Recombinant human decorin upregulates p57KIP2 expression in HepG2 hepatoma cell lines

  • Authors:
    • Abdu Selim Hamid
    • Jinran Li
    • Yali Wang
    • Xiaodong Wu
    • Hassan Abdellah Ahmed Ali
    • Zhenwu Du
    • Lihua Bo
    • Yucheng Zhang
    • Guizhen Zhang
  • View Affiliations

  • Published online on: June 6, 2013     https://doi.org/10.3892/mmr.2013.1510
  • Pages: 511-516
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Increasing the expression of cyclin-cyclin-dependent kinase inhibitors (cyclin-CDK) using small molecule inhibitors is a therapeutic strategy used to suppress cancer cell growth. Decorin (DCN), a functional component of the extracellular matrix, has been implicated in the suppression of cell proliferation by upregulating p21, a cyclin-CDK inhibitor. The purpose of this study was to examine the effect of recombinant decorin on the reactivation of p57KIP2, whose expression is silenced in hepatocellular carcinoma (HCC). Cell viability assay, cell cycle analysis, apoptosis assay and quantitative real time-PCR experiments were performed in three groups of HepG2 human cells: Uninfected HepG2 cells (control group), pcDNA3.1 vector-infected HepG2 cells (pcDNA3.1 group) and pcDNA3.1-DCN-infected HepG2 cells (pcDNA3.1‑DCN group). Our results revealed that recombinant human decorin inhibited cell proliferation, induced G0/G1 phase arrest and induced apoptosis by increasing the expression of caspase-3 in the pcDNA3.1-DCN group. The expression of p57KIP2 mRNA in the pcDNA3.1-DCN group was higher than in the pcDNA3.1 and control groups (P<0.05); however, there was no statistically significant difference between the control and pcDNA3.1 groups (P>0.05). In conclusion, recombinant human decorin reactivated p57KIP2 expression in HepG2 cells. As the expression level of p57KIP2 is downregulated in HCC, our finding may serve as a basis for the therapy and prognosis of HCC, although further studies are required.

Introduction

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. It is the third-leading cause of cancer-related mortality and the fifth most common type of cancer worldwide, with >600,000 cases diagnosed annually (13). HCC is normally diagnosed at an advanced stage and typically has a poor prognosis even following surgical resection and liver transplantation (4). Due to various etiologies, the prevention and treatment of HCC remains a challenge worldwide (5). HCC is more prevalent in developing countries with ~80% of the total incidence occurring in Asia and sub-Saharan Africa (6,7). The prevalence of HCC is increasing in western countries due to the increasing prevalence of hepatitis C virus (HCV) infection (810). Thus, with the global incidence of HCC on the rise, there have been increasing calls for the development of new and improved approaches for the treatment of HCC.

Decorin, a functional component of the extracellular matrix (ECM), has multiple biological functions, which include regulating matrix assembly and fibrillogenesis, and controlling cell proliferation (1113). Recently, an in vivo investigation demonstrated that decorin-null mice developed severe liver fibrosis with a significantly delayed healing process (14). Decorin is often downregulated in various types of cancer of epithelial origin (15). In addition to its important biological functions, decorin inhibits cancer growth in vitro and in vivo. The mechanism for the suppression of tumor growth is independent of the functional p53 tumor suppressor gene; however, requires p21 to be functional (16). Decorin causes a rapid phosphorylation of the epidermal growth factor receptor (EGFR), leading to the activation of mitogen-activated protein kinase and the upregulation of p21, a cyclin-dependent kinase (CDK) inhibitor, and ultimately growth arrest (17). In an in vivo experiment, mice that were decorin and p53 null (DCN/ and p53/) developed a more aggressive form of lymphoma than those that were only p53 null (12), suggesting that decorin deficiency is permissive for tumorigenesis. Another study by Bi et al, using a decorin-deficient mouse model, demonstrated that the intestinal tumorigenesis in DCN/ mice was linked to the downregulation of p21 and p27 (18), implying that the role of decorin in repressing tumorigenesis requires the upregulation of cyclin-dependent kinase inhibitors (CKIs).

Cyclin-CDK inhibitors include three proteins: p21CIP1, p27KIP1 and p57KIP2. CDK inhibitors regulate the cell cycle of mammalian cells by binding to cyclin-CDK complexes. In addition to cell cycle regulation, CKIs have CDK-independent functions, including regulating transcription, apoptosis, cell migration and the cytoskeleton (19). Unlike p21CIP1 and p27KIP1, p57KIP2 has a unique role in embryogenesis, in which the genetic deletion of p57KIP2 has been demonstrated to be lethal in p57KIP2 null mice (20,21). Relative to p21CIP1 and p27KIP1, p57KIP2 is the newest and least studied CIP/KIP member.

The p57KIP2 human gene is located on chromosome 11 at the 11p15.5 locus and encodes a protein 316 amino acids long. Structurally, p57KIP2 is almost identical to p27KIP1 and functionally, it leads to cell cycle arrest in the G1 phase. In addition, it has been reported that p27KIP1 and p57KIP2 play conducive roles in neuronal migration and may, when working together, coordinate the timing of neuronal differentiation, migration and, potentially, cell cycle arrest in neocortical development. p57KIP2 may serve as an important domain for protein interactions implicated in functions other than the CDK-inhibitory role (21). Additionally, p57KIP2, but not p21CIP1/WAF1 or p27KIP1, interacts in vivo and in vitro through its amino-terminal domain with transcription factor B-Myb. Mutations of p57KIP2 occur in Beckwith-Wiedemann syndrome and its reduced protein expression in breast, lung, liver, prostate, colorectal and bladder cancer is suggestive of its tumor suppressive properties (2226). There is mounting evidence that the p57KIP2 protein level is normally downregulated in these types of cancer through several mechanisms, including maternal-specific loss of heterozygosity (LOH), loss of imprinting and promoter methylation (19). A recent study revealed that the downregulation of p57KIP2 accelerates the growth and invasion of HCC (27), indicating that the upregulation of this tumor suppressor is essential for the prevention and therapy of HCC. Activating the cyclin-CDK inhibitors using a small molecule inhibitor to restore their regulatory role in the cell cycle, proliferation and differentiation is an attractive therapeutic strategy for cancer treatment. As the expression of p57KIP2 is downregulated in several types of cancer, it may have therapeutic and prognostic uses. Hence, an investigation into upregulating its expression is essential. In this study, we demonstrated that recombinant human decorin upregulated the expression of p57KIP2, a CDK inhibitor, in HepG2 cell lines.

Materials and methods

Cell culture

HepG2 cell lines were purchased from the American Type Culture Collection (Manassas, VA, USA); cells were grown in DMEM (Invitrogen Life Technologies, Carlsbad, CA, USA) plus 10% fetal bovine serum, and supplemented with 100 μ/ml penicillin and 100 mg/ml streptomycin (Sigma, St. Louis, MO, USA). Cells were maintained at 37°C in a humidified atmosphere containing 95% air and 5% CO2. Recombinant human decorin was synthesized in our laboratory (28).

Transient transfection of HepG2 cell lines

HepG2 cells were divided into three groups: The pcDNA3.1-DCN group, the pcDNA3.1 group and the untransfected group (control group). Transient transfection was performed using Lipotap liposomal reagent according to the manufacturer's instructions (Beyotime Institute of Biotechnology, Haimen, Jiangsu, China).

Cell viability assay

Cells were seeded in 96-well plates (104 cells per well). The cell proliferation assay was performed in all three HepG2 groups using the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] method (Sigma, 5 mg/ml). After 72 h of transfection, 20 μl (5 mg/ml) of MTT was added to each well for a 4-h incubation at 37°C. The supernatant was removed and 150 μl DMSO (Sigma) was added following 10 min of oscillation. The optical density (OD) value was determined with an ELISA machine (Biotek Synergy HT, Winooski, VT, USA) at 490 nm and the assays were performed in triplicate. In each group, the cells were analyzed and the data are presented as the means ± SD.

Cell cycle detection by flow cytometry

Cells were plated (3×105 cells per well) in 6-well plates, treated with pcDNA3.1-DCN, pcDNA3.1 and/or with neither pcDNA3.1 nor pcDNA3.1-DCN for 72 h and then trypsinized, washed using 1X buffer A, fixed with 70% ice-cold ethanol and incubated overnight. RNase A (up to 0.25 mg/ml) was added and the DNA was labeled with propidium iodide (PI; 5 μl; Becton-Dickinson, Franklin Lakes, NJ, USA). The cells were analyzed using flow cytometry (FC500, Beckman Coulter, Miami, FL, USA). The experiment was performed in triplicate.

Analysis of apoptosis by annexin V-FITC/PI assay

Analysis was conducted by cultivating cells in 6-well plates (3×105 cells per well) treated with pcDNA3.1-DCN, pcDNA3.1 and/or with neither pcDNA3.1 nor pcDNA3.1-DCN for 72 h and then trypsinized. Apoptosis was determined using the Annexin V-FITC Apoptosis kit (Becton-Dickinson). The cells were analyzed using flow cytometry in triplicate. The experiment was performed in triplicate.

RNA extraction

RNA was extracted using TRIzol RNA reagent (Sangon Biotech Co. Ltd., Shanghai, China) from three groups of HepG2 cells after 72 h transient transfection according to the manufacturer's instructions. Briefly, adhering to protocol, cells (3×105 cells) from each group were washed three times using 0.1M PBS, trypsinized, and then transferred to an RNase-free Eppendorf tube and centrifuged at a low speed. The supernatant was discarded, 0.5 ml of TRIzol reagent was added to lyse the cells and they were thoroughly mixed. Samples were allowed to sit at room temperature for 5 min and 0.2 ml of chloroform was added. The samples were mixed by hand for 15 sec and allowed to stand for 5–10 min at room temperature. The resulting mixture was centrifuged at 12,000 × g for 15 min at 4°C. The uppermost supernatant aqueous phase was transferred to a fresh RNase-free microcentrifuge tube, an equal amount of 70% ethanol was added, mixed and then transferred to a column. RPE solution (0.5 ml) was then added, incubated at room temperature and centrifuged. The purity and concentration of the RNA was checked using NanoDrop 1000 (Thermo Scientific, West Palm Beach, FL, USA) spectrophotometry and the OD 260/280 nm ratio was between 1.85–1.95 for each RNA sample group. The quality of the total RNA was verified by running an agarose gel and the total RNA was stored at −70°C.

Complementary DNA (cDNA) synthesis

Total RNA (1 μg) from each HepG2 cell group was reverse transcribed into complementary DNA (cDNA) using the First Strand cDNA Synthesis kit (GeneCopoeia, Rockville, MD, USA). Briefly, 1 μl random primer was added, and ddH2O (RNase/DNase free) was added up to 13 μl volume and the mixture was incubated for 10 min at 65°C, cooled and centrifuged. The final volume of 25 μl was kept at 37°C for 1 h followed by enzyme deactivation at 85°C for 5 min. The final volume was stored at −20°C.

Real time-PCR

The relative expression levels of mRNA p57KIP2 and caspase-3 from each group of cells were determined by quantitative PCR using the SYBR All-in-One qPCRMix (GeneCopoeia) with GADPH as a reference (Takara Bio, Inc., Shiga, Japan). Samples were run in separate tubes on an ABI Prism 7500 according to the manufacturer's instructions. In brief, the 25 μl samples were treated at 95°C for 10 min followed by 40 cycles of 95°C for 20 sec and 60°C for 30 sec, and a final extension of 5 min at 72°C. The real time-PCR (RT-PCR) primers were synthesized by Sangon Biotech Co. Ltd., and the sequences (5′ to 3′) were as follows: p57KIP2, forward: 5′-CAGAACCGCTGGGATTACGA-3′, reverse: 5′-CACCGAGTCGCTGTCCACTT-3′ and caspase-3, forward: 5′-GAGTGCTCGCAGCTCATACCT-3′, reverse: 5′-CCTCACGGCCTGGGATTT-3′. GAPDH was purchased from Takara Bio, Inc., and was used as an endogenous reference and its primer sequence was as follows: forward: 5′-TGCACCACCAACTGCTTAGC-3′ and reverse: 5′-GGCATGGACTGTGGTCATGAG-3′. The mRNA expression of p57KIP2 and caspase-3 was determined from each group of HepG2 cell cultures and performed in triplicate. Relative quantitation using the comparative CT method was performed for each sample group.

Statistical analysis

The Student's t-test was used to identify statistically significant differences among the samples for cell proliferation, cell cycle, apoptosis and quantitative PCR assays. The experiments were performed with three replicates and repeated three times. P<0.05 was considered to indicate a statistically significant result.

Results

Recombinant human decorin inhibits cell growth in HepG2 hepatoma cells

To investigate the cell proliferation inhibitory role of recombinant human decorin, an MTT assay was performed. As shown in Fig. 1, the control group cells and pcDNA3.1 group cells exhibited a higher OD after 72 h transient transfection, whereas in the pcDNA3.1-DCN group, cell proliferation was markedly inhibited at a statistically significant level. This result revealed that recombinant human decorin represses cell growth in HepG2 cells after 72 h of transient transfection and so all of the following experiments were conducted after culturing for 72 h.

Recombinant human decorin induces G0/G1 cell cycle arrest in HepG2 cells

Flow cytometry was used to investigate the cell cycle regulatory role of recombinant human decorin in HepG2 cells. Our result revealed that recombinant human decorin caused cell cycle arrest at the G0/G1 phase in HepG2 cells at 72 h following transient transfection (Fig. 2). More HepG2 cell cultures that were treated with pcDNA3.1-DCN accumulated in the G0/G1 phase compared with the control and pcDNA3.1 groups. The percentage of cells in the G0/G1 phase was 66.126±2.701, 57.116±1.421 and 55.323±1.641% in the pcDNA3.1-DCN, pcDNA3.1 and control groups, respectively (Table I).

Table I

Cell cycle distribution of pcDNA3.1-DCN, pcDNA3.1 and control group cells (n=3).

Table I

Cell cycle distribution of pcDNA3.1-DCN, pcDNA3.1 and control group cells (n=3).

Cell cycle distribution (%)

Group G0/G1S G2/M
Control55.323±1.64121.045±1.44223.632±1.815
pcDNA3.157.116±1.42121.284±1.73521.602±1.912
pcDNA3.1-DCN 66.126±2.701a22.108±1.915 11.776±1.043a

a P<0.05, compared with the control group.

Recombinant human decorin induces apoptosis in HepG2 cells

We examined the effect of recombinant human decorin on the induction of apoptosis and, as shown in Fig. 3, recombinant decorin induced apoptosis. The proportion of cells stained with annexin V and PI was higher in the pcDNA3.1-DCN group compared with the control and pcDNA3.1 groups (Table II). We measured caspase-3 expression by quantitative RT-PCR in the three human HepG2 groups. As illustrated in Fig. 4, caspase-3 expression was markedly increased in the pcDNA3.1-DCN group compared with the pcDNA-3.1 and control groups. There was no statistically significant difference in the expression of caspase-3 between the control group and pcDNA3.1 group (P>0.05).

Table II

Role of recombinant human decorin in inducing apoptosis in HepG2 group cells (n=3).

Table II

Role of recombinant human decorin in inducing apoptosis in HepG2 group cells (n=3).

GroupApoptosis (%)
Control3.1±0.3
pcDNA3.12.8±0.2
pcDNA3.1-DCN10.2±0.6a

a P<0.05, compared with the control group.

p57KIP2 expression in the three HepG2 cell groups

To determine the effect of human recombinant decorin on the relative expression of p57KIP2 mRNA between each cell group, quantitative RT-PCR was used. GAPDH was used as an endogenous reference for normalization. The p57KIP2 mRNA level was more highly expressed in the pcDNA3.1-DCN group than in the control and pcDNA3.1 groups.

Discussion

To the best of our knowledge, our findings demonstrate for the first time that recombinant human decorin upregulates p57KIP2 mRNA levels in HepG2 cells. By using quantitative RT-PCR, we examined the expression of p57KIP2 transcriptional mRNA levels and identified that expression was higher in recombinant human decorin-treated HepG2 cells compared with the control and pcDNA3.1 groups.

Earlier studies have revealed that decorin, a member of the family of small leucine-rich proteoglycans, inhibits the growth of cancer cells. For instance, Hu et al demonstrated that decorin suppressed prostate cancer cells through the EGFR and androgen receptor pathways (29). In addition, a study conducted by De Luca et al revealed that the inhibitory effect of decorin was correlated with the overexpression of p21, a CDK inhibitor (30).

Furthermore, previous studies have revealed that the decorin protein core causes cell death in both in vivo and in vitro experiments by activating the caspase-3 enzyme (31). In agreement with this, our study demonstrated that recombinant human decorin induced apoptosis via the activation of caspase-3 in the HepG2 cell line. In this study, the caspase-3 gene was significantly overexpressed in the recombinant human decorin-transfected HepG2 cells compared with the control and pcDNA3.1 groups. Caspase-3 is a crucial enzyme for apoptosis (32). Thus, one of the tumor suppressive properties of recombinant decorin in HepG2 cells promotes cell death via activation of the caspase-3 enzyme.

Growing evidence indicates that CDK inhibitors, including p57KIP2, are important in regulating cell proliferation and differentiation, cell cycle and cell apoptosis (19,20). Enhancing the expression of CKIs in order to suppress the activity of CDK in cancer has become a focus of cancer therapy research. Thus, reactivating the cyclin-CDK inhibitors using a small tumor inhibitor molecule, such as recombinant human decorin, provides an attractive therapeutic strategy for cancer treatment. We recently demonstrated that recombinant human decorin represses the growth of HepG2 cells by upregulating p21 via the p53-independent pathway (28). Ma and Cress demonstrated that p57KIP2 was significantly upregulated using small molecule CDK inhibitors, for instance BMS-387032 (SNS-032), in a breast cancer cell line (33). These findings led us to examine the role of recombinant decorin in the reactivation of p57KIP2, a family member of the CDK inhibitors in HepG2 culture cells. It is well documented that p57KIP2 is a potential tumor suppressor gene (34). However, the expression of this multifunctional CDK inhibitor is generally silenced in many types of cancer. A recent study by Guo et al demonstrated that the downregulation of p57KIP2 accelerates the growth and invasion of HCC (27). It has also been reported that p57KIP2 expression correlates with the malignant transformation of hepatocytes (35). Furthermore, another recent study revealed that the decreased expression of decorin and p57KIP2 correlated with poor survival rates and lymphatic metastasis in lung cancer patients (36). These studies suggest that p57KIP2 has a role in tumor inhibition. Notably, besides the possible function in tumorigenesis, the decreased expression of p57KIP2 may provide important prognostic implications for patients with ovarian, hepatocellular and colorectal cancer, and acute lymphoblastic leukemia (23,3739). Previously, it had been reported that the marked loss of p57KIP2 expression is a frequent event in HCC and so it may be important in the differentiation of HCC (25). Taken together, these observations imply that the reactivation of p57KIP2 suppresses the growth of cancer cells. Thus, restoring the normal function of p57KIP2 by increasing its expression using small molecule inhibitors may have therapeutic value. The results from our study demonstrated an increase in the expression of p57KIP2 in the pcDNA3.1-DCN HepG2-infected group compared with the control and pcDNA3.1 groups. Statistical analysis of the changes in p57KIP2 mRNA levels revealed a significant induction of p57KIP2 expression by recombinant decorin in HepG2 culture cells.

In this study, our results demonstrate that recombinant decorin induces the reactivation of p57KIP2 transcriptional mRNA in the HepG2 cell line. Therefore, it may suppress cell growth in the HepG2 cell line by upregulating p21 and also by reactivating p57KIP2, a cyclin-CDK inhibitor identified in pcDNA3.1-DCN-infected HepG2 cells.

In conclusion, our results demonstrated that recombinant human decorin increases the expression of p57KIP2 mRNA in the HepG2 cell line. Since p57 protein expression is silenced in various types of cancer, its reactivation may have a therapeutic use in clinical practice, in addition to other prognostic implications. In the present study, the statistical analysis of the changes in p57KIP2 mRNA levels revealed a significant induction of p57KIP2 expression by recombinant decorin in the HepG2 culture cells. However, for a more detailed understanding, particularly regarding the mechanism of recombinant decorin in the upregulation of p57KIP2 in the HepG2 cell line, further studies are required.

Acknowledgements

This study was supported by a grant from the Department of Public Health (2009Z080) in the Jilin Province, China.

References

1 

Colak D, Chishti MA, Al-Bakheet AB, et al: Integrative and comparative genomics analysis of early hepatocellular carcinoma differentiated from liver regeneration in young and old. Mol Cancer. 9:1462010. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar

3 

El-Serag HB, Davila JA, Petersen NJ and McGlynn KA: The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med. 139:817–823. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Zhang MF, Zhang ZY, Fu J, Yang YF and Yun JP: Correlation between expression of p53, p21/WAF1, and MDM2 proteins and their prognostic significance in primary hepatocellular carcinoma. J Transl Med. 7:1102009. View Article : Google Scholar

5 

Jain S, Singhal S, Lee P and Xu R: Molecular genetics of hepatocellular neoplasia. Am J Transl Res. 2:105–118. 2010.

6 

McGlynn KA and London WT: Epidemiology and natural history of hepatocellular carcinoma. Best Pract Res Clin Gastroenterol. 19:3–23. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Sherman M: Hepatocellular carcinoma: epidemiology, risk factors, and screening. Semin Liver Dis. 25:143–154. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Farazi PA and DePinho RA: Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer. 6:674–687. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Altekruse SF, McGlynn KA and Reichman ME: Hepatocellular carcinoma incidence, mortality, and survival trends in the United States from 1975 to 2005. J Clin Oncol. 27:1485–1491. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Nguyen MH, Whittemore AS, Garcia RT, et al: Role of ethnicity in risk for hepatocellular carcinoma in patients with chronic hepatitis C and cirrhosis. Clin Gastroenterol Hepatol. 2:820–824. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Iozzo RV: Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem. 67:609–652. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Iozzo RV, Moscatello DK, McQuillan DJ and Eichstetter I: Decorin is a biological ligand for the epidermal growth factor receptor. J Biol Chem. 274:4489–4492. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Reed CC and Iozzo RV: The role of decorin in collagen fibrillogenesis and skin homeostasis. Glycoconj J. 19:249–255. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Baghy K, Dezso K, László V, et al: Ablation of the decorin gene enhances experimental hepatic fibrosis and impairs hepatic healing in mice. Lab Invest. 91:439–451. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Gu Y, Zhang S, Wu Q, et al: Differential expression of decorin, EGFR and cyclin D1 during mammary gland carcinogenesis in TA2 mice with spontaneous breast cancer. J Exp Clin Cancer Res. 29:62010. View Article : Google Scholar : PubMed/NCBI

16 

Moscatello DK, Santra M, Mann DM, McQuillan DJ, Wong AJ and Iozzo RV: Decorin suppresses tumor cell growth by activating the epidermal growth factor receptor. J Clin Invest. 101:406–412. 1998. View Article : Google Scholar : PubMed/NCBI

17 

Csordás G, Santra M, Reed CC, et al: Sustained down-regulation of the epidermal growth factor receptor by decorin. A mechanism for controlling tumor growth in vivo. J Biol Chem. 275:32879–32887. 2000.PubMed/NCBI

18 

Bi X, Tong C, Dockendorff A, et al: Genetic deficiency of decorin causes intestinal tumor formation through disruption of intestinal cell maturation. Carcinogenesis. 29:1435–1440. 2008. View Article : Google Scholar

19 

Besson A, Dowdy SF and Roberts JM: CDK inhibitors: cell cycle regulators and beyond. Dev Cell. 14:159–169. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Kavanagh E and Joseph B: The hallmarks of CDKN1C (p57, KIP2) in cancer. Biochim Biophys Acta. 1816:50–56. 2011.PubMed/NCBI

21 

Pateras IS, Apostolopoulou K, Koutsami M, et al: Downregulation of the KIP family members p27(KIP1) and p57(KIP2) by SKP2 and the role of methylation in p57(KIP2) inactivation in nonsmall cell lung cancer. Int J Cancer. 119:2546–2556. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Oya M and Schulz WA: Decreased expression of p57(KIP2)mRNA in human bladder cancer. Br J Cancer. 83:626–631. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Li JQ, Wu F, Usuki H, et al: Loss of p57KIP2 is associated with colorectal carcinogenesis. Int J Oncol. 23:1537–1543. 2003.PubMed/NCBI

24 

Larson PS, Schlechter BL, King CL, et al: CDKN1C/p57kip2 is a candidate tumor suppressor gene in human breast cancer. BMC Cancer. 8:682008. View Article : Google Scholar : PubMed/NCBI

25 

Ito Y, Takeda T, Sakon M, Tsujimoto M, Monden M and Matsuura N: Expression of p57/Kip2 protein in hepatocellular carcinoma. Oncology. 61:221–225. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Atasoy P, Yilmaz E, Bozdogan O, Ayva S and Batislam E: Expression profile and prognostic importance in prostate lesions of the reverse transcriptase component of human telomerase (hTERT) and of cyclin-dependent kinase inhibitor p57 (p57kip2a). Int Urol Nephrol. 41:55–60. 2009. View Article : Google Scholar

27 

Guo H, Lv Y, Tian T, et al: Downregulation of p57 accelerates the growth and invasion of hepatocellular carcinoma. Carcinogenesis. 32:1897–1904. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Y, Wang Y, Du Z, et al: Recombinant human decorin suppresses liver HepG2 carcinoma cells by p21 upregulation. Onco Targets Ther. 5:143–152. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Hu Y, Sun H, Owens RT, et al: Decorin suppresses prostate tumor growth through inhibition of epidermal growth factor and androgen receptor pathways. Neoplasia. 11:1042–1053. 2009.PubMed/NCBI

30 

De Luca A, Santra M, Baldi A, Giordano A and Iozzo RV: Decorin-induced growth suppression is associated with up-regulation of p21, an inhibitor of cyclin-dependent kinases. J Biol Chem. 271:18961–18965. 1996.PubMed/NCBI

31 

Seidler DG, Goldoni S, Agnew C, et al: Decorin protein core inhibits in vivo cancer growth and metabolism by hindering epidermal growth factor receptor function and triggering apoptosis via caspase-3 activation. J Biol Chem. 281:26408–26418. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Lakhani SA, Masud A, Kuida K, et al: Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science. 311:847–851. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Ma Y and Cress WD: Transcriptional upregulation of p57 (Kip2) by the cyclin-dependent kinase inhibitor BMS-387032 is E2F dependent and serves as a negative feedback loop limiting cytotoxicity. Oncogene. 26:3532–3540. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Scandura JM, Boccuni P, Massagué J and Nimer SD: Transforming growth factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation. Proc Natl Acad Sci USA. 101:15231–15236. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Nan KJ, Guo H, Ruan ZP, Jing Z and Liu SX: Expression of p57(kip2) and its relationship with clinicopathology, PCNA and p53 in primary hepatocellular carcinoma. World J Gastroenterol. 11:1237–1240. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Biaoxue R, Xiguang C, Hua L, et al: Decreased expression of decorin and p57(KIP2) correlates with poor survival and lymphatic metastasis in lung cancer patients. Int J Biol Markers. 26:9–21. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Shen L, Toyota M, Kondo Y, et al: Aberrant DNA methylation of p57KIP2 identifies a cell-cycle regulatory pathway with prognostic impact in adult acute lymphocytic leukemia. Blood. 101:4131–4136. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Nakai S, Masaki T, Shiratori Y, et al: Expression of p57(KIP2) in hepatocellular carcinoma: relationship between tumor differentiation and patient survival. Int J Oncol. 20:769–775. 2002.PubMed/NCBI

39 

Sui L, Dong Y, Ohno M, Watanabe Y, Sugimoto K and Tokuda M: Expression of p57kip2 and its clinical relevance in epithelial ovarian tumors. Anticancer Res. 22:3191–3196. 2002.PubMed/NCBI

Related Articles

Journal Cover

August 2013
Volume 8 Issue 2

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
Hamid AS, Li J, Wang Y, Wu X, Ali HA, Du Z, Bo L, Zhang Y and Zhang G: Recombinant human decorin upregulates p57KIP2 expression in HepG2 hepatoma cell lines. Mol Med Rep 8: 511-516, 2013
APA
Hamid, A.S., Li, J., Wang, Y., Wu, X., Ali, H.A., Du, Z. ... Zhang, G. (2013). Recombinant human decorin upregulates p57KIP2 expression in HepG2 hepatoma cell lines. Molecular Medicine Reports, 8, 511-516. https://doi.org/10.3892/mmr.2013.1510
MLA
Hamid, A. S., Li, J., Wang, Y., Wu, X., Ali, H. A., Du, Z., Bo, L., Zhang, Y., Zhang, G."Recombinant human decorin upregulates p57KIP2 expression in HepG2 hepatoma cell lines". Molecular Medicine Reports 8.2 (2013): 511-516.
Chicago
Hamid, A. S., Li, J., Wang, Y., Wu, X., Ali, H. A., Du, Z., Bo, L., Zhang, Y., Zhang, G."Recombinant human decorin upregulates p57KIP2 expression in HepG2 hepatoma cell lines". Molecular Medicine Reports 8, no. 2 (2013): 511-516. https://doi.org/10.3892/mmr.2013.1510