G-patch domain containing 2, a gene highly expressed in testes, inhibits nuclear factor-κB and cell proliferation

  • Authors:
    • Fen Hu
    • Lixia Gou
    • Qing Liu
    • Wendian Zhang
    • Mengmeng Luo
    • Xiujun Zhang
  • View Affiliations

  • Published online on: November 5, 2014     https://doi.org/10.3892/mmr.2014.2870
  • Pages: 1252-1257
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

G-patch domain containing 2 (GPATC2), a human gene that is highly expressed in the testes, was implicated as a novel cancer/testis antigen. The present study investigated GPATC2 expression in a number of human cell lines and rat tissues, and its potential biological function in 293T cells. Semi‑quantitative reverse transcription-polymerase chain reaction analysis showed that GPATC2 was widely expressed in 15 human cell lines (representing different lineages) and in 11 different rat tissues, and that the GPATC2 mRNA relative expression level was significantly higher in the testis than it was in other tissues. 293T cells were transiently transfected with GPATC2-p enhanced green fluorescent protein (EGFP)‑N1 or GPATC2-pEGFP-C3 and the nuclei were stained with 4',6'‑diamidino‑2‑phenylindole. The results showed that GPATC2 is predominantly expressed in the nucleus of 293T cells. Overexpression of GPATC2 may inhibit transcription of the NF-κB reporter gene. The role of GPATC2 in proliferation was analyzed with cell counting kit-8, colony-forming efficiency and flow cytometry assays. The results indicated that over‑expression of GPATC2 in 293T cells significantly inhibited cell proliferation by decreasing the number of cells in S phase. By contrast, GPATC2 knockdown by RNA interference exhibited the opposite effect, suggesting that GPATC2 may be involved in inhibiting G1-S phase transition in 293T cells. In conclusion, these results provide novel insight into the breadth of expression of GPATC2 and its role in cell proliferation.

Introduction

The mammalian testis serves two main functions: The synthesis of steroids and the production of spermatozoa. This second role is achieved through a process of transcriptional, translational and posttranslational regulation (1). A number of spermatogenesis-related genes, which are involved in specific phases of germ cell development, have been identified during the past few years, and certain genes have been shown to be important in spermatogenesis (23). However, spermatogenesis-derived factors involved in this process and the underlying molecular mechanisms remain largely obscure.

Cell division and proliferation, which are the most basic features of living organisms, are crucial in cell survival. Apoptosis, or programmed cell death, is the process by which excess or dysfunctional cells are eliminated, and is critical to maintain tissue homeostasis (4). Preserving the balance between cell proliferation and death is essential in sustaining differentiated tissue homeostasis and also forms a defense mechanism against pathogens (5). Nuclear factor (NF)-κB is a transcription factor that is important in regulating inflammation, immunity, cell proliferation and apoptosis (69).

Homo sapiens G-patch domain containing 2 (GPATC2; GenBank accession no. BC042193.1) also termed CT110, PPP1R30 and RP11-361K17.1, was first cloned by The National Institutes of Health Mammalian Gene Collection program in October, 2002, which is a multi-institutional effort to identify and sequence a cDNA clone containing a complete open reading frame for each human and mouse gene (10). It has been shown using northern blot analysis that the GPATC2 gene is highly expressed in the testes (11). The functions and mechanisms of action of the GPATC2 gene have not yet been elucidated. As no suitable male germ cell lines are currently available, a somatic cell line was used to investigate the function of GPATC2 in apoptosis, cell proliferation and the cell cycle.

The aim of the present study was to investigate the expression of GPATC2 in different human cell lines and rat tissues, and to determine the sub-cellular localization of the GPATC2 protein in 293T cells. In addition, the function of GPATC2 in 293T cell cycle progression was studied in order to provide novel insight into the breadth of expression of GPATC2 and its role in cell proliferation.

Materials and methods

Plasmid construction

The generation of vector pCMV-SPORT6-GPATC2 has been cloned by The Chinese National Human Genome Research Center (CHGB; Beijing, China), full-length GPATC2 the coding sequence was obtained by double enzyme digestion from pCMV-SPORT6-GPATC2 and cloned into the pcDNA3.1/myc-His (12) B vectors (pcDB; Invitrogen, Carlsbad, CA, USA), enhanced green fluorescent protein plasmid-N1 (pEGFP-N1) and pEGFP-C3, using the same enzyme restriction sites for EcoRI and BamHI.

Preparation of small interfering RNAs (siRNA)

The target sequence of the siRNA was 5′-GGAGCTGGTTCATGACCTT-3′ for human GPATC2. Sense and antisense oligonucleotides with the internal loop were synthesized by Takara Bio Inc., (Tokyo, Japan). These were annealed and ligated into the BamHI and HindIII sites of pSilencer 4.1-CMVneo (Ambion, Carlsbad, CA, USA) to construct the GPATC2-specific siRNA expression plasmid, according to the manufacturer’s instructions. pSilencer 4.1-CMVneo expressing scrambled siRNAs (Ambion) was used as a control.

Immunofluorescence microscopy

293T cells were transfected with si-GPATC2 or si-control and were grown on glass chamber slides. The cells were fixed using 4% paraformaldehyde in phosphate-buffered saline (PBS) for 30 min, permeabilized in 0.1% Triton X-100 for 30 min and then blocked with 0.5% bovine serum albumin in PBS for 30 min at room temperature (RT). After washing with PBS, the cells were incubated with polyclonal mouse anti-human GPATC2 antibody (H00055105-B01; Abnova, Walnut, CA, USA) for 2 h at 37°C. After washing with PBS and Tween-20, the cells were incubated with rabbit anti-mouse fluorescein isothiocyanate-conjugated secondary antibody (F-4143; Sigma, St. Louis, MO, USA) and stained with 4′,6-diamidino-2-phenylindole (DAPI; Roche, Basel, Switzerland). Images were visualized and captured using an Olympus microscope (Olympus, Tokyo, Japan).

Cell culture and transient transfection

H520, Hela, Hepg2, H1299, THP-1, Jurkas, U937, 786-O, HL60, A498, Caki, G401, MCF-7, T47d and 293T cells (American Type Culture Collection, Manassas, VA, USA) were maintained in Dulbecco’s modified Eagle’s medium or RPMI-1640 (HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum (Gibco-BRL, Grand Island, NY, USA) under 5% CO2 at 37°C. 293T cells seeded at a density of 1×104 cells/well in 96-well plates or 3×105 cells/well in 6-well plates and were transfected with empty vector pcDB or pcDB-GPATC2 using Vigorous transfection reagent (Vigorous Instruments Co., Ltd., Beijing, China), according to the manufacturer’s instructions.

RNA isolation and reverse transcription-polymerase chain reaction (RT-PCR)

Male Sprague-Dawley rats were purchased from Peking University Laboratory Animal Center (Beijing, China). The study was approved by the Ethics Committee of Hebei United University (Tangshan, China). Rats were sacrificed by cervical dislocation. Appendix, liver, heart muscle, lung, small intestine, skeletal muscle, pancreas, brain, kidney, stomach and testis tissues were obtained from three rats of the same age. Total mRNA was extracted from all tissues and cell lines using the TRIzol RNA isolation protocol (Gibco-BRL). Samples (0.25 μg) were used for cDNA synthesis using the PrimeScript RT Master mix (Perfect Real Time) (Takara Bio Inc.). GPATC2 and GAPDH internal control transcripts were amplified by RT-PCR using the following primers: Forward: 5′-GCGCAGGCCGTCATCAAC-3′ and reverse: 5′-TCCCACCAGGGCACAACTC-3′ for human GPATC2; forward: 5′-GCCAGTATCCCGAGTTTAT-3′ and reverse: 5′-GCCCGACTCCTTCTCATA-3′ for rat GPATC2; and forward: 5′-AAGAAGGTGGTGAAGCAGGC-3′ and reverse: 5′-TCCACCACCCTGTTGCTGTA-3′ for GAPDH.

Subcellular localization

293T cells seeded at a density of 1×104 cells/well in 96-well plates were transfected with pEGFP-N1-GPATC2 or pEGFP-C3-GPATC2. After 24 h, cells were washed with PBS, fixed with 4% paraformaldehyde for 10 min at RT, washed again with PBS and stained with DAPI (5 μg/ml in PBS) at RT for 5 min. Cells were visualized using an inverted fluorescence microscope (Nikon TE 2000-U, Nikon, Tokyo, Japan).

Dual-luciferase reporter assay

293T cells were seeded into 96-well plates at 1×104 cells/well. After 24 h, cells were co-transfected with 80 ng/well pcDB-GPATC2 or pcDB, 40 ng pNF-κB-Luc plasmid containing the firefly luciferase reporter gene (PathDetect, Stratagene, La Jolla, CA, USA) and 4 ng internal control pRL-TK plasmid containing the renilla luciferase gene (Promega Corporation, Madison, WI, USA). Each transfection was conducted in triplicate wells. After 24 h, cells were lysed in standard lysis buffer (Promega Corporation) and cell lysates were assayed for firefly and renilla luciferase activities using the dual-luciferase reporter (DLR) Assay system (Promega Corporation), according to the manufacturer’s instructions. Light emission was measured using a GENios Pro reader (Tecan, Männedorf, Switzerland). Luciferase activity was normalized to renilla luciferase activity. Duplicate experiments were performed.

Cell counting kit-8 (CCK-8) assay

293T cells transfected with pcDB or pcDB-GPATC2 were seeded into 96-well plates at a density of 2×103 cells/well. At the indicated time point, the cell counting kit proliferation assay (Dojindo, Tokyo, Japan) was performed by adding 10 μl CCK-8 solution to each well and incubating at 37°C for 2 h. Absorbance at 450 nm was then measured using a microplate plate reader (Synergy 2 Multi-Mode Microplate Reader, BioTek, Winooski, VT, USA).

Flow cytometry

293T cells were synchronized by serum starvation for 12 h and then transfected with 2.5 μg pcDB or pcDB-GPATC2. Cells were trypsinized 48 h after GPATC2 transfection, washed twice with PBS and fixed with ice-cold 70% ethanol. Fixed cells were pelleted, washed and resuspended in PBS. Samples were then treated with 1 μl of 10 mg/ml DNase free RNase A (Promega Corporation) incubated at 37°C for 30 min and then 50 μl of 300 mg/ml propidium iodide (Boehringer, Mannheim, Germany) containing Triton X-100 was added and cells were incubated in the dark for 30 min. Cell cycle data were obtained by flow cytometry (FACSCalibur, BD Biosciences, Franklin Lakes, NJ, USA).

Results

Characterization and expression profile of the GPATC2 gene

The putative GPATC2 protein is comprised of 376 amino acids with a deduced molecular mass of ~43 kDa and a theoretical isoelectric point of 7.65. The putative GPATC2 protein has no transmembrane helices as predicted by the program TMHMM Server v.2.0 (http://www.cbs.dtu.dk/services/TMHMM/), and no other domains, including signal peptides, were found by the InterProScan program (http://www.ebi.ac.uk/Tools/InterProScan/). Human GPATC2 is located on chromosome 1q41. As shown in Fig. 1A, GPATC2 mRNA was widely expressed in the 15 human cell lines examined, which represented different lineages, as measured by RT-PCR. This technique was also performed to analyze GPATC2 mRNA expression in 11 different rat tissues, including appendix, liver, heart muscle, lung, small intestine, skeletal muscle, pancreas, brain, kidney, stomach and testis tissues. As shown in Fig. 1B, GPATC2 mRNA was expressed in a number of these tissues, although the GPATC2 mRNA relative expression level was significantly higher in the testis than it was in other tissues.

Over-expressed GPATC2 localizes to the nucleus and inhibits NF-κB

To determine the sub-cellular localization of GPATC2, GPATC2 cDNA was fused to the 3′ end of the green fluorescent protein (GFP) cDNA in the pEGFP-C3 vector and to the 5′ end of the GFP cDNA in the pEGFP-N1 vector. As shown in Fig. 2A, GPATC2-EGFP was predominantly expressed in the nucleus of 293T cells. The DLR assay system showed that GPATC2 over-expression in 293T cells inhibited the NF-κB reporter gene compared with the control group (Fig. 2B).

GPATC2 over-expression inhibits cell proliferation

As shown in Fig. 3A and B, cell numbers decreased significantly 24 h after GPATC2 over-expression was established compared with empty vector controls. As shown in Fig. 3C, colony-forming efficiency was 3.0-fold higher in controls cells than in cells over-expressing GPATC2 (Fig. 3C). This indicates a growth-inhibiting effect of GPATC2 on 293T cells. Furthermore, flow cytometry showed that a reduced proportion of 293T cells were in S phase 48 h after GPATC2 transfection, with 39.2% of cells over-expressing GPATC2 compared with 51.3% of controls in S phase (Fig. 3D). In addition, GPATC2 over-expression increased the proportion of cells in the G0/G1phase. These data suggest that GPATC2 over-expression significantly inhibits cell cycle G1-S phase progression in 293T cells.

GPATC2 knockdown by RNA interference promotes cell proliferation

Based on the above results, it was hypothesized that knockdown of GPATC2 using RNA interference results in an increased number of cells in S phase. In order to test this hypothesis, an siRNA expression plasmid targeting GPATC2 or a scrambled control siRNA plasmid was transiently transfected into 293T cells. The reduction in GPATC2 expression was demonstrated by the use of immunofluorescence (Fig. 4A). As shown in Fig. 4B, in 293T cells depleted of GPATC2, cell numbers increased significantly after two days, compared with the empty vector-transfected cells. These data suggest that GPATC2 knockdown significantly promotes cell proliferation in 293T cells.

Discussion

GPATC2 exhibits high RNA expression in rat testis, an important organ involved in the synthesis of steroids and production of spermatozoa. Spermatogenesis is a complex biological process involving the mitotic proliferation of spermatogonia and the meiotic division of spermatocytes, followed by the morphogenic differentiation of spermatids to mature spermatozoa. This intricate process is reflected in the complex gene expression in the testis (1314). This study shows that GPATC2 is a nuclear factor, which suggests that it may be important in spermatogenesis.

NF-κB is a generic term for a dimeric transcription factor formed by the heterodimerization or homodimerization of members of the Rel/NF-κB family (7). NF-κB-regulated genes are important for cell differentiation, embryonic development, the immune response and inflammation (1517), as well as the development, progression and drug resistance of cancer cells (18,19). The present study demonstrated that GPATC2 inhibits the NF-κB pathway, suggesting that it may be an important regulator of a variety of cell functions. Furthermore, the results showed that GPATC2 inhibits 293T cell cycle progression by decreasing the number of cells in S phase, indicating that GPATC2 may be an important regulator of cell cycle progression. Lin et al (11) demonstrated that the interaction of GPATC2 protein with hPrp43, an RNA-dependent ATPase, significantly enhanced the ATPase activity of hPrp43 and induced a growth-promoting effect on mammalian cells. However, the mechanism through which GPATC2 inhibits 293T cell proliferation requires further elucidation.

In conclusion, GPATC2, which is highly expressed in the testis and is largely localized to the nucleus of 293T cells, is capable of inhibiting NF-κB and 293T cell cycle progression in vitro. However, the mechanism of GPATC2 function may be far more complex in vivo and further investigations are required to elucidate the role and molecular mechanisms of GPATC2 in the process of spermatogenesis.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81170616, 81072093, 30671092 and 81302323); and the Natural Science Foundation of Hebei Province (grant nos. C2009001260, C2012401039, H2013209194, C2013209024 and C2014209140).

References

1 

Bettegowda A and Wilkinson MF: Transcription and post-transcriptional regulation of spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 365:1637–1651. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Liu ML, Cheng YM and Jia MC: LM23 is essential for spermatogenesis in Rattus norvegicus. Front Biosci (Elite Ed). 2:187–194. 2010. View Article : Google Scholar

3 

De Gendt K, Swinnen JV, Saunders PT, et al: A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci USA. 101:1327–1332. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Hale AJ, Smith CA, Sutherland LC, et al: Apoptosis: molecular regulation of cell death. Eur J Biochem. 237:8841996.PubMed/NCBI

5 

Kim JH, Yang YI, Lee KT, et al: Costunolide induces apoptosis in human endometriotic cells through inhibition of the prosurvival Akt and nuclear factor kappa B signaling pathway. Biol Pharm Bull. 34:580–585. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Gebel HM, Braun DP, Tambur A, et al: Spontaneous apoptosis of endometrial tissue is impaired in women with endometriosis. Fertil Steril. 69:1042–1047. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Karin M and Ben-Neriah Y: Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annu Rev Immunol. 18:621–663. 2000. View Article : Google Scholar

8 

Huxford T, Malek S and Ghosh G: Structure and mechanism in NF-kappa B/I kappa B signaling. Cold Spring Harb Symp Quant Biol. 64:533–540. 1999. View Article : Google Scholar

9 

Baldwin AS Jr: The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 14:649–683. 1996. View Article : Google Scholar : PubMed/NCBI

10 

Strausberg RL, Feingold EA, Grouse LH, et al: Mammalian Gene Collection Program Team: Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. Proc Natl Acad Sci USA. 99:16899–16903. 2002. View Article : Google Scholar

11 

Lin ML, Fukukawa C, Park JH, et al: Involvement of G-patch domain containing 2 overexpression in breast carcinogenesis. Cancer Sci. 100:1443–1450. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Sekido R, Murai K, Funahashi J, et al: The delta-crystallin enhancer-binding protein delta EF1 is a repressor of E2-box-mediated gene activation. Mol Cell Biol. 14:5692–5700. 1994. View Article : Google Scholar : PubMed/NCBI

13 

Dufau ML, Tsai-Morris C, Tang P and Khanum A: Regulation of steroidogenic enzymes and a novel testicular RNA helicase. J Steroid Biochem Mol Biol. 76:187–197. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Walker WH: Molecular mechanisms of testosterone action in spermatogenesis. Steroids. 74:602–607. 2009. View Article : Google Scholar

15 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Luo JL, Maeda S, Hsu LC, et al: Inhibition of NF-kappaB in cancer cells converts inflammation-induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell. 6:297–305. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Luo JL, Tan W, Ricono JM, et al: Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature. 446:690–694. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Ikezoe T, Yang Y, Bandobashi K, et al: Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways. Mol Cancer Ther. 4:578–586. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Karin M, Cao Y, Greten FR and Li ZW: NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2:301–310. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2015
Volume 11 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
Hu F, Gou L, Liu Q, Zhang W, Luo M and Zhang X: G-patch domain containing 2, a gene highly expressed in testes, inhibits nuclear factor-κB and cell proliferation. Mol Med Rep 11: 1252-1257, 2015
APA
Hu, F., Gou, L., Liu, Q., Zhang, W., Luo, M., & Zhang, X. (2015). G-patch domain containing 2, a gene highly expressed in testes, inhibits nuclear factor-κB and cell proliferation. Molecular Medicine Reports, 11, 1252-1257. https://doi.org/10.3892/mmr.2014.2870
MLA
Hu, F., Gou, L., Liu, Q., Zhang, W., Luo, M., Zhang, X."G-patch domain containing 2, a gene highly expressed in testes, inhibits nuclear factor-κB and cell proliferation". Molecular Medicine Reports 11.2 (2015): 1252-1257.
Chicago
Hu, F., Gou, L., Liu, Q., Zhang, W., Luo, M., Zhang, X."G-patch domain containing 2, a gene highly expressed in testes, inhibits nuclear factor-κB and cell proliferation". Molecular Medicine Reports 11, no. 2 (2015): 1252-1257. https://doi.org/10.3892/mmr.2014.2870