehyde in phosphate-buffered saline (PBS), pH 7.4, and permeabilized in PBS containing 0.2% Triton X-100. After washing with PBS, the cells were incubated with 20% N102 blocking reagent in PBS. For F-actin staining, the cells were incubated with rhodamine-phalloidin (1 U/ml) for 30 min at room temperature, followed by three washes with PBS.

Zymography

MMP-9 activity was assessed by zymography, essentially as previously described (3,4,10,15,16,30). Briefly, conditioned media were collected from the cultures and centrifuged to remove cellular debris. The supernatants were mixed with 2.5 volumes of acetone and centrifuged. The precipitated proteins were dissolved in non-reducing sample buffer for sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The samples were electrophoresed on 7.5% polyacrylamide gels containing 0.1% gelatin. The gels were washed with 2.5% Triton X-100 containing 50 mM Tris-HC1 (pH 7.5) and 0.1 M NaCl at room temperature for 2 h, and incubated in 50 mM Tris-HC1 (pH 7.5) containing 10 mM CaCl2 at 37°C for 20 h, and stained with Coomassie brilliant blue R-250.

Pull-down assays

Active RhoA was measured using RhoA Pull-down Activation Assay Biochem kits. Because the GTP-bound form of active RhoA specifically binds rhotekin peptide, consisting of the Rho binding region (amino acids 7–89), fused to glutathione S-transferase (GST-rhotekin), this complex was purified using glutathione affinity beads and active RhoA level analyzed by immunoblotting with anti-RhoA antibody as previously described (15). Briefly, the complexes were separated by 10% SDS-PAGE and transferred onto Immobilon-P PVDF membranes. The membranes were blocked with TBS-T (20 mM Tris-HCl, pH 7.5, 150 mM NaCl, and 0.05% Tween-20) containing 20% blocking reagent N102, treated with anti-RhoA antibody, and incubated with biotin-conjugated secondary antibody and avidin-conjugated horseradish peroxidase. Signals were detected with enhanced chemiluminescence reagents. As a positive control, total RhoA level was analyzed in cell lysates.

Mmp-9 promoter assay

MMP-9 promoter activity was measured using luciferase reporter gene assays with an MMP-9 promoter construct, as previously described (15). Transfection efficiency was monitored by co-transfection with Renilla luciferase reporter vector (pRL-CMV), with reporter activity measured using a dual reporter assay kit, according to the manufacturer's protocol.

Gene knockout of PLD isozymes

Cells with knockout of either the PLD1 or PLD2 were generated using the CRISPR-Cas9 system (31). Single guide RNA (sgRNA) sequences targeting PLD1 and PLD2 were 5′-gtggaatatgacgcatctccagg-3′ and 5′-ttgaggtccaggtcggaaaaagg-3′, respectively. Vectors were transfected with Xfect Transfection reagent. Clones showing lack of target gene expression were selected by the colony formation method.

PLD activity

PLD activity was indirectly measured using PLD assay kits (BioVision, Milpitas, CA, USA), according to the manufacturer's protocol. Because PLD cleaves phospha-tidylcholine to choline and phophatide, choline was oxidized with choline oxidase, yielding H2O2, which was measured using a H2O2-sensitive probe at 570 nm.

Reverse transcription quantitative polymerase chain reaction (RT-qPCR)

Total RNA was purified with Isogen and reverse-transcribed to cDNA using a High-Capacity cDNA Reverse Transcription kit. Sequences corresponding to Pld1, Pld2, MMP-9 and Actb (β-actin) mRNAs were amplified using specific primers and SYBR Premix Ex Taq II in a Thermal Cycler Dice real-time system (TP-870; Takara Bio). The specific primer sequences were 5′-acacaccggtgtgcggatct-3′ (upstream) and 5′-cgtggtctgggtgtctcatc-3′ (downstream) for Pld1 (134 bp); 5′-catcagcatgacagctatgcc-3′ (upstream) and 5′-ttcttccgccttcctcttga-3′ (downstream) for Pld2 (219 bp); 5′-gccctggaactcacacgaca-3′ (upstream) and 5′-ttggaaactcacacgccagaa-3′ (downstream) for Mmp-9 (85 bp); and 5′-catccgtaaagacctctatgccaac-3′ (upstream) and 5′-atggagccaccgatccaca-3′ (downstream) for β-actin (85 bp). The levels of expression of Pld1, Pld2 and Mmp-9 mRNAs were normalized to those of β-actin mRNA in the same samples.

Statistical analysis

Statistical significance was determined by the Student's t-test. P-values <0.05 were considered to indicate a statistically significant result.

Results

Reduction of Rho inhibits acidic pHe-induced MMP-9 production

We have reported that acidic pHe -induced morphological changes in B16-F10 melanoma (3) and Lewis lung carcinoma (10) cell lines, with these cells showing a fibroblastic morphology. Similarly, an acidic pHe-induced morphological changes, independently of cell density, in B16-BL6 cells (Fig. 1A), and enhanced the accumulation of F-actin aggregates, most of which were associated with the plasma membrane (Fig. 1B). Because small G-proteins such as RhoA play important roles in actin reorganization (32), we tested whether cytochalasin D and C3 exoenzyme, which specifically inhibit actin polymerization and Rho, respectively, reduced acidic pHe-induced MMP-9 production, finding that both of these agents inhibited MMP-9 induction (Fig. 2A). Because geranylgeranylation is essential for Rho activation and statins can inactivate Rho by reducing geranylgeranylation (32), we tested the effects of simvastatin on these cultures, finding that simvastatin inhibited acidic pHe-induced MMP-9 production in a dose-dependent manner (Fig. 2B). To assess the contribution of RhoA to the acidic pHe signaling pathway responsible for the induction of MMP-9, we treated cells with RhoA siRNA. We found that RhoA siRNA significantly inhibited MMP-9 induction, whereas Rac1 siRNA and Cdc42 siRNA did not (Fig. 3A). Pull-down assays showed that acidic pHe increased the level of active RhoA (Fig. 3B). Furthermore, transduction of a constitutively active RhoA expression vector into B16-BL6 cells resulted in the induction of MMP-9, even at physiological pHe (Fig. 3C). Taken together, these findings suggested that RhoA is a molecule that transduces acidic signaling to induce expression of MMP-9.

PLD1 is a downstream effector of acidic pHe signaling in MMP-9 induction

We previously reported that PLD is important for transducing acidic pHe signaling (15,16). Moreover, RhoA has been found to activate PLD (26). Therefore, we tested the effects of RhoA siRNA on PLD activity. RhoA siRNA reduced not only PLD activity at pHe 5.9 but also the basal PLD activity at pHe 7.4 (Fig. 4). Because PLD has two isozymes, PLD1 and PLD2 (26), we determined which one is involved in the acidic pHe induction of MMP-9. We found that the specific PLD1 inhibitor VU0359595 significantly reduced acidic pHe-induced MMP-9 production, whereas the specific PLD2 inhibitor CAY10594 did not (Fig. 5A and B). This was confirmed by RT-qPCR assessments of the level of Mmp-9 mRNA expression (Fig. 5C). Furthermore, we used the CRISPR-Cas9 system to knockout either PLD isozyme (Fig. 6A), finding that PLD1 KO completely inhibited acidic pHe-induced MMP-9 production whereas PLD2 KO had no effect on its expression (Fig. 6B). This result was also confirmed by RT-qPCR for Mmp-9 mRNA (Fig. 6B). Acidic pHe also enhanced the levels of both PLD1 and PLD2 mRNAs (Fig. 6C). Taken together, these findings suggest that RhoA-PLD1 plays an important role in acidic pHe induction of MMP-9.

Discussion

The present study expands on our previous findings, showing that acidic pHe activates PLD leading to MMP-9 induction. The present study initially focused on the mechanism of PLD activation. PLD activity has been closely associated with actin reorganization, as small GTPases such as the Rho family and Arf synergistically activate PLD (26). RhoA is a more potent PLD activator than other members of the Rho family such as Rac1 and Cdc42, with the latter two shown to rescue PLD activity when RhoA was inactivated (33). This study showed that RhoA knockdown by a specific siRNA reduced acidic pHe-induced MMP-9 expression, whereas Rac1 and Cdc42 siRNAs had no effect. These findings indicate that Rac1 and Cdc42 are not involved in acidic pHe signaling and that they cannot rescue RhoA activity in the PLD-MMP-9 axis. These findings are in good agreement with results showing the importance of RhoA activation in MMP-9 expression (34,35). Our and other research groups have investigated the role of acidic pHe in the induction of EMT (10,12), showing that RhoA activation is important for actin reorganization in EMT. Cytochalasin D was found to enhance mesenchymal epithelial transition along with E-cadherin by reducing RhoA activity and reduced expression of vimentin and N-cadherin (13). Sustained Ras activity was shown to reduce Rac, which can inhibit Rho function, whereas induction of Rho can result in EMT in transformed cells. Cytoskeletal rearrangement induced by acidic pHe is therefore important for EMT through the RhoA-PLD axis.

TPA is an analog of diacylglycerol that activates protein kinase C (PKC) isoforms such as conventional PKC (DAG and Ca2+-dependent; α, βI, βII and γ) and novel PKC (DAG-dependent, but Ca2+-independent; δ, ɛ, η and θ), contributing to the activation of AP-1 and PLD (36). We have shown that TPA did not induce MMP-9 expression in B16-BL6 cells at neutral pHe but enhanced MMP-9 expression at acidic pHe, at least in part through NF-κB, but not AP-1 activation (15,16). However, bisindolylmaleimide III, a potent PKCa inhibitor, attenuated MMP-9 expression at low pHe (data not shown).

PLD has two major isozymes, PLD1 and PLD2 (26). To determine which of these isozymes contributed to acidic pHe signaling, we used isozyme specific inhibitors and gene knockout technique for each PLD. These experiments showed that PLD1 was responsible for the induction of Mmp-9 mRNA expression at acidic pHe. Human melanoma cells predominantly express PLD1 (28). Although PLD1 activity is inducible, PLD2 activity is constitutive and shows a reduced response to stimulation by small GTPases (27). TPA was found to enhance Pld1, but not Pld2, promoter activity, resulting in MMP-9 secretion through PKCβII-Ras-MAP kinase-NF-κB signaling (37). Our results also showed that PLD1, but not PLD2, was associated with acidic pHe induced MMP-9 production. We also found that increased PLD activity may be due to increases in both Pld1 gene expression and RhoA-induced PLD1 activation, as well as to Ca2+ influx (16), suggesting the existence of an intracellular signaling loop for gain of function of PLD1. Acidic pHe increased the expression of Pld2 mRNA in PLD1-KO cells, but these cells did not respond to acidic medium. This finding suggests that PLD2 cannot compensate for PLD1 in the acidic pHe signaling.

We have reported that PLD activation leads to MMP-9 expression through NF-κB (15). Deletion of the NF-κB-binding-site from the Mmp-9 promoter construct attenuated acidic pHe-induced Mmp-9 promoter activity when compared with wild-type (WT) promoter (15). In contrast, the promoter activity ratio between neutral and acidic pHe was similar in the mutant and WT constructs, suggesting that PLD-NF-κB signaling is essential for Mmp-9 expression. In agreement, this study showed that RhoA siRNA reduced PLD activities but had no effect on the activity ratio of neutral and acidic pHe. Taken together, these findings indicate that PLD1 activity is essential in the induction of Mmp-9 mRNA, with additional signaling, such as acidic sphingomyelinase, required for further induction of Mmp-9 gene expression by acidic pHe (16).

Generally, tumor cells become hypoxic ~100 μm from blood vessels. Because the expression of metabolic enzymes in glycolysis, such as glyceraldehyde dehydrogenase (GAPDH) and enolase, is dependent on hypoxia-inducible factor (HIF) (38), increased tumor acidity may occur in hypoxic areas, although hypoxia may not be essential for tumor acidity (2). Increases in glucose products, such as pyruvate and lactate, can result in the accumulation of HIF-induced proteins under aerobic conditions (39), indicating that glycolysis can associate the signaling of both hypoxia and acidic pHe. Hypoxia has been shown to induce the EMT in gastric cancer cells through TGFβ signaling in an autocrine manner (40). TGF acts synergistically with other growth factors, including epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) (41,42). Further analysis is required to determine relationships between growth factors and acidic pHe signaling

In conclusion, the present study investigated whether acidic pHe, a hallmark of malignant tumors, contributes to the metastatic phenotype through the RhoA-PLD1 axis. Inhibition of isoprenylation by statins such as simvastatin is useful for preventing metastasis driven by acidic pHe.

Acknowledgements

We thank Dr Alan Hall (Memorial Sloan Kettering Cancer Center, deceased 3rd May, 2015), Dr Douglas D. Boyd (MD Anderson Cancer Center, University of Texas, TX, USA) and Kaoru Miyazaki (Kanagawa Cancer Center Research Institute, Japan) for providing constitutive RhoA expression vector, the luciferase reporter gene constructs driven by the 5′-flanking region of human MMP-9 (-670), and B16-BL6 cells, respectively. We also thank Dr Ryu-Ichiro Hata (Kanagawa Dental University, Japan) for his critical suggestions. The present study was partly supported by Youth Encouragement Grants (to S.Y.) from Ohu University, Japan.

References

1 

Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T and Baba Y: Acidic extracellular microenvironment and cancer. Cancer Cell Int. 13:892013. View Article : Google Scholar : PubMed/NCBI

2 

Helmlinger G, Sckell A, Dellian M, Forbes NS and Jain RK: Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res. 8:1284–1291. 2002.PubMed/NCBI

3 

Kato Y, Nakayama Y, Umeda M and Miyazaki K: Induction of 103-kDa gelatinase/type IV collagenase by acidic culture conditions in mouse metastatic melanoma cell lines. J Biol Chem. 267:11424–11430. 1992.PubMed/NCBI

4 

Kato Y, Ozono S, Shuin T and Miyazaki K: Slow induction of gelatinase B mRNA by acidic culture conditions in mouse meta-static melanoma cells. Cell Biol Int. 20:375–377. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Martínez-Zaguilán R, Seftor EA, Seftor RE, Chu YW, Gillies RJ and Hendrix MJ: Acidic pH enhances the invasive behavior of human melanoma cells. Clin Exp Metastasis. 14:176–186. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Lora-Michiels M, Yu D, Sanders L, Poulson JM, Azuma C, Case B, Vujaskovic Z, Thrall DE, Charles HC and Dewhirst MW: Extracellular pH and P-31 magnetic resonance spectroscopic variables are related to outcome in canine soft tissue sarcomas treated with thermoradiotherapy. Clin Cancer Res. 12:5733–5740. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Gatenby RA, Gawlinski ET, Gmitro AF, Kaylor B and Gillies RJ: Acid-mediated tumor invasion: A multidisciplinary study. Cancer Res. 66:5216–5223. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Moellering RE, Black KC, Krishnamurty C, Baggett BK, Stafford P, Rain M, Gatenby RA and Gillies RJ: Acid treatment of melanoma cells selects for invasive phenotypes. Clin Exp Metastasis. 25:411–425. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Robey IF, Baggett BK, Kirkpatrick ND, Roe DJ, Dosescu J, Sloane BF, Hashim AI, Morse DL, Raghunand N, Gatenby RA, et al: Bicarbonate increases tumor pH and inhibits spontaneous metastases. Cancer Res. 69:2260–2268. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Suzuki A, Maeda T, Baba Y, Shimamura K and Kato Y: Acidic extracellular pH promotes epithelial mesenchymal transition in Lewis lung carcinoma model. Cancer Cell Int. 14:1292014. View Article : Google Scholar : PubMed/NCBI

11 

Curry JM, Sprandio J, Cognetti D, Luginbuhl A, Bar-ad V, Pribitkin E and Tuluc M: Tumor microenvironment in head and neck squamous cell carcinoma. Semin Oncol. 41:217–234. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Peppicelli S, Bianchini F, Torre E and Calorini L: Contribution of acidic melanoma cells undergoing epithelial-to-mesenchymal transition to aggressiveness of non-acidic melanoma cells. Clin Exp Metastasis. 31:423–433. 2014. View Article : Google Scholar

13 

Shankar J and Nabi IR: Actin cytoskeleton regulation of epithelial mesenchymal transition in metastatic cancer cells. PLoS One. 10:e01199542015. View Article : Google Scholar : PubMed/NCBI

14 

Dobrowolski JM and Sibley LD: Toxoplasma invasion of mammalian cells is powered by the actin cytoskeleton of the parasite. Cell. 84:933–939. 1996. View Article : Google Scholar : PubMed/NCBI

15 

Kato Y, Lambert CA, Colige AC, Mineur P, Noël A, Frankenne F, Foidart JM, Baba M, Hata R, Miyazaki K, et al: Acidic extra-cellular pH induces matrix metalloproteinase-9 expression in mouse metastatic melanoma cells through the phospholipase D-mitogen-activated protein kinase signaling. J Biol Chem. 280:10938–10944. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Kato Y, Ozawa S, Tsukuda M, Kubota E, Miyazaki K, St-Pierre Y and Hata R: Acidic extracellular pH increases calcium influx-triggered phospholipase D activity along with acidic sphingomyelinase activation to induce matrix metalloproteinase-9 expression in mouse metastatic melanoma. FEBS J. 274:3171–3183. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Xu L, Fukumura D and Jain RK: Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway: Mechanism of low pH-induced VEGF. J Biol Chem. 277:11368–11374. 2002. View Article : Google Scholar

18 

Chen Y, Kung HN, Chen CH, Huang SH, Chen KH and Wang SM: Acidic extracellular pH induces p120-catenin-mediated disruption of adherens junctions via the Src kinase-PKCδ pathway. FEBS Lett. 585:705–710. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Sarosi GA Jr, Jaiswal K, Herndon E, Lopez-Guzman C, Spechler SJ and Souza RF: Acid increases MAPK-mediated proliferation in Barrett's esophageal adenocarcinoma cells via intracellular acidification through a Cl/HCO3 exchanger. Am J Physiol Gastrointest Liver Physiol. 289:G991–G997. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Lee YJ, Lee DM and Lee SH: Production of Cyr61 protein is modulated by extracellular acidification and PI3K/Akt signaling in prostate carcinoma PC-3 cells. Food Chem Toxicol. 58:169–176. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Riemann A, Schneider B, Ihling A, Nowak M, Sauvant C, Thews O and Gekle M: Acidic environment leads to ROS-induced MAPK signaling in cancer cells. PLoS One. 6:e224452011. View Article : Google Scholar : PubMed/NCBI

22 

Mochimaru Y, Azuma M, Oshima N, Ichijo Y, Satou K, Matsuda K, Asaoka Y, Nishina H, Nakakura T, Mogi C, et al: Extracellular acidification activates ovarian cancer G-protein-coupled receptor 1 and GPR4 homologs of zebra fish. Biochem Biophys Res Commun. 457:493–499. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Kotake M, Sato K, Mogi C, Tobo M, Aoki H, Ishizuka T, Sunaga N, Imai H, Kaira K, Hisada T, et al: Acidic pH increases cGMP accumulation through the OGR1/phospholipase C/Ca2+/neuronal NOS pathway in N1E-115 neuronal cells. Cell Signal. 26:2326–2332. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Murata N, Mogi C, Tobo M, Nakakura T, Sato K, Tomura H and Okajima F: Inhibition of superoxide anion production by extra-cellular acidification in neutrophils. Cell Immunol. 259:21–26. 2009. View Article : Google Scholar

25 

Mogi C, Tobo M, Tomura H, Murata N, He XD, Sato K, Kimura T, Ishizuka T, Sasaki T, Sato T, et al: Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J Immunol. 182:3243–3251. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Bruntz RC, Lindsley CW and Brown HA: Phospholipase D signaling pathways and phosphatidic acid as therapeutic targets in cancer. Pharmacol Rev. 66:1033–1079. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Colley WC, Sung TC, Roll R, Jenco J, Hammond SM, Altshuller Y, Bar-Sagi D, Morris AJ and Frohman MA: Phospholipase D2, a distinct phospholipase D isoform with novel regulatory properties that provokes cytoskeletal reorganization. Curr Biol. 7:191–201. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Riebeling C, Müller C and Geilen CC: Expression and regulation of phospholipase D isoenzymes in human melanoma cells and primary melanocytes. Melanoma Res. 13:555–562. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Saito M, Iwadate M, Higashimoto M, Ono K, Takebayashi Y and Takenoshita S: Expression of phospholipase D2 in human colorectal carcinoma. Oncol Rep. 18:1329–1334. 2007.PubMed/NCBI

30 

Maeda T, Suzuki A, Yuzawa S, Baba Y, Kimura Y and Kato Y: Mineral trioxide aggregate induces osteoblastogenesis via Atf6. Bone Rep. 2:36–43. 2015. View Article : Google Scholar

31 

Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA and Zhang F: Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 8:2281–2308. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Xiao Y, Liang L, Pan Y, Lian F, Li L, Lin H, Fu D, Fan J, Yang X, Sun L, et al: Inhibitory effects of simvastatin on migration and invasion of rheumatoid fibroblast-like synoviocytes by preventing geranylgeranylation of RhoA. Rheumatol Int. 33:389–399. 2013. View Article : Google Scholar

33 

Kuribara H, Tago K, Yokozeki T, Sasaki T, Takai Y, Morii N, Narumiya S, Katada T and Kanaho Y: Synergistic activation of rat brain phospholipase D by ADP-ribosylation factor and rhoA p21, and its inhibition by Clostridium botulinum C3 exoenzyme. J Biol Chem. 270:25667–25671. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Abécassis I, Olofsson B, Schmid M, Zalcman G and Karniguian A: RhoA induces MMP-9 expression at CD44 lamellipodial focal complexes and promotes HMEC-1 cell invasion. Exp Cell Res. 291:363–376. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Chandrasekar N, Jasti S, Alfred-Yung WK, Ali-Osman F, Dinh DH, Olivero WC, Gujrati M, Kyritsis AP, Nicolson GL, Rao JS, et al: Modulation of endothelial cell morphogenesis in vitro by MMP-9 during glial-endothelial cell interactions. Clin Exp Metastasis. 18:337–342. 2000. View Article : Google Scholar

36 

Bosco R, Melloni E, Celeghini C, Rimondi E, Vaccarezza M and Zauli G: Fine tuning of protein kinase C (PKC) isoforms in cancer: Shortening the distance from the laboratory to the bedside. Mini Rev Med Chem. 11:185–199. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Kang DW, Park MH, Lee YJ, Kim HS, Kwon TK, Park WS and Min S: Phorbol ester up-regulates phospholipase D1 but not phospholipase D2 expression through a PKC/Ras/ERK/NFkappaB-dependent pathway and enhances matrix metalloproteinase-9 secretion in colon cancer cells. J Biol Chem. 283:4094–4104. 2008. View Article : Google Scholar

38 

Semenza GL: Regulation of metabolism by hypoxia-inducible factor 1. Cold Spring Harb Symp Quant Biol. 76:347–353. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Lu H, Forbes RA and Verma A: Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem. 277:23111–23115. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Matsuoka J, Yashiro M, Doi Y, Fuyuhiro Y, Kato Y, Shinto O, Noda S, Kashiwagi S, Aomatsu N, Hirakawa T, et al: Hypoxia stimulates the EMT of gastric cancer cells through autocrine TGFβ signaling. PLoS One. 8:e623102013. View Article : Google Scholar

41 

Uttamsingh S, Bao X, Nguyen KT, Bhanot M, Gong J, Chan JL, Liu F, Chu TT and Wang LH: Synergistic effect between EGF and TGF-beta1 in inducing oncogenic properties of intestinal epithelial cells. Oncogene. 27:2626–2634. 2008. View Article : Google Scholar

42 

Shirakihara T, Kawasaki T, Fukagawa A, Semba K, Sakai R, Miyazono K, Miyazawa K and Saitoh M: Identification of integrin α3 as a molecular marker of cells undergoing epithelial-mesenchymal transition and of cancer cells with aggressive phenotypes. Cancer Sci. 104:1189–1197. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 48 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Maeda T, Yuzawa S, Suzuki A, Baba Y, Nishimura Y and Kato Y: RhoA mediates the expression of acidic extracellular pH-induced matrix metalloproteinase-9 mRNA through phospholipase D1 in mouse metastatic B16-BL6 melanoma cells. Int J Oncol 48: 1251-1257, 2016
APA
Maeda, T., Yuzawa, S., Suzuki, A., Baba, Y., Nishimura, Y., & Kato, Y. (2016). RhoA mediates the expression of acidic extracellular pH-induced matrix metalloproteinase-9 mRNA through phospholipase D1 in mouse metastatic B16-BL6 melanoma cells. International Journal of Oncology, 48, 1251-1257. https://doi.org/10.3892/ijo.2016.3322
MLA
Maeda, T., Yuzawa, S., Suzuki, A., Baba, Y., Nishimura, Y., Kato, Y."RhoA mediates the expression of acidic extracellular pH-induced matrix metalloproteinase-9 mRNA through phospholipase D1 in mouse metastatic B16-BL6 melanoma cells". International Journal of Oncology 48.3 (2016): 1251-1257.
Chicago
Maeda, T., Yuzawa, S., Suzuki, A., Baba, Y., Nishimura, Y., Kato, Y."RhoA mediates the expression of acidic extracellular pH-induced matrix metalloproteinase-9 mRNA through phospholipase D1 in mouse metastatic B16-BL6 melanoma cells". International Journal of Oncology 48, no. 3 (2016): 1251-1257. https://doi.org/10.3892/ijo.2016.3322