FAM172A induces S phase arrest of HepG2 cells via Notch 3

Retraction in: /10.3892/or.2023.8554

  • Authors:
    • Zhiqiang Feng
    • Hongqi Li
    • Shunai Liu
    • Jun Cheng
    • Guoan Xiang
    • Jinqian Zhang
  • View Affiliations

  • Published online on: January 11, 2013     https://doi.org/10.3892/or.2013.2235
  • Pages: 1154-1160
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Our previous results revealed that FAM172A was significantly downregulated in liver tissue from hepatocellular carcinoma or cirrhotic patients. The present study was designed to elucidate the regulatory role of FAM172A in HepG2 cells. In order to determine the expression of the FAM172A protein, western blot analysis was performed. Confocal laser scanning technique was used to observe the localization of FAM172A in HepG2 cells. Surface plasmon resonance experiments were used to determine the binding activity of FAM172A and active single sugar and Ca2+. The cell cycle progression of HepG2 cells was assessed by flow cytometry. The FAM172A protein was localized in the endoplasmic reticulum of HepG2 cells. This protein was moderately expressed in normal liver tissue, but was significantly decreased in liver tissue of patients with chronic hepatitis B When co-cultured with the FAM172A recombinant protein, HepG2 cells exhibited complete cell cycle arrest in the S phase at a high concentration (100 ng/ml). Proliferation of HepG2 cells treated with the FAM172A recombinant protein was prominently inhibited compared with that of the control cells. Western blot analysis showed that upregulation of Notch 3 and cyclin E may be related with the cell cycle control. Our results indicate that FAM172A may be a novel tumor-suppressor gene, which plays an important role in cell cycle control and tumor cell proliferation. G1/S phase arrest may be mediated, at least partially, by the Notch 3 signaling pathway.

Introduction

Hepatocellular carcinoma (HCC) is the most common primary solid tumor of the liver. Numerous evidence indicates that hepatitis B virus (HBV) is one of the major etiological factors responsible for the development of HCC (1,2). Approximately 350 million people are chronically infected with HBV worldwide, among whom at least 200 million reside in China (3,4). Although great efforts have been carried out by numerous reseachers, to date, the molecular mechanism of hepatocarcinogenesis remains unknown. Identification of the ‘critical’ genes that play a pivotal role in the initiation and/or promotion of the development of HCC must be achieved (5).

The Notch pathway is implicated in many aspects of liver development and functions. Notch signaling acts as a molecular gate which is tightly associated with liver cell fate decisions (6). Jagged-1 and its receptor, Notch, are involved in the regulation of biliary epithelial growth and liver development (7,8). Mutation in the human Jagged 1 gene (MIM 601920) causes liver defects (9). Aberrant Jagged-1 expression has been observed in the liver during the development of cirrhosis (10). Recently, many investigations have demonstrated that the Notch pathway is involved in hepatocarcinogenesis. Although Notch 3 and 4 are not expressed in normal liver and in chronic hepatitis surrounding HCC, the abnormal accumulation of Notch 3 and 4 was observed (11). Although the Notch receptor is implicated in HCC, a detailed understanding of this signaling pathway in the process of hepatocarcinogenesis is not fully elucidated. Discovery of novel human proteins provides new opportunities for the development of drug therapies for the treatment of HCC for which there is still a demand (12).

In recent years, our results showed that one functionally unknown gene, FAM172A (C5orf21, NM_032042), was significantly downregulated in the tissues of patients with HCC or cirrhosis (13). Based on bioinformatic analysis, FAM172A encodes a protein as a precursor 416aa (48 kDa). The 1–18aa of the N-terminal is a signal peptide, and the 413–416aa in the C-terminal can prevent secretion from the endoplasmic reticulum (ER). The present investigation was designed to elucidate the regulatory role of FAM172A in HepG2 cells.

Materials and methods

The study protocol was approved by the Ethics Committees of the Air Force General Hospital of PLA and Beijing Ditan Hospital, Capital Medical University, China. The tissue samples were obtained from the Department of Pathology, Air Force General Hospital of PLA and Beijing Ditan Hospital, Capital Medical University.

Cell culture

Human L02, HepG2 and HepG2.2.15 cells were maintained in our laboratory. Cells were cultured in DMEM medium supplemented with 10% fetal bovine serum (FBS) in a 95% air, 5% CO2-humidified atmosphere at 37°C.

Preparation of the FAM172A recombinant protein and its polyclonal antibody

Preparation of the FAM172A recombinant protein and its polyclonal antibody was previously described (14). Briefly, FAM172A cDNA was amplified from FAM172A mRNA (PubMed: NM-032042), using the upstream primer (5′-ggtaccatgtctatttccttgagctc-3′) and the downstream primer (5′-aagcttcagctcttcgtgcttgatg-3′). The restriction sites, BamHI and HindIII, were incorporated into the primer sequences for cloning purposes. The PCR product was purified and cloned into pET-32a (+) expression vector [pET-32a (+) - FAM172A]. The BL21 E. coli cells transferred with pET-32a-FAM172A were induced. After purification, the FAM172A recombinant protein was used to inoculate rabbits and its polyclonal antibody was prepared.

Western blot analysis for detection of the histological expression of FAM172A

For determining the expression of FAM172A, Notch, and other proteins, western blot analysis was performed. Equal amounts of protein were added on 12% SDS-PAGE for electrophoresis, blotted onto PVDF membranes (Millipore, Billerica, MA, USA), and treated with anti-actin (1:500) (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-FAM172A (1:200), anti-GRp78 (1:500) (Abcam, UK), anti-Notch 1, 2, 3, and 4 (1:500) (Santa Cruz Biotechnology, Inc.) antibodies followed by goat anti-rabbit IgG (1:2,000) (Bio-Rad, USA). A quantitative measurement of the band intensity was performed using the GE Typhoon Trio (GE, USA).

Confocal laser scanning

Calreticulin (CALR) was cloned using forward primer (5′-ctcgagatggcgggatcc-3′) and reverse primer (5′-ggtaccggaaagaattttttggc-3′). The restriction sites, XhoI and KpnI, were incorporated into the primer sequences for cloning purposes. The PCR product was purified and cloned into the pDS-RED1-N1 expression vector (pDS-RED1-N1-CALR).

The pEGFP-C1-FAM172A and pDS-RED1-N1-CALR vectors were cotransfected into HepG2 cells. After transfection for 48 h, cells were treated with 4% paraformaldehyde for 10 min, washed 3 times with PBS and stained with 0.1 μg/ml 4′,6-diamidino-2-phenylindole (DAPI) for 30 min at 37°C. Then cells were imaged using an LSM510 microscope (Zeiss, Germany).

Surface plasmon resonance experiments

All experiments were performed at 25°C in HBS buffer (10 mM HEPES pH 7.4 containing 150 mM NaCl, 3 mM EDTA and 0.005% Surfactant P20) on a Biacore 3000 instrument, as previously described (14).

Cell count and cell cycle analysis

HepG2 cells were treated with different concentrations (0, 0.1, 1.0, 10 and 100 ng/ml) of FAM172A recombinant proteins. Cells were then trypsinized and washed gently with PBS, and then fixed in ice-cold 70% ethanol for at least 30 min. After fixation, cells were collected and stained with propidium iodide (PI) (5 mg/ml) for 30 min. The cells treated with PBS were used as the control group. Cells were assessed by flow cytometry (BD Biosciences, USA) and the results were analyzed with Modifit software.

Evaluation of endoplasmic reticulum stress

For determining the role of FAM172A in endoplasmic reticulum stress, we co-cultured HepG2 cells with tunicamycin (0.25, 0.5 and 1 μM) for 48 h. Total proteins were extracted from the separated HepG2 cells, respectively. Then 20 μg of protein was loaded onto each lane and was electrophoresed on 12% SDS-PAGE and transferred to PVDF membranes via a standard protocol. The membranes were probed for anti-GADD, anti-FAM172A and anti-GRp78 using the respective specific antibodies. β-actin was used as a loading control.

Statistical analysis

Results are presented as means ± SEM. Significance of the differences between means was assessed by one-way analysis of variance (ANOVA) or the two-tailed Student's t-test. P-value <0.05 was considered to indicate a statistically significant result. Unless stated otherwise, studies were performed on three independent occasions.

Results

FAM172A is localized in the endoplasmic reticulum and is moderately expressed in normal liver tissue

After HepG2 cells were co-transfection with plasmids pEGFP-C1-FAM172A and pDS-RED1-N1-CALR for 48 h, the confocal scanning demonstrated that the FAM172A recombinant protein was localization in the endoplasmic reticulum (Fig. 1B). Compared with normal liver tissues, FAM172A displayed almost no expression in the tissues of patients with chronic hepatitis B (Fig. 1B). In order to ascertain the role of FAM172A in the pathogenesis of the liver, we observed the specific expression of FAM172A protein in the human liver cell lines, L02, HepG2, and HepG2.2.15. The results demonstrated that FAM172A was strongly expressed in human hepatic normal cells (L02), moderately expressed in human hepatoma cells (HepG2), and weakly expressed in HepG2.2.15 cells (Fig. 1C).

Potent Ca2+ binding activity and downregulation of FAM172A during ER stress

Since the FAM172A protein is localized in the ER, we further determined whether this recombinant protein possesses nucleoside sugar and Ca2+ binding activity. At different concentrations (10−3, 10−2, 10−1 and 1 M) none of the active sugars, including UDP-GlcNAc, UDP-Gal, UDP-Glc, UDP-GalNAc, UDP-Fuc, GDP-Man and CMP-sialic acid (Sigma, USA), displayed any binding activity with this recombinant protein. However, the recombinant protein was able to bind with Ca2+ at concentrations of 10−3 M (169 RU), 10−2 M (350 RU), 10−1 M (451 RU) and 1 M (923 RU), respectively. The feature of binding between FAM172A and Ca2+ involved the rapid association to a steady-state response and rapid dissociation. According to Rmax, the binding ratio between FAM172A and Ca2+ was not 1:1, but appeared to be 1:n (Fig. 2A).

Based on FAM172A localization in the endoplasmic reticulum, we first speculated that FAM172A may be implicated in ER stress. Subsequently, ER stress was induced with tunicamycin. The expression of GADD, GRp78 and FAM172A was detected by western blot analysis. The results showed that the expression of GADD and GRp78 in cells treated with different concentrations of tunicamycin was higher than that of the control cells. In contrast to the markers of ER stress, GADD and GRp78, the expression of FAM172A was significantly downregulated in the cells treated with different concentrations of tunicamycin when compared with that of the control cells (Fig. 2B).

FAM172A causes S phase arrest of HepG2 cells and inhibits cell proliferation

To determine the effect of FAM172A on the cell cycle, we co-cultured HepG2 cells with different concentrations of the FAM172A recombinant protein. After 48 h, the total number of cells was counted under a microscope. Based on our results, FAM172A recombinant protein inhibited proliferation of HepG2 cells in a dose-dependent manner (Fig. 3A). Flow cytometric analysis also demonstrated that the percentage of cells in the S phase of the cell cycle was decreased as the concentration of the FAM172A recombinant protein increased. At the highest concentration of FAM172A recombinant protein (100 ng/ml), the percentage of HepG2 cells in the S phase was decreased to 2.27% (Fig. 4A-D). The results of the cytometric analysis showed that the FAM172A recombinant protein significantly suppressed HepG2 cell proliferation at a concentration >10 ng/ml.

Upregulation of Notch 3 and cyclin E is related to cell cycle control

We investigated the manner by which FAM172A regulates the cell cycle. Notch receptors are known to play a key role in ‘cell fate decision’. Thus, we examined whether FAM172A recombinant protein has any effects on Notch molecular expression. Subsequently, we co-cultured HepG2 cells with different concentrations (0, 0.1, 1.0, 10 and 100 ng/ml) of the FAM172A recombinant protein for 48 h. Cell proteins were extracted and assessed by western blot analysis. Blots were probed with anti-Notch 1, 2, 3, and 4 antibodies respectively. As shown in Fig. 3B, only the expression of Notch 3 was upregulated after recombinant protein treatment. However, no difference in expression of Notch 1, 2 and 4 in the HepG2 cells was noted after co-culture with different concentrations of the FAM172A recombinant protein for 48 h.

Since the cyclins control cell cycle transition, we aimed to ascertain which cyclin is implicated in the cell cycle control mediated by FAM172A. Therefore, after HepG2 cells were co-cultured with different concentrations (0, 0.1, 1.0, 10, and 100 ng/ml) of the FAM172A recombinant protein, the expression of cyclin A, B, D and E in the HepG2 cells was evaluated. Our results showed that only cyclin E was upregulated with increasing concentrations of FAM172A in the supernatant (Fig. 4E).

Discussion

FAM172A was cloned, the expression was assessed in vitro, and the polyclonal antibody was prepared. Our results showed that FAM172A may be implicated in the proliferation of hepatocytes. Our data revealed that FAM172A can arrest HepG2 cells in the S phase of the cell cycle at a higher concentration. This effect may be negatively related to the regulation of hepatocarcinogenesis. In particular, high levels of FAM172A recombinant protein or transfection with the FAM172A expression plasmid for 48 h, caused elimination of HepG2 cells in the S phase (data not show). The proliferation of HepG2 cells was also significantly inhibited at that concentration (>100 ng/ml) of the FAM172A recombinant protein.

We aimed to ascertain the signaling pathway involved in the cell cycle arrest after HepG2 cells were treated with FAM172A. We hypothesized that the Notch pathway may be involved in this process since Notch signaling acts as a molecular gate of hepatocytes that are tightly associated with liver cell fate decisions (6,9). However, based on aforementioned results, the expression of three genes in the HepG2 cells was significantly increased, i.e. Notch 3, NF-κB and cyclin E, after FAM172A recombinant protein treatment or transfection with the FAM172A expression plasmid.

The findings of endoplasmic reticulum (ER) stress induced by tunicamycin are in overall agreement with the observation of the clinical samples. Regardless of how FAM172A is involved in this process, we confirmed that a decrease in FAM172A exacerbated ER stress, as a number of Ca2+ were freed from FAM172A (16). Since ER stress plays a critical role in hepatocarcinogenesis (17,18), based on the above results, we could extrapolate that the decrease in FAM172A expression in hepatocytes may be involved in ER stress, ultimately resulting in hepatocarcinogenesis.

Based on our results, we first considered by which manner Notch 3, NF-κB, and cyclin E are harmonized with each other in cell cycle control. According to a previous report, all 4 Notch receptors are expressed in neoplastic cells of HCC, and are involved in the development of hepatocellular carcinoma (19), but only Notch 3 may be involved in controlling the cell differentiation of HCC (20). It seems that the Notch signaling pathway is activated and probably represents an important contributor to hepatocyte proliferation (21). Furthermore, the NF-κB pathway is an effector of gene expression downstream of the Notch signaling pathway (22,23). However, in most circumstances, the activation of NF-κB signaling also facilitates cell proliferation instead of differentiation. It is unclear what inhibited proliferation when HepG2 cells were exposed to a high concentration of FAM172A recombinant protein, and simultaneously, Notch 3 and NF-κB were significantly upregulated. Another investigation demonstrated that although cyclin E controls the transition of quiescent cells into the cell cycle, after partial hepatectomy, cyclin E2 (−/−) mice displayed accelerated and sustained DNA synthesis (24). Our results suggest that overexpression of cyclin E may also delay cell cycle progression from G1 phase to S phase. As a downstream signal molecule of the NF-κB pathway (25), cyclin E may play an important role in FAM172A-induced cell cycle arrest.

Herein, we propose a model to explain the FAM172A-dependent responses to Notch/NF-κB/cyclin E pathway activation in HepG2 cells (Fig. 5). Based on our results and other investigations, we considered that FAM172A may play an important role in (HepG2) G1/S cell cycle arrest via two pathways. First, when expression of FAM172A is increased, upregulation of Notch 3 expression occurs, and Notch 3 prompts the expression of NF-κB (26). NF-κB downregulates the expression of cyclin E (27). FAM172A may diminish the NF-κB inhibiting role, and G1/S phase arrest is induced by high expression of NF-κB or indirectly by cyclin E (28,29). Secondly, when expression of FAM172A is decreased, Ca2+ is released from the ER, and cdk2 is activated, prompting cell cycle progression into the S phase (30,31). Since Ca2+ plays a pivotal role in ER stress, the cell cycle control of FAM172A may also be mediated by ER stress (32).

Bioinformatics has demonstrated that this functional gene, FAM172A, is 456.69 kb, including 10 exons, belonging to the family of sequence similarity 172, member A. The 1–18aa of the N-terminal is a signal peptide, and the 413–416aa (HEEL) in the C-terminal prevents secretion from ER1. The precursor protein may be located in the endoplasmic reticulum (Fig. 1A-D), being further processed into a mature secreted isoform, 292aa (34 kDa). Consistent with the above information and our results, we speculate that G1/S arrest induced by FAM172A may be mediated by this secreted isoform. The results of the supernatant interchange confirmed this view. (Fig. 4). The 34-kDa isoform may be a new cytokine related with immunoregulation, since the 34-kDa isoform was also observed in spleen and thymus tissues (Fig. 1G).

In conclusion, our data indicate that FAM172A may be a new tumor-suppressor gene, which plays an important role in cell cycle control and tumor cell proliferation. Cell cycle arrest may be mediated, at least partially, by upregulated expression of Notch 3.

Acknowledgements

This research was supported by a grant from the National Natural Science Foundation of China (no. 30600524), and funding of the Capital Health Research and Development (2007-1017).

Abbreviations:

FAM172A

the protein coded by C5orf21

HCC

hepatocellular carcinoma

HBV

hepatitis B virus

FBS

fetal bovine serum

CALR

calreticulin

UDP-GlcNAc

UDP-acetylglucosamine

UDP-Gal

UDP-galactose

UDP-Glc

UDP-glucose

UDP-Fuc

UDP-fucose

GDP-Man

GDP-mannose

UDP-GalNAc

UDP-acetylgalactosamine

GADD

DNA damage-inducible transcription factor

GRp78

78-kDa glucose-regulated protein

ANOVA

one-way analysis of variance

HEEL

histidine-glutamic acid-glutamic acid-leucine

References

1 

Beasley RP: Hepatitis B virus. The major etiology of hepatocellular carcinoma. Cancer. 61:1942–1956. 1998. View Article : Google Scholar

2 

Papatheodoridis GV, Lampertico P, Manolakopoulos S and Lok A: Incidence of hepatocellular carcinoma in chronic hepatitis B patients receiving nucleos(t)ide therapy: a systematic review. J Hepatol. 53:348–356. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Fan X, Fang D, Bin D, Xueyong L, Jun C and Hongshan W: Serum levels of surface large envelope protein: prognostic marker for hepatitis B e antigen-negative patients with adefovir dipivoxil treatment. Antivir Ther. 14:1149–1156. 2009. View Article : Google Scholar

4 

Wong DK, Yuen MF, Poon RT, Yuen JC, Fung J and Lai CL: Quantification of hepatitis B virus covalently closed circular DNA in patients with hepatocellular carcinoma. J Hepatol. 45:553–559. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Barone M, Daniela Spano D, D'Apolito M, Centra M, Lasalandra C, Capasso M, Di Leo A, Volinia S, Arcelli D, Rosso N, Francavilla A, Tiribelli C and Iolascon A: Gene expression analysis in HBV transgenic mouse liver: a model to study early events related to hepatocarcinogenesis. Mol Med. 12:115–123. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Ehebauer M, Hayward P and Arias AM: Notch, a universal arbiter of cell fate decisions. Science. 314:1414–1415. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Louis AA, Van Eyken P, Haber BA, Hicks C, Weinmaster G, Taub R and Rand EB: Hepatic jagged1 expression studies. Hepatology. 30:1269–1275. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Geisler F, Nagl F, Mazur PK, Lee M, Zimber-Strobl U, Strobl LJ, Radtke F, Schmid RM and Siveke JT: Liver-specific inactivation of Notch2, but not Notch1, compromises intrahepatic bile duct development in mice. Hepatology. 48:607–616. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Yuan ZR, Kobayashi N and Kohsaka T: Human Jagged 1 mutants cause liver defect in Alagille syndrome by overexpression of hepatocyte growth factor. J Mol Biol. 356:559–568. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Nijjar SS, Wallace L, Crosby HA, Hubscher SG and Strain AJ: Altered Notch ligand expression in human liver disease: further evidence for a role of the Notch signaling pathway in hepatic neovascularization and biliary ductular defects. Am J Pathol. 160:1695–1703. 2002. View Article : Google Scholar

11 

Gramantieri L, Giovannini C, Lanzi A, Chieco P, Ravaioli M, Venturi A, Grazi GL and Bolondi L: Aberrant Notch3 and Notch4 expression in human hepatocellular carcinoma. Liver Int. 27:997–1007. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Siegel AB, Olsen SK, Magun A and Brown RS Jr: Sorafenib: where do we go from here? Hepatology. 52:360–369. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Li GL, Wei HS, Song SJ, Guo J and Cheng J: The effects of angiotensin II on gene expression of hepatic stellate cells. Zhonghua Gan Zang Bing Za Zhi. 14:914–919. 2006.(In Chinese).

14 

Gutiérrez Gallego R, Haseley SR, van Miegem VF, Vliegenthart JF and Kamerling JP: Identification of carbohydrates binding to lectins by using surface plasmon resonance in combination with HPLC profiling. Glycobiology. 14:373–386. 2004.PubMed/NCBI

15 

Chen X, Kintner DB, Luo J, Baba A, Matsuda T and Sun D: Endoplasmic reticulum Ca2+ dysregulation and endoplasmic reticulum stress following in vitro neuronal ischemia: role of Na+-K+-Cl cotransporter. J Neurochem. 106:1563–1576. 2008.

16 

Shuda M, Kondoh N, Imazeki N, Tanaka K, Okada T, Mori K, Hada A, Arai M, Wakatsuki T, Matsubara O, Yamamoto N and Yamamoto M: Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J Hepatol. 38:605–614. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Arai M, Kondoh N, Imazeki N, Hada A, Hatsuse K, Kimura F, Matsubara O, Mori K, Wakatsuki T and Yamamoto M: Transformation-associated gene regulation by ATF6alpha during hepatocarcinogenesis. FEBS Lett. 580:184–190. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Gao J, Song Z, Chen Y, Xia L, Wang J, Fan R, Du R, Zhang F, Hong L, Song J, Zou X, Xu H, Zheng G, Liu J and Fan D: Deregulated expression of Notch receptors in human hepatocellular carcinoma. Dig Liver Dis. 40:114–121. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Giovannini C, Lacchini M, Gramantieri L, Chieco P and Bolondi L: Notch3 intracellular domain accumulates in HepG2 cell line. Anticancer Res. 26:2123–2127. 2006.PubMed/NCBI

20 

Xu HY, Li BJ, Wang RF and Andersson R: Alterations of Notch/Jagged mRNA and protein expression after partial hepatectomy in rats. Scand J Gastroenterol. 43:1522–1528. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Monsalve E, Ruiz-García A, Baladrón V, Ruiz-Hidalgo MJ, Sánchez-Solana B, Rivero S, García-Ramírez JJ, Rubio A, Laborda J and Díaz-Guerra MJ: Notch1 upregulates LPS-induced macrophage activation by increasing NF-kappaB activity. Eur J Immunol. 39:2556–2570. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Itoh M, Fu L and Tohda S: NF-κB activation induced by Notch ligand stimulation in acute myeloid leukemia cells. Oncol Rep. 22:631–634. 2009.

23 

Nevzorova YA, Tschaharganeh D, Gassler N, Geng Y, Weiskirchen R, Sicinski P, Trautwein C and Liedtke C: Aberrant cell cycle progression and endoreplication in regenerating livers of mice that lack a single E-type cyclin. Gastroenterology. 137:691–703. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Cho JW, Lee KS and Kim CW: Curcumin attenuates the expression of IL-1β, IL-6, and TNF-α as well as cyclin E in TNF-α-treated HaCaT cells; NF-κB and MAPKs as potential upstream targets. Int J Mol Med. 19:469–474. 2007.

25 

Morga E, Mouad-Amazzal L, Felten P, Heurtaux T, Moro M, Michelucci A, Gabel S, Grandbarbe L and Heuschling P: Jagged1 regulates the activation of astrocytes via modulation of NFkappaB and JAK/STAT/SOCS pathways. Glia. 57:1741–1753. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Feng B, Cheng S, Hsia CY, King LB, Monroe JG and Liou HC: NF-kappaB inducible genes BCL-X and cyclin E promote immature B-cell proliferation and survival. Cell Immunol. 232:9–20. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Bharti AC, Takada Y, Shishodia S and Aggarwal BB: Evidence that receptor activator of nuclear factor (NF)-kappaB ligand can suppress cell proliferation and induce apoptosis through activation of a NF-kappaB-independent and TRAF6-dependent mechanism. J Biol Chem. 279:6065–6076. 2004. View Article : Google Scholar

28 

Chen E and Li CC: Association of Cdk2/cyclin E and NF-kappa B complexes at G1/S phase. Biochem Biophys Res Commun. 249:728–734. 1998. View Article : Google Scholar : PubMed/NCBI

29 

Pujol MJ, Jaime M, Serratosa J, Jaumot M, Agell N and Bachs O: Differential association of p21Cip1 and p27Kip1 with cyclin E-CDK2 during rat liver regeneration. J Hepatol. 33:266–274. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Koroxenidou L, Ohlson LC and Porsch Hällström I: Long-term 17alpha-ethinyl estradiol treatment decreases cyclin E and cdk2 expression, reduces cdk2 kinase activity and inhibits S phase entry in regenerating rat liver. J Hepatol. 43:478–484. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Lin SS, Huang HP, Yang JS, Wu JY, Hsia TC, Lin CC, Lin CW, Kuo CL, Gibson Wood W and Chung JG: DNA damage and endoplasmic reticulum stress mediated curcumin-induced cell cycle arrest and apoptosis in human lung carcinoma A-549 cells through the activation of caspase cascade- and mitochondrial-dependent pathway. Cancer Lett. 272:77–90. 2008. View Article : Google Scholar

32 

Derkx PM and Madrid SM: The foldase CYPB is a component of the secretory pathway of Aspergillus niger and contains the endoplasmic reticulum retention signal HEEL. Mol Genet Genomics. 266:537–545. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Feng Z, Li H, Liu S, Cheng J, Xiang G and Zhang J: FAM172A induces S phase arrest of HepG2 cells via Notch 3 Retraction in /10.3892/or.2023.8554. Oncol Rep 29: 1154-1160, 2013
APA
Feng, Z., Li, H., Liu, S., Cheng, J., Xiang, G., & Zhang, J. (2013). FAM172A induces S phase arrest of HepG2 cells via Notch 3 Retraction in /10.3892/or.2023.8554. Oncology Reports, 29, 1154-1160. https://doi.org/10.3892/or.2013.2235
MLA
Feng, Z., Li, H., Liu, S., Cheng, J., Xiang, G., Zhang, J."FAM172A induces S phase arrest of HepG2 cells via Notch 3 Retraction in /10.3892/or.2023.8554". Oncology Reports 29.3 (2013): 1154-1160.
Chicago
Feng, Z., Li, H., Liu, S., Cheng, J., Xiang, G., Zhang, J."FAM172A induces S phase arrest of HepG2 cells via Notch 3 Retraction in /10.3892/or.2023.8554". Oncology Reports 29, no. 3 (2013): 1154-1160. https://doi.org/10.3892/or.2013.2235