PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype

  • Authors:
    • Xiaoshan Cai
    • Hongguang Zhu
    • Ying Li
  • View Affiliations

  • Published online on: September 26, 2017     https://doi.org/10.3892/mmr.2017.7634
  • Pages: 8301-8306
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate protein kinase C ζ type (PKCζ), matrix metalloproteinase (MMP)‑2 and MMP‑9 expression in lung adenocarcinoma and to define their association with in vitro invasion and metastatic capacity. PKCζ, MMP‑2 and MMP‑9 expression was assessed by immunohistochemistry in 110 cases of lung adenocarcinoma. PKCζ small interfering (si)RNA was transfected into A549 cells, and western blotting was used to confirm PKCζ‑knockdown in transfected cells and to measure MMP‑2 and MMP‑9 levels. A Transwell invasion assay was used to detect in vitro invasive capacity. The rates of positive PKCζ, MMP‑2 and MMP‑9 staining in lung adenocarcinoma tissues were 52.73, 55.45 and 61.82%, respectively. PKCζ expression was increased in malignant tissues compared with adjacent normal lung tissues and was associated with lymph node metastasis (P<0.05), although it was not associated with any other clinicopathological parameters, including sex, age, tumor size, smoking status or distant metastases (all P>0.05). PKCζ, MMP‑2 and MMP‑9 expression was markedly decreased in siPKCζ‑treated A549 cells, which exhibited a significantly decreased invasive capacity in the Transwell invasion assay (P<0.05). In conclusion, PKCζ promoted lung adenocarcinoma invasion and metastasis, and its expression was associated with MMP‑2 and MMP‑9 expression. PKCζ may be a potential target for gene therapy in lung adenocarcinoma.

Introduction

Lung cancer is one of the commonest malignant carcinomas in the world, and its incidence is increasing in a number of countries. Worldwide, lung cancer is the leading cause of death from malignant tumor, accounting for ~30% of all cancer-associated mortality (1). Lung adenocarcinoma is the principal subtype of lung cancer, and metastasis is the leading cause of mortality in patients with lung adenocarcinoma.

The protein kinase C (PKC) family regulates cell growth, differentiation, metabolism and transcriptional activation. PKCs may affect the invasion and metastasis of tumor cells. PKCζ is a member of the PKC family that serves important roles in cell growth, metabolism and other associated signal transduction pathways (2,3). It has been established that PKCζ is a tumor suppressor for numerous types of human cancer (4). However, studies have additionally identified pro-oncogenic functions of PKCζ, although a complete understanding of the detailed molecular mechanisms is lacking (24). Additionally, it has been suggested that PKCζ may be involved in inflammatory responses to diverse stimuli in vitro and in vivo (58). However, PKCζ expression in lung adenocarcinoma and the possible outcomes of PKCζ signaling in the context of lung adenocarcinoma remain to be completely elucidated.

Matrix metalloproteinases (MMPs) are able to degrade the extracellular matrix and basement membrane, and serve important roles in promoting tumor invasion and metastasis (9,10). MMPs proteolytically activate or degrade a variety of non-matrix substrates, including cytokines and chemokines, exerting a regulatory function in inflammation and immunity (11). At present, the most well-established roles for MMPs are in colorectal carcinogenesis, wherein MMP-2 and MMP-9 have been implicated in colon cancer progression and metastasis (12). Studies into the role of metalloproteinases and their inhibitors in lung adenocarcinoma are limited, and the results have been varied (13,14).

Recently, studies have demonstrated that PKCs may promote the metastasis of tumor cells in breast cancer, glioma and other malignancies (15,16). PKCζ is able to activate the mitogen-activated protein kinase (MAPK) signaling pathway, which terminates with extracellular signal-regulated kinase (ERK) phosphorylation and consequent promotion of MMP-2 and MMP-9 secretion, which may facilitate invasion and metastasis (17,18). However, there have been few studies focusing on lung adenocarcinoma, and whether PKCζ may mediate the invasion and metastasis of lung adenocarcinoma by regulating MMP-2 and MMP-9 secretion remains unknown.

In the present study, PKCζ, MMP-2 and MMP-9 expression was assessed in lung adenocarcinoma and adjacent normal lung tissues using immunohistochemistry, and associations between their relative expression levels were analyzed. PKCζ was knocked down in the lung adenocarcinoma cell line A549, and invasive capacity, and MMP-2 and MMP-9 expression were observed, in order to examine the effects of PKCζ on invasion and metastasis in lung adenocarcinoma and to provide a novel method for the treatment of lung adenocarcinoma.

Materials and methods

Specimen collection

The present study included 110 patients with invasive lung adenocarcinoma (including all subtypes) who underwent histological diagnosis at the Second People's Hospital of Weifang (Weifang, China) between January 2012 and December 2014. Cases with preoperative therapy or a history of other known malignancies were excluded. Medical records were reviewed for clinicopathological features, including sex, age, tumor size, smoking status, lymph node metastasis, distant metastasis and pathological tumor, node, metastasis (pTNM) stage. Patients were divided into two groups by age (≤60 years and >60 years) and smoking status [smokers (>5 pack-year history) and non-smokers]. The pTNM stage was evaluated in accordance with the 7th lung cancer TNM classification and staging system (19). Adjacent normal lung tissue (taken 5 cm from the edge of the cancerous tissue) was used as the control.

Among the 110 lung adenocarcinoma patients: 59 were male and 51 were female; 66 were ≤60 years old and 44 were >60; and 44 were smokers and 66 were non-smokers. Regarding pTNM stage, 36 were stages I+II and 74 were stages III+IV. The present study was approved by the Institutional Ethics Committee of Second People's Hospital of Weifang, and written informed consent was obtained from all participants.

Reagents

Anti-PKCζ (TA312044), anti-MMP-2 (TA806846) and anti-MMP-9 (TA353338) antibodies were purchased from OriGene Technologies, Inc. (Beijing, China). Cell culture plates, Matrigel and Transwell chambers were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Lipofectamine 2000 transfection reagent was purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and RIPA lysis buffer was purchased from Beyotime Institute of Biotechnology (Haimen, China). The A549 cell line was purchased from the American Type Culture Collection (Manassas, VA, USA).

Immunohistochemistry

Immunohistochemical staining was performed on 4-µm, formalin-fixed, paraffin-embedded sections. PKCζ primary antibody was diluted 1:200 and manually applied to sections. All steps were performed in accordance with the manufacturer's protocol. MMP-2 and MMP-9 were not diluted for these experiments. PBS was used as the negative control. Staining intensity and the percentage of positive cells were evaluated under a microscope (BX53; Olympus Corporation, Tokyo, Japan) in 5 high-magnification fields of vision, and 100 cells were counted in each field. The specific methods were performed according to a previous study (18).

Cell culture

The lung adenocarcinoma cell line A549 was cultured in F12K culture medium (21127–022; Invitrogen; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (Gibco, Thermo Fisher Scientific, Inc.) at 37°C in 5% CO2. Experiments were performed on cells in the logarithmic growth phase. The cells were divided into 3 groups as follows: Control group, A549 cells without any treatment; Scr/A549 group, A549 cells transiently transfected with empty plasmid; and small interfering (si)PKCζ/A549 group, A549 cells transiently transfected with the PKCζ target fragment 5′-GAGGAAGTGAGAGACATGTGT-3′. A total of 0.4 µg plasmid/siRNA were transfected into the Scr/A549 group and the (si)PKCζ/A549 group. All the vectors were synthesized by Shanghai GeneChem Co., Ltd. (Shanghai, China). Transfections were performed using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The subsequent experimentation commenced 48 h following transfection.

Western blotting

For Western blot analysis, cells or tissues were directly lysed in RIPA lysis buffer. Aliquots of 50 µg protein were separated by 10% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked in 5% skimmed milk for 1 h at room temperature, and then immunoblotted using the appropriate primary antibodies at 4°C overnight and the HRP conjugated secondary antibodies at 37°C for 2 h. They were visualized by using enhanced chemiluminescence reagents ECL (Pierce; Thermo Fisher Scientific, Inc.). Western blot data in the present study are representative from three independent experiments. The intensities of bands in western blots were quantified by densitometry analysis using AlphaImager HP (version 3.4.0; ProteinSimple, San Leandro, CA, USA) and NIH ImageJ software (version 1.44; National Institutes of Health, Bethesda, MD, USA). The following commercial antibodies were used in this study: PKCζ (TA312044; 1:1,000), MMP-2 (TA806846; 1:1,000) and MMP-9 (TA353338; 1:1,000) (all from OriGene Technologies, Inc.), β-actin (4970; 1:1,000) and HRP-linked anti-rabbit IgG antibody (7074; 1:2,000) (both from Cell Signaling Technology, Inc., Danvers, MA, USA).

Transwell invasion assay

Matrigel was added to the top chamber of a Transwell system to form the matrix layer. To this matrix was added 100 µl (1×105) Scr/A549 or siPKCζ/A549 cells; epidermal growth factor was added into the lower chamber (500 µl/well). The Transwell device was placed in an incubator (37°C; 5% CO2) for 24 h. Following incubation, invaded cells were fixed for 1 min in precooled methanol and Giemsa stained for 30 min at room temperature. All experiments were repeated at least three times. The number of invading cells was counted under a microscope (IX71; Olympus Corporation) in five predetermined fields, total magnification, ×200, using CellSens Standard (version 1.7; Olympus Corporation).

Statistical analysis

All statistical analyses were performed using SPSS version 19.0 (IBM Corp., Armonk, NY, USA). Data are presented as the mean ± standard deviation. Statistical significance was evaluated using Student's t-test or χ2 test. P<0.05 was considered to indicate a statistically significant difference.

Results

Immunohistochemical findings

PKCζ was expressed in the cytoplasm of lung adenocarcinoma cells (Fig. 1A). Positive PKCζ staining was detected in 58 (52.73%) lung adenocarcinoma samples, while only 5 (4.50%) normal lung tissues exhibited weak positive staining. The difference was statistically significant (χ2=62.479; P<0.01). The rate of positive PKCζ staining in lung adenocarcinomas with lymph node metastases (64.30%) was increased compared with non-metastatic samples (45.60%) (P=0.017). The differences among other clinicopathological parameters were not significant (Table I).

Table I.

Expression of PKCζ in lung adenocarcinoma and association with clinical pathological indices.

Table I.

Expression of PKCζ in lung adenocarcinoma and association with clinical pathological indices.

PKCζ

Clinical pathological indexCase no.PositiveNegativeχ2P-value
Gender
  Male5932270.1160.848
  Female512625
Age, years
  ≤606630363.5010.08
  >60442816
Diameter of tumor, cm
  ≤34017232.6380.116
  >3704129
Smoker
  Yes4421230.7360.439
  No663729
Metastasis of LN
  Yes3926134.7100.045
  No713239
Distant metastasis
  Yes4227153.6420.077
  No683137
TNM stage
  I+II3616201.4730.309
  III+IV744232

[i] LN, lymph node; TNM, tumor, node, metastasis; PKCζ, protein kinase C ζ type.

MMP-2 and MMP-9 were primarily expressed in the cytoplasm of lung adenocarcinomas (Fig. 1B and C); the rate of positive staining was 55.45 and 61.82%, respectively. PKCζ expression was associated with MMP-2 (P=0.012) and MMP-9 (P=0.006) expression in lung adenocarcinoma (Table II).

Table II.

Association between PKCζ, MMP-2 and MMP-9 expression in lung adenocarcinoma.

Table II.

Association between PKCζ, MMP-2 and MMP-9 expression in lung adenocarcinoma.

PKCζ

GroupCase no.PositiveNegativeχ2P-value
MMP-2
  Positive6139226.9000.012
  Negative491930
MMP-9
  Positive6843257.8890.006
  Negative421527

[i] PKCζ, protein kinase C ζ type; MMP, matrix metalloproteinase.

Western blot analysis results

PKCζ expression siPKCζ/A549 cells was markedly decreased compared with Scr/A549 cells, confirming that the reagent successfully disrupted the expression of the target gene (Fig. 2). In addition, MMP-2 and MMP-9 protein expression in siPKCζ/A549 cells was markedly decreased compared with Scr/A549 cells (Fig. 2).

Transwell invasion assay findings

Fewer siPKCζ/A549 cells invaded through the membrane and into the bottom chamber compared with Scr/A549 cells (P<0.05), suggesting that PKCζ downregulation was able to decreased the invasive ability of lung adenocarcinoma cells (Fig. 3).

Discussion

In the present study, it was observed that positive PKCζ expression in lung adenocarcinoma was associated with lymph node metastasis, and MMP-2 and MMP-9 expression. It was additionally observed that MMP-2 and MMP-9 expression was decreased in A549 cells following PKCζ knockdown by siRNA, which weakened the invasive ability of the cells in vitro.

PKCs are lipid-dependent serine/threonine protein kinases present in mammalian cells that serve important roles in growth, metabolism, proliferation and cytoskeletal remodeling. PKCs are also important intracellular signaling molecules that have been demonstrated to act as oncogenes and tumor suppressors, depending on the cellular context and upon which protein adaptors interact with which PKC isoforms (2023). PKC isozymes comprise three classes: Conventional (cPKC, α, β and γ), novel (nPKC, δ, ε, η and θ) and atypical (aPKC, ζ and ι). Different PKC isotypes are known to serve distinct regulatory roles. PKCζ is an important subtype of atypical PKCs that is involved in numerous signal transduction pathways.

PKC family proteins have been intensively studied due to their association with cancer. Previous studies have demonstrated that PKCζ may promote tumor cell chemotaxis in glioma, liver cancer and breast cancer, thus promoting cancer cell invasion and metastasis (16,24). When PKCζ is activated, it may phosphorylate Lim domain kinase 1 and cofilin, promoting F-actin depolymerization and polymerization, respectively, which affects the cytoskeleton structure and inhibits cancer cell chemotaxis and migration. Moreover, PKCζ is able to activate integrin-β1, which enhances adhesion between cells, activates the MAPK pathway, and promotes vascular endothelial growth factor (VEGF) expression and angiogenesis, which may consequently promote tumor invasion and metastasis (17,18).

Ma et al (25) observed that PKCζ is involved in lung cancer cell adhesion and chemotaxis, and thus may affect the invasion and metastasis of lung cancer. In the present study, it was demonstrated that the rate of positive PKCζ staining in patient-derived lung adenocarcinoma paraffin sections assayed by immunohistochemistry was significantly increased compared with adjacent tissues, and that PKCζ expression was associated with lymph node metastasis. This result also suggested that PKCζ affected the invasion and metastasis of lung adenocarcinoma. In vitro Transwell invasion experiments using A549 lung adenocarcinoma cells further confirmed that reducing PKCζ inhibited the invasive capacity of tumor cells. Therefore, the results of the present study demonstrated that PKCζ was able to promote the invasion and metastasis of lung adenocarcinoma through in vitro and in vivo methods.

MMPs are a family of Zn2+-dependent endopeptidases that are able to degrade the extracellular matrix and basement membrane, and serve an important role in physiological and pathological processes. They have been regarded as critical factors that promote tumor cell invasion. MMP-2 and MMP-9 are the most important enzymes for type IV collagen degradation, and serve important roles in tumor angiogenesis, invasion and metastasis (26,27). The mechanism underlying this effect involves the increase of VEGF secretion from tumor cells induced by MMP-2 and MMP-9, promoting invasion and metastasis, which is dependent on MAPK activation and ERK phosphorylation. In addition, MMP-9 expression is known to cause emphysema in chronic obstructive pulmonary disorder and angiogenesis/metastasis in lung cancer (28).

Studies have demonstrated that MMP-2 and MMP-9 expression in non-small cell lung cancer is significantly increased compared with normal tissue adjacent to the cancer, and that their expression levels are associated with pathological grading and staging, invasion and metastasis (20,29). PKCζ was able to activate MAPK and the MAPK signaling pathway, and promote VEGF overexpression, angiogenesis, tumor invasion and metastasis (17). In the present study, it was observed that PKCζ expression was associated with the expression of MMP-2 and MMP-9 in lung adenocarcinoma, using immunohistochemical detection. By decreasing the expression of PKCζ in A549 cells, the invasiveness of siPKCζ/A549 cells decreased significantly; decreased PKCζ expression coincided with reduced secretion of MMP-2 and MMP-9. The above results suggested that the PKCζ may promote lung cancer invasion and metastasis by affecting MMP-2 and MMP-9 secretion in lung adenocarcinoma cells.

In conclusion, PKCζ expression was associated with the invasion and metastasis of lung adenocarcinoma, making PKCζ a potential target for gene therapy in lung cancer and providing a theoretical basis for enhancing the survival rate of patients with lung adenocarcinoma. PKCζ, MMP-2 and MMP-9 synergistically promoted lung cancer invasion and metastasis, although the specific mechanism remains unclear and requires further research.

Acknowledgements

The present study was supported by the Program of Weifang Health Bureau in China (grant no. 2012012) and the Program of Bureau of Science and Technology in Weifang Kuiwen District in China (grant no. 201620).

References

1 

Lu J, Wang W, Xu M, Li Y, Chen C and Wang X: A global view of regulatory networks in lung cancer: An approach to understand homogeneity and heterogeneity. Semin Cancer Biol. 42:31–38. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Wu J, Zhang B, Wu M, Li H, Niu R, Ying G and Zhang N: Screening of a PKC zeta-specific kinase inhibitor PKCzI257.3 which inhibits EGF-induced breast cancer cell chemotaxis. Invest New Drugs. 28:268–275. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Butler AM, Scotti BML, Li S, Smith KE, Fields AP and Murray NR: Protein kinase C zeta regulates human pancreatic cancer cell transformed growth and invasion through a STAT3-dependent mechanism. PLoS One. 8:e720612013. View Article : Google Scholar : PubMed/NCBI

4 

Queisser MA, Dada LA, Deiss-Yehiely N, Angulo M, Zhou G, Kouri FM, Knab LM, Liu J, Stegh AH, DeCamp MM, et al: HOIL-1L functions as the PKCζ ubiquitin ligase to promote lung tumor growth. Am J Respir Crit Care Med. 190:688–698. 2014. View Article : Google Scholar : PubMed/NCBI

5 

LaVallie ER, Chockalingam PS, Collins-Racie LA, Freeman BA, Keohan CC, Leitges M, Dorner AJ, Morris EA, Majumdar MK and Arai M: Protein kinase Czeta is up-regulated in osteoarthritic cartilage and is required for activation of NF-kappaB by tumor necrosis factor and interleukin-1 in articular chondrocytes. J Biol Chem. 281:24124–24137. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Abdel-Halim M, Darwish SS, ElHady AK, Hoppstädter J, Abadi AH, Hartmann RW, Kiemer AK and Engel M: Pharmacological inhibition of protein kinase C (PKC)ζ downregulates the expression of cytokines involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). Eur J Pharm Sci. 93:405–409. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Diaz-Meco MT and Moscat J: The atypical PKCs in inflammation: NF-κB and beyond. Immunol Rev. 246:154–167. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Morin C, Fortin S and Rousseau E: Bronchial inflammation induced PKCζ over-expression: Involvement in mechanical properties of airway smooth muscle. Can J Physiol Pharmacol. 90:261–269. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Xu M, Wang HF and Zhang HZ: Expression of RECK and MMPs in hepatoblastoma and neuroblastoma and comparative analysis on the tumor metastasis. Asian Pac J Cancer Prev. 16:4007–4011. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Merdad A, Karim S, Schulten HJ, Dallol A, Buhmeida A, Al-Thubaity F, Gari MA, Chaudhary AG, Abuzenadah AM and Al-Qahtani MH: Expression of matrix metalloproteinases (MMPs) in primary human breast cancer: MMP-9 as a potential biomarker for cancer invasion and metastasis. Anticancer Res. 34:1355–1366. 2014.PubMed/NCBI

11 

Parks WC, Wilson CL and López-Boado YS: Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol. 4:617–629. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Babykutty S, Suboj P, Srinivas P, Nair AS, Chandramohan K and Gopala S: Insidious role of nitric oxide in migration/invasion of colon cancer cells by upregulating MMP-2/9 via activation of cGMP-PKG-ERK signaling pathways. Clin Exp Metastasis. 29:471–492. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Kim HIe, Lee HS, Kim TH, Lee JS, Lee ST and Lee SJ: Growth-stimulatory activity of TIMP-2 is mediated through c-Src activation followed by activation of FAK PI3-kinase/AKT, and ERK1/2 independent of MMP inhibition in lung adenocarcinoma cells. Oncotarget. 6:42905–42922. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Yamamura T, Nakanishi K, Hiroi S, Kumaki F, Sato H, Aida S and Kawai T: Expression of membrane-type-1-matrix metalloproteinase and metalloproteinase-2 in nonsmall cell lung carcinomas. Lung Cancer. 35:249–255. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Zhang F, Dong W, Zeng W, Zhang L, Zhang C, Qiu Y, Wang L, Yin X, Zhang C and Liang W: Naringenin prevents TGF-β1 secretion from breast cancer and suppresses pulmonary metastasis by inhibiting PKC activation. Breast Cancer Res. 18:382016. View Article : Google Scholar : PubMed/NCBI

16 

Paul A, Danley M, Saha B, Tawfik O and Paul S: PKCζ Promotes Breast Cancer Invasion by Regulating Expression of E-cadherin and Zonula Occludens-1 (ZO-1) via NFκB-p65. Sci Rep. 5:125202015. View Article : Google Scholar : PubMed/NCBI

17 

McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, et al: Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 1773:1263–1284. 2007. View Article : Google Scholar : PubMed/NCBI

18 

S LH Yang, Li W YL, Ni M ZL and Yin CaZB: Expression and correlation of PKCζ, MMP-2 and MMP-9 in breast cancer. Chin J Clin Exp Pathol. 30:958–962. 2014.

19 

Mirsadraee S, Oswal D, Alizadeh Y, Caulo A and van Beek E Jr: The 7th lung cancer TNM classification and staging system: Review of the changes and implications. World J Radiol. 4:128–134. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Rajagopalan S, Moyle MW, Joosten I and Long EO: DNA-PKcs controls an endosomal signaling pathway for a proinflammatory response by natural killer cells. Sci Signal. 3:ra142010. View Article : Google Scholar : PubMed/NCBI

21 

Lim PS, Sutton CR and Rao S: Protein kinase C in the immune system: From signalling to chromatin regulation. Immunology. 146:508–522. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Fields AP and Regala RP: Protein kinase C iota: Human oncogene, prognostic marker and therapeutic target. Pharmacol Res. 55:487–497. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Antal CE, Hudson AM, Kang E, Zanca C, Wirth C, Stephenson NL, Trotter EW, Gallegos LL, Miller CJ, Furnari FB, et al: Cancer-associated protein kinase C mutations reveal kinase's role as tumor suppressor. Cell. 160:489–502. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Sun R, Gao P, Chen L, Ma D, Wang J, Oppenheim JJ and Zhang N: Protein kinase C zeta is required for epidermal growth factor-induced chemotaxis of human breast cancer cells. Cancer Res. 65:1433–1441. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Ma NZF, Guo HZX, Cao SNR and Zhang N: Correlation between PKCζ expression and invasion, metastasis and prognosis of adenoearcinoma of the lung. Chin J Clin Oncol. 37:557–560. 2010.

26 

Kuo HY, Huang YS, Tseng CH, Chen YC, Chang YW, Shih HM and Wu CW: PML represses lung cancer metastasis by suppressing the nuclear EGFR-mediated transcriptional activation of MMP2. Cell Cycle. 13:3132–3142. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Yu Y, Ding Z, Jian H, Shen L, Zhu L and Lu S: Prognostic value of MMP9 activity level in resected stage I B lung adenocarcinoma. Cancer Med. 5:2323–2331. 2016. View Article : Google Scholar : PubMed/NCBI

28 

King PT: Inflammation in chronic obstructive pulmonary disease and its role in cardiovascular disease and lung cancer. Clin Transl Med. 4:682015. View Article : Google Scholar : PubMed/NCBI

29 

González-Arriaga P, Pascual T, García-Alvarez A, Fernández-Somoano A, López-Cima MF and Tardón A: Genetic polymorphisms in MMP 2, 9 and 3 genes modify lung cancer risk and survival. BMC Cancer. 12:1212012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

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
Cai X, Zhu H and Li Y: PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype. Mol Med Rep 16: 8301-8306, 2017
APA
Cai, X., Zhu, H., & Li, Y. (2017). PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype. Molecular Medicine Reports, 16, 8301-8306. https://doi.org/10.3892/mmr.2017.7634
MLA
Cai, X., Zhu, H., Li, Y."PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype". Molecular Medicine Reports 16.6 (2017): 8301-8306.
Chicago
Cai, X., Zhu, H., Li, Y."PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype". Molecular Medicine Reports 16, no. 6 (2017): 8301-8306. https://doi.org/10.3892/mmr.2017.7634