Skip to main content
Medical Science Monitor: International Medical Journal of Experimental and Clinical Research logoLink to Medical Science Monitor: International Medical Journal of Experimental and Clinical Research
. 2018 Apr 4;24:1997–2002. doi: 10.12659/MSM.905892

Cyclic Guanosine Monophosphate (cGMP)-Dependent Protein Kinase II Blocks Epidermal Growth Factor (EGF)/Epidermal Growth Factor Receptor (EGFR)-Induced Biological Effects on Osteosarcoma Cells

Dapeng Li 1,A,E,G,*, Ye Hua 1,D,E,*, Lu Jiang 2,C,F, Yonghui Huang 1,B,D, Jiawei Yue 1,C,E, Yan Wu 2,A,E,G,, Yongchang Chen 2,D,F
PMCID: PMC5900801  PMID: 29617357

Abstract

Background

The present work was performed to detect the potential inhibitory effect of cyclic guanosine monophosphate (cGMP)-dependent protein kinase II (PKG II) on epidermal growth factor (EGF) receptor-induced biological activity and related signal cascades in osteosarcoma cells.

Material/Methods

We transfected the osteosarcoma MG-63 cell line with an adenoviral vector encoding PKG II cDNA (Ad-PKGII) and incubated the transfected cells with 250 μM 8-pCPT-cGMP to activate the PKG II. We stimulated the MG-63 cells with100 ng/ml EGF, and then detected their proliferation using a CCK-8 assay. Transwell assay was used to examine MG-63 cell migration; and Western blot analysis was used to detect expression of matrix metalloproteinase 9 (MMP-9) and activation of ERK and Akt.

Results

Stimulating cells by 100 ng/ml EGF promoted MG-63 cell proliferation and migration, ERK and Akt phosphorylation, and MMP-9 expression. These effects of EGF were inhibited in MG-63 cells infected with Ad-PKGII and incubated with 8-pCPT-cGMP.

Conclusions

Our results demonstrate that Ad-PKGII infection significantly inhibited EGF-induced proliferation and migration, as well as the associated-signal cascades; which indicates that PKG II might be a potential anti-cancer factor.

MeSH Keywords: Cell Proliferation; Cyclic GMP-Dependent Protein Kinase Type II; Epidermal Growth Factor; Osteosarcoma; Receptor, Epidermal Growth Factor; Transcellular Cell Migration

Background

Osteosarcoma (OS) is the third most common malignant tumor in adolescents, and is the most common primary bone malignant tumor [1]. OS has been proposed to originate from mesenchymal stem cells or osteoblasts, based on different clinical and experimental data [2]. OS occurs predominantly in the long bones, specifically in the metaphyseal areas [3], and exhibits a high degree of malignancy, rapid growth, and aggressive invasion. Hematogenous metastasis occurs early, with high incidence of lung metastasis [4]. The main clinical treatment of OS is definitive surgery with radiotherapy and chemotherapy [5]. However, the mortality rate remains high, especially in patients with metastatic disease.

Epidermal growth factor receptor (EGFR) is a membrane surface receptor with tyrosine kinase activity. EGF binds to EGFR and causes EGFR-specific tyrosine residue phosphorylation, thereby leading to activation of signal transduction pathways, including MAPK/ERK and PI3K/Akt pathways [68]. EGFR is highly expressed in a variety of human malignancies [9,10]. In recent years, important advances have been made in the study of EGFR, including indications that EGFR-mediated signal transduction is closely linked to the occurrence, development, and prognosis of malignant tumors [11,12]. EGFR expression in OS is well documented [1315], and correlates with worse prognosis. EGFR may play an important role in OS development; therefore, the EGFR pathway inhibition could represent a viable treatment option for OS [16].

PKG II, a serine/threonine protein kinase, exists in eukaryotic cells and is abundant in the brain and intestinal mucosa [17]. The function of PKG II was long unclear. However, in recent years, our studies [1822] have provided data showing that PKG II is closely associated with cell proliferation and migration, and the occurrence and development of tumors. The present work aimed to detect the inhibitory effect of PKG II on OS cell proliferation, migration, and other biological activities induced by activation of EGFR.

Material and Methods

Cell lines and reagents

The MG-63 human osteosarcoma cell line was provided by the Institute of Cell Biology (Shanghai, China). Adenoviral vectors containing cDNA encoding β-galactosidase (pAd-LacZ) and PKG II (pAd-PKG II) were kind gifts from Prof. Gerry and Prof. Renate, University of California, San Diego, CA, USA. Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were obtained from Gibco (Grand Island, NY, USA). The following primary antibodies were used in this study: anti-PKG II (Abgent, San Diego, CA, USA), goat anti-β-actin (Santa Cruz, CA, USA), mouse anti-phosphorylated EGFR (tyrosine 1068); (Cell Signaling, Danvers, MA, USA), rabbit anti-EGFR (Cell Signaling), rabbit anti-p-ERK (thr202/tyr204; Cell Signaling), and rabbit anti-ERK (Cell Signaling). HRP-conjugated secondary antibodies were obtained from Santa Cruz. EGF was purchased from Sigma (St. Louis, MO, USA); electrochemiluminescence reagents were obtained from Millipore (Billerica, MA, USA); and the Transwell system was purchased from Corning (Tewksbury, MA, USA).

Cell culture and adenoviral vector infection

MG-63 cells were cultured in DMEM with 10% FBS at 37°C and 5% CO2. The cells were sub-cultured when they reached 90% confluence. The cells were freshly plated at 70–80% confluence, and the infection was subsequently performed following our lab’s protocol [19].

Cell-counting kit (CCK)-8 assay

MG-63 cells were plated in 96-well plates (5×103 cells per well). After the cells were treated, 10 μl CCK-8 was added to every well, and then incubated for 2 h. The optical density at 450 nm was detected using a microplate reader. The data are presented as a percentage of the value obtained for the control group.

Cell migration assay

We plated 5×103 cells into the upper chamber of a Transwell system containing DMEM without FBS. Cells that migrated to the bottom side of the membrane were induced by DMEM containing 10% FBS in the lower chamber for 12 h at 37°C. The cells were then fixed with 4% paraformaldehyde solution for 30 min and stained with Giemsa solution for 10 min. The stained cells were examined with a microscope. The cells that migrated were counted in 5 randomly selected fields per insert.

Western blotting

MG-63 cells were lysed in radioimmunoprecipitation assay buffer supplemented with proteinase inhibitors. Equal amounts of total protein were loaded onto and separated on a 10% polyacrylamide gel by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Following electrophoresis, the proteins were transferred to a polyvinylidene difluoride membrane (PVDF), which was then blocked in 5% (w/v) non-fat milk and incubated with primary antibodies overnight at 4°C. The membrane was washed 3 times with Tris-buffered saline/Tween, then incubated with horseradish peroxidase-conjugated secondary antibody for 1 h at 37°C. Electrochemiluminescence was used to visualize positive bands on the membrane.

Statistical analysis

Statistical analyses were performed using the Statistical Package for Social Sciences System Version 14.0 (SPSS, Chicago, IL, USA). Data are shown as means ± standard deviation (SD). Statistical significance was assessed using ANOVA, and P<0.05 was considered statistical significance.

Results

PKG II inhibits EGF-induced cell proliferation

CCK-8 assays were performed to assess the effect of PKG II on EGF-stimulated proliferation of MG-63 cells. EGF treatment resulted in 75–90% more cells than control, which was abrogated by cGMP in Ad-PKG II transduced MG-63 cells (P<0.05 Ad-PKG II + cGMP + EGF vs. Ad-PKG II + EGF and Ad-PKG II + cGMP + EGF vs. Ad-LacZ + cGMP + EGF) but not in parental MG-63 or Ad-LacZ transduced MG-63 cells (Figure 1).

Figure 1.

Figure 1

PKG II inhibits EGF-induced cell proliferation. The MG-63 cells were infected with Ad-LacZ or Ad-PKG II for 24 h and then placed in serum-free overnight. Next, the cells were incubated with 8-pCPT-cGMP (250 μM, 1 h), and then treated with 100 ng/ml EGF for 24 h. Data are means ±SD from 3 independent experiments.

PKG II inhibits EGF-induced cell migration

To investigate whether PKG II influenced MG-63 migration, Transwell migration assay was performed. EGF-treated MG-63 cells had more than 40% more migrating cells, which was abolished by cGMP in PKG II-overexpressed MG-63 cells (P<0.05 Ad-PKG II + cGMP + EGF vs. Ad-PKG II + EGF and Ad-PKG II + cGMP + EGF vs. Ad-LacZ + cGMP + EGF) but not in control Ad-LacZ MG-63 cells (Figure 2).

Figure 2.

Figure 2

PKG II inhibits EGF-induced cell migration. The MG-63 cells were infected with Ad-LacZ or Ad-PKG II for 24 h and then seeded into the Transwell assay system. Treatment with 100 ng/ml EGF for 12 h can induce MG-63 cell migration (Ad-LacZ+EGF vs. Ad-LacZ, Ad-PKG II+EGF vs. Ad-PKG II). However, activated PKG II can inhibit EGF-induced cell migration (Ad-PKG II+cGMP+EGF vs. Ad-PKG II+EGF). Data are means ±SD from 3 independent experiments.

PKG II inhibits EGF-induced MMP-9 expression

Because EGFR-induced MMP-9 played significant roles in osteosarcoma invasion [23], we then evaluated the effect of PKG II on EGFR-induced MMP-9 expression in MG-63 cells. Western blot analysis demonstrated that treating both Ad-LacZ- and Ad-PKG II-infected cells with 100 ng/ml EGF increased the expression of MMP-9. Moreover, Ad-PKG II infection facilitated the inhibition of EGF-induced MMP-9 expression (Ad-PKG II + cGMP + EGF group vs. Ad-PKG II + EGF group) (Figure 3).

Figure 3.

Figure 3

PKG II inhibits expression of MMP-9 induced by EGF. The MG-63 cells were infected with Ad-LacZ or Ad-PKG II for 24 h and then placed on serum-free medium overnight. Next, the cells were incubated with 250 μM 8-pCPT-cGMP for 1 h, and then treated with 100 ng/ml EGF for 48 h. Data are means ±SD from 3 independent experiments.

PKG II inhibits EGF-induced tyrosine 1068 phosphorylation of EGFR and its downstream pathways

To determine how activated PKG II inhibited EGFR-induced proliferation and migration of MG-63 osteosarcoma cells, we performed Western blot analysis with specific antibodies to monitor the changes of the phosphorylation of EGFR, ERK, and Akt. Treating both Ad-LacZ- and Ad-PKG II-infected cells with 100 ng/ml EGF promoted EGFR, ERK, and Akt phosphorylation (Ad-LacZ + EGF group vs. Ad-LacZ group; Ad-PKG II + EGF group vs. Ad-PKG II group) (Figure 4). EGF-induced EGFR activation and phosphorylation of ERK and Akt were inhibited by cGMP only when PKG II was presented (Ad-PKG II + cGMP + EGF group vs. Ad-PKG II + EGF group) (Figure 4).

Figure 4.

Figure 4

PKG II inhibits activation of ERK and Akt induced by EGF. The MG-63 cells were infected with Ad-LacZ or Ad-PKG II for 24 h and then placed on serum-free medium overnight. Cells were incubated with 8-pCPT-cGMP (250 MM, 1 h), and then treated with 100 ng/ml EGF for 5 h. Data are means ±SD from 3 independent experiments.

Discussion

EGF treatment caused activation of EGFR and its downstream ERK and Akt, which in turn induced overexpression of MMP-9 and proliferation and migration of MG-63 osteosarcoma cells, which was abolished by cGMP-activated PKG II.

EGFR, a product of the proto-oncogene c-erbB1, is widely distributed on the surface of mammalian epithelial cells, fibroblasts, glial cells, keratinocytes, and other cell types. EGFR is a 170-kDa protein consisting of 3 domains: an extracellular domain that binds ligands, a transmembrane domain, and an intracellular domain with tyrosine kinase activity [24]. EGF binds with EGFR and induces tyrosine kinase phosphorylation of its intracellular domain [25], which then recruits effector proteins to its phosphorylated C-terminal domain and activates effector protein-mediated signal pathways that regulate proliferation, survival, differentiation, migration, invasion, and injury repair of cells [26]. EGFR is expressed in many malignant tumor cell types [2729]. Overexpression of EGFR is associated with metastasis, invasion, and poor prognosis of cancers [30]. The present study indicated that EGFR activation induced proliferation and migration of MG-63 osteosarcoma cells.

EGF, a ligand of EGFR, is an active polypeptide, that plays a critical role in intercellular interactions [7,31,32]. EGF binds with EGFR to induce cell growth, migration, and other biological activities [33]. EGF and EGFR also are involved in neoplastic transformation. Up-regulated EGF or EGFR has been detected in various malignancies, including OS [34].

MAPK/ERK and the PI3K/Akt pathway are the most important signal transduction pathways downstream of EGFR [35,36]. They are both related to cell proliferation, anti-apoptosis, angiogenesis, migration, adhesion, and invasion. Here, Western blot analyses revealed that EGF promoted the EGFR phosphorylation, and activated the ERK and Akt cascades.

Concluisons

PKG II is a major downstream effector of cGMP in chondrocytes [37], and PKG II-deficient mice show dwarfism due to impaired endochondral ossification. Our previous results indicated that PKG II may be a potential cancer suppressor by blocking proliferation and migration and inducing apoptosis of tumor cells [22,38]. In the present study, we demonstrated that activated PKG II inhibited the proliferation and migration of MG-63 cells induced by EGFR activation. Furthermore, we found that activated PKG II inhibited EGF-induced activation of the PI3K/Akt and ERK pathways in MG-63 cells, indicating that PKG II is a potential cancer suppressor of OS.

Footnotes

Source of support: This study was supported by the National Natural Science Foundation of China (81601931, 81201959), the Natural Science Foundation of Jiangsu Province (BK20150475), and the Youth Medical Talents Project of Jiangsu Province (QNRC2016844)

Conflict of interests

None.

References

  • 1.Avnet S, Di Pompo G, Chano T, et al. Cancer-associated mesenchymal stroma fosters the stemness of osteosarcoma cells in response to intratumoral acidosis via NF-kappaB activation. Int J Cancer. 2017;140(6):1331–45. doi: 10.1002/ijc.30540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lin YH, Jewell BE, Gingold J, et al. Osteosarcoma: Molecular pathogenesis and iPSC modeling. Trends in Mol Med. 2017;23(8):737–55. doi: 10.1016/j.molmed.2017.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kang S, Lee JS, Park J, et al. Staged lengthening and reconstruction for children with a leg-length discrepancy after excision of an osteosarcoma around the knee. Bone Joint J. 2017;99-B(3):401–8. doi: 10.1302/0301-620X.99B3.38018. [DOI] [PubMed] [Google Scholar]
  • 4.Paioli A, Rocca M, Cevolani L, et al. Osteosarcoma follow-up: Chest X-ray or computed tomography? Clin Sarcoma Res. 2017;7:3. doi: 10.1186/s13569-017-0067-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Anghelescu DL, Steen BD, Wu H, et al. Prospective study of neuropathic pain after definitive surgery for extremity osteosarcoma in a pediatric population. Pediatr Blood Cancer. 2017;64(3) doi: 10.1002/pbc.26162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jang JW, Song Y, Kim SH, et al. CD133 confers cancer stem-like cell properties by stabilizing EGFR-AKT signaling in hepatocellular carcinoma. Cancer Lett. 2017;389:1–10. doi: 10.1016/j.canlet.2016.12.023. [DOI] [PubMed] [Google Scholar]
  • 7.Xu L, Hou Y, Tu G, et al. Nuclear Drosha enhances cell invasion via an EGFR-ERK1/2-MMP7 signaling pathway induced by dysregulated miRNA-622/197 and their targets LAMC2 and CD82 in gastric cancer. Cell Death Dis. 2017;8(3):e2642. doi: 10.1038/cddis.2017.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kumari R, Chouhan S, Singh S, et al. Constitutively activated ERK sensitizes cancer cells to doxorubicin: Involvement of p53-EGFR-ERK pathway. J Biosci. 2017;42(1):31–41. doi: 10.1007/s12038-017-9667-8. [DOI] [PubMed] [Google Scholar]
  • 9.Zhou C, Zhu L, Ji J, et al. EGFR High Expression, but not KRAS status, predicts sensitivity of pancreatic cancer cells to nimotuzumab treatment in vivo. Curr Cancer Drug Targets. 2017;17(1):89–97. doi: 10.2174/1568009616666161013101657. [DOI] [PubMed] [Google Scholar]
  • 10.Canueto J, Cardenoso E, Garcia JL, et al. EGFR expression is associated with poor outcome in cutaneous squamous cell carcinoma. Br J Dermatol. 2017;176:1279–87. doi: 10.1111/bjd.14936. [DOI] [PubMed] [Google Scholar]
  • 11.Wang J, Wang B, Chu H, et al. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. Onco Targets Ther. 2016;9:3711–26. doi: 10.2147/OTT.S106399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yen HY, Liu YC, Chen NY, et al. Effect of sialylation on EGFR phosphorylation and resistance to tyrosine kinase inhibition. Proc Natl Acad Sci USA. 2015;112(22):6955–60. doi: 10.1073/pnas.1507329112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sevelda F, Mayr L, Kubista B, et al. EGFR is not a major driver for osteosarcoma cell growth in vitro but contributes to starvation and chemotherapy resistance. J Exp Clin Cancer Res. 2015;34:134. doi: 10.1186/s13046-015-0251-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lee JA, Ko Y, Kim DH, et al. Epidermal growth factor receptor: Is it a feasible target for the treatment of osteosarcoma? Cancer Res Treat. 2012;44(3):202–9. doi: 10.4143/crt.2012.44.3.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Selvarajah GT, Verheije MH, Kik M, et al. Expression of epidermal growth factor receptor in canine osteosarcoma: Association with clinicopathological parameters and prognosis. Vet J. 2012;193(2):412–19. doi: 10.1016/j.tvjl.2012.02.009. [DOI] [PubMed] [Google Scholar]
  • 16.Mantovani FB, Morrison JA, Mutsaers AJ. Effects of epidermal growth factor receptor kinase inhibition on radiation response in canine osteosarcoma cells. BMC Vet Res. 2016;12:82. doi: 10.1186/s12917-016-0707-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rangaswami H, Marathe N, Zhuang S, et al. Type II cGMP-dependent protein kinase mediates osteoblast mechanotransduction. J Biol Chem. 2009;284(22):14796–808. doi: 10.1074/jbc.M806486200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wu Y, Yao X, Zhu M, et al. PKG II reverses HGF-triggered cellular activities by phosphorylating serine 985 of c-Met in gastric cancer cells. Oncotarget. 2016;7(23):34190–200. doi: 10.18632/oncotarget.9074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jiang LU, Wu M, Wu Y, et al. Type II cyclic guanosine monophosphate-dependent protein kinase inhibits epidermal growth factor receptor activation in different cancer cell lines. Oncol Lett. 2015;9(6):2781–86. doi: 10.3892/ol.2015.3067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jiang L, Lan T, Chen Y, et al. PKG II inhibits EGF/EGFR-induced migration of gastric cancer cells. PloS One. 2013;8(4):e61674. doi: 10.1371/journal.pone.0061674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jiang L, Chen Y, Sang J, et al. Type II cGMP-dependent protein kinase inhibits activation of key members of the RTK family in gastric cancer cells. Biomed Rep. 2013;1(3):399–404. doi: 10.3892/br.2013.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wang Y, Chen Y, Li Y, et al. Type II cGMPdependent protein kinase inhibits RhoA activation in gastric cancer cells. Mol Med Rep. 2014;9(4):1444–52. doi: 10.3892/mmr.2014.1960. [DOI] [PubMed] [Google Scholar]
  • 23.Wang Q, Cai J, Wang J, et al. MiR-143 inhibits EGFR-signaling-dependent osteosarcoma invasion. Tumour Biol. 2014;35(12):12743–48. doi: 10.1007/s13277-014-2600-y. [DOI] [PubMed] [Google Scholar]
  • 24.Han YH, Abdul Hamid MR, Telisinghe PU, et al. Overexpression of EGFR protein in Bruneian lung cancer patients. Asian Pac J Cancer Prev. 2015;16(1):233–37. doi: 10.7314/apjcp.2015.16.1.233. [DOI] [PubMed] [Google Scholar]
  • 25.Guo T, Zhao L, Zhang Y, et al. A monoclonal antibody targeting the dimer interface of epidermal growth factor receptor (EGFR) Immunol Lett. 2016;180:39–45. doi: 10.1016/j.imlet.2016.10.011. [DOI] [PubMed] [Google Scholar]
  • 26.Han T, Guo M, Zhang T, et al. A novel glutaminase inhibitor-968 inhibits the migration and proliferation of non-small cell lung cancer cells by targeting EGFR/ERK signaling pathway. Oncotarget. 2017;8:28063–73. doi: 10.18632/oncotarget.14188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kaufman M, Mehrotra B, Limaye S, et al. EGFR expression in gallbladder carcinoma in North America. Int J Med Sci. 2008;5(5):285–91. doi: 10.7150/ijms.5.285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lee CM, Shrieve DC, Zempolich KA, et al. Correlation between human epidermal growth factor receptor family (EGFR, HER2, HER3, HER4), phosphorylated Akt (P-Akt), and clinical outcomes after radiation therapy in carcinoma of the cervix. Gynecol Oncol. 2005;99(2):415–21. doi: 10.1016/j.ygyno.2005.05.045. [DOI] [PubMed] [Google Scholar]
  • 29.Nishimura Y, Ezawa R, Ishii J, et al. Affibody-displaying bio-nanocapsules effective in EGFR, typical biomarker, expressed in various cancer cells. Bioorg Med Chem Lett. 2017;27(2):336–41. doi: 10.1016/j.bmcl.2016.11.038. [DOI] [PubMed] [Google Scholar]
  • 30.Lopez F, Llorente JL, Oviedo CM, et al. Gene amplification and protein overexpression of EGFR and ERBB2 in sinonasal squamous cell carcinoma. Cancer. 2012;118(7):1818–26. doi: 10.1002/cncr.26451. [DOI] [PubMed] [Google Scholar]
  • 31.Ruan L, Wang GL, Chen Y, et al. Identification of tyrosine phosphoproteins in signaling pathway triggered TGF-a by using functional proteomics technology. Med Oncol. 2010;27(4):1407–14. doi: 10.1007/s12032-009-9394-6. [DOI] [PubMed] [Google Scholar]
  • 32.Singh B, Carpenter G, Coffey RJ. EGF receptor ligands: Recent advances. F1000Res. 2016 Sep;8:5. doi: 10.12688/f1000research.9025.1. pii: F1000 Faculty Rev-2270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yuan JQ, Zhang YL, Li HT, et al. EGFR mutation testing in blood for guiding EGFR tyrosine kinase inhibitor treatment in patients with nonsmall cell lung cancer: A protocol for systematic review and meta-analysis. Medicine. 2017;96(7):e6151. doi: 10.1097/MD.0000000000006151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pahl JH, Ruslan SE, Buddingh EP, et al. Anti-EGFR antibody cetuximab enhances the cytolytic activity of natural killer cells toward osteosarcoma. Clin Cancer Res. 2012;18(2):432–41. doi: 10.1158/1078-0432.CCR-11-2277. [DOI] [PubMed] [Google Scholar]
  • 35.Mikula M, Skrzypczak M, Goryca K, et al. Genome-wide co-localization of active EGFR and downstream ERK pathway kinases mirrors mitogen-inducible RNA polymerase 2 genomic occupancy. Nucleic Acids Res. 2016;44(21):10150–64. doi: 10.1093/nar/gkw763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Liu B, Su Y, Li T, et al. CMTM7 knockdown increases tumorigenicity of human non-small cell lung cancer cells and EGFR-AKT signaling by reducing Rab5 activation. Oncotarget. 2015;6(38):41092–107. doi: 10.18632/oncotarget.5732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kamemura N, Murakami S, Komatsu H, et al. Type II cGMP-dependent protein kinase negatively regulates fibroblast growth factor signaling by phosphorylating Raf-1 at serine 43 in rat chondrosarcoma cells. Biochem Biophys Res Commun. 2017;483(1):82–87. doi: 10.1016/j.bbrc.2017.01.001. [DOI] [PubMed] [Google Scholar]
  • 38.Wu Y, Chen Y, Qu R, et al. Type II cGMP-dependent protein kinase inhibits EGF-triggered signal transduction of the MAPK/ERK-mediated pathway in gastric cancer cells. Oncol Rep. 2012;27(2):553–58. doi: 10.3892/or.2011.1507. [DOI] [PubMed] [Google Scholar]

Articles from Medical Science Monitor : International Medical Journal of Experimental and Clinical Research are provided here courtesy of International Scientific Information, Inc.

RESOURCES