Microarray-based analysis of the gene expression profile in GC-1 spg cells transfected with spermatogenesis associated gene 12

  • Authors:
    • Yiting Lin
    • Zhiwen Liu
    • Xuanming Liu
    • Yunsheng Zhang
    • Zhuoxian Rong
    • Dan Li
  • View Affiliations

  • Published online on: December 27, 2012     https://doi.org/10.3892/ijmm.2012.1225
  • Pages: 459-466
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The unique differentiation mechanisms of spermatogenesis suggest the existence of cell type- and stage-specific molecules. Herein, a microarray-based approach was used to identify changes in the gene expression profile in mouse GC-1 spg germ cells transfected with spermatogenesis associated gene 12 (SPATA12) . One hundred and eighty-two upregulated genes and 104 downregulated genes with fold changes of ≥2 or ≤0.5 (P≤0.05) in expression were identified. Ten genes were selected for validation of the microarray results using quantitative RT-PCR. The real-time quantitative RT-PCR results were consistent with that of the microarray. The gene ontology (GO) terms for the biological functions of the differentially expressed genes induced by SPATA12 included binding activity and immune response. Biological pathway analysis identified several related pathways which are associated with immune responses, cell adhesion and the developmental process. In addition, we observed that SPATA12 may interact with the β-catenin signaling pathway and that SPATA12 may negatively regulate β-catenin signaling during spermatogenesis.

Introduction

Infertility affects approximately 15% of couples worldwide, and the male factor is at least partly responsible in approximately 50% of infertile couples (1). Genetic abnormalities, i.e. obstacles to spermatogenesis, are thought to account for 15–30% of male factor infertility (2). Spermatogenesis is the process by which male germ cells undergo a complex differentiation where morphological alterations lead to the formation of differentiated sperm. The unique differentiation mechanisms of spermatogenesis suggest the existence of cell type- and stage-specific molecules. With the advent of genetics and molecular biology, a variety of genes involved in spermatogenesis have been identified during the last decade (39).

Recently, we identified a novel protein referred to as spermatogenesis associated gene 12 (SPATA12) (10,11). In the testis, SPATA12 is specifically expressed in spermatocytes, spermatids and spermatozoa, and may be involved in the development of testicular maturation. In contrast, the SPATA12 gene is located on chromosome 3p14. Chromosomal abnormalities including homozygous deletions, loss of heterozygosity and expressional deficiencies in genes located at 3p14 have been frequently reported in many tumor types, suggesting that this locus is likely to contain tumour-suppressor genes (12,13). SPATA12 was also found to be absent in testicular germ cell tumors such as seminoma, yolk sac, teratoma and embryonal carcinoma. Flow cytometric analysis of SPATA12 in both mouse GC-1 spg germ cells and human HeLa cells indicates that the expression of the SPATA12 gene may delay G1 to S phase progression in the cell cycle. In addition, SPATA12 was shown to inhibit tumor cell colony formation. These findings suggest that SPATA12 could be an inhibitor suppressing cell proliferation in the process of germ cell development and in tumorigenesis. However, the transcriptional regulations of SPATA12 in spermatogenesis remain unclear.

The emerging technology of cDNA microarray hybridization offers the possibility of providing a rapid, high-throughput method for the efficient and accurate simultaneous expression measurement of thousands of genes (1417). Studies based on microarray or expressed sequence tag analyses have been successfully used during spermatogenesis, and have aided in the understanding of the molecular mechanisms and genetic determinants of male infertility (1820). Based on this background, we hypothesized that analysis of the gene expression profile induced by SPATA12 in GC-1 spg germ cells may contribute to an understanding of the function of SPATA12 and the possible pathways in which SPATA12 is involved during spermatogenesis.

Materials and methods

Cell line

The mouse GC-1 spg germ cell line (ATCC CRL-2053) was cultured in Dulbecco’s modified Eagle’s medium supplemented with 10% FBS, 100 μg/ml penicillin-streptomycin, and was maintained in 5% CO2 and a 95% humidified air atmosphere at 37°C.

Transient transfection

Transfections were performed with Lipofectamine 2000 (Invitrogen). Cells were plated to 50–70% confluent culture in each well of a 6-well plate or a 60-mm dish 24 h before transfection. According to the manufacturer’s instructions, the GC-1 spg cells were transfected with 4 μg pRevTRE plasmid or pRevTRE-SPATA12 plasmid, respectively.

RNA isolation

Total RNA was extracted with the TRIzol reagent (Invitrogen) according to the manufacturer’s protocol, digested by RNase-free DNase (Fermentas), dissolved in diethyl pyrocarbonate-treated water, and stored at −80°C prior to use. For quality control, RNA purity and integrity were evaluated by agrose gel electrophoresis and the OD260/OD280 ratio.

Microarray assay and data analysis

32K Mouse Genome Array (CapitolBio Corporation, Beijing, China), covering 99% of the current assembly of the mouse genome and including 32,256 Oligo DNA with 70 mer length, was applied to investigate the possible changes in the mRNA level in GC-1 spg cells following SPATA12 gene transfection by comparison to control GC-1 spg cells. Total RNA was extracted using TRIzol reagent (Invitrogen) and then applied to synthesize Cy3- or Cy5-conjugated dUTP-labeled cDNA probe using the RNA Fluorescence Labeling Core kit (M-MLV version 2.0; Takara, Dalian, China) and following the manufacturer’s instructions. Hybridization, scanning, and data extraction were conducted at CapitalBio Corporation. The number of genes affected by SPATA12 was determined using the scatter plots of control GC-1 spg cells vs. GC-1 spg cells transfected with SPATA12 (named GC-1 spg-SPATA12 cells). In addition, identified genes were also categorized specifically using biological process ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathways using the software Molecule Annotation System 2.0 (http://:bioinfo.capitalbio.com/MAS/) (CapitolBio Corporation).

RT-PCR and SYBR-Green real-time PCR analysis

For cDNA synthesis, 2 μg of total RNA was reverse transcribed using M-MLV reverse transcriptase (Promega). Then PCR was performed in a 10 μl reaction volume containing 5.7 μl of nuclease-free water, 0.1 μl of Takara Taq (5 U/μl), 1 μl of 10X PCR buffer, 0.8 μl of dNTP mixture (2.5 mM), 2 μl of cDNA and 0.2 μl each of the 20 μM gene-specific primers. After initial denaturation for 10 min at 94°C, 27–30 cycles of PCR were performed. Each cycle consisted of a denaturing period (30 sec at 94°C), an annealing phase (55–60°C), and an extension period (60 sec at 72°C). After the last cycle, all samples were incubated for an additional 10 min at 72°C. PCR products were separated by 1.5% agarose gel electrophoresis, and the DNA bands were stained with ethidium bromide. RT-PCR signals were normalized to the signals of the murine glyceraldehyde-3-phosphate dehydrogenase (gapdh) gene.

For quantitative RT-PCR, a SYBR-Green real-time RT-PCR protocol (Invitrogen) was applied using an MX3000 (Stratagene, USA) instrument. PCR was performed in a 10 μl reaction volume containing 2.2 μl of nuclease-free water, 5 μl of Mix, 2 μl of cDNA and 0.4 μl each of the 2.5 μM gene-specific primers. The PCR profile was 95°C for 5 min followed by 94°C for 30 sec, 58°C for 20 sec, and 72°C for 20 sec for 40 cycles, with a final extension at 72°C for 10 min and storage at 4°C. The level of gene mRNA was evaluated in automated analysis by MxPro. All the primers for RT-PCR are shown in Table I.

Table I.

Primers used for real-time RT-PCR and RT-PCR experiments.

Table I.

Primers used for real-time RT-PCR and RT-PCR experiments.

Gene nameForward primer (5′→3′)Reverse primer (5′→3′)Product size (bp)
Gapdh1 GAAGGGTGGAGCCAAAAGG TTGCTGACAATCTTGAGTGAGTTG111
Ccl5 GCCCACGTCAAGGAGTATTTCT TCTCTGGGTTGGCACACACTT100
Fbxo39 AGCCTGAGGAGTTGCTATTTCAGT ACGTTAACTTCTGCAGGGTGTTC100
Wnt10a CTTCAGCCGAGGTTTTCGAGA CCGCAAGCCTTCAGTTTACC108
Rtp4 CATCTTTGGGTGAGAAGGTGACT GAGATCTGGGTGGTTTTACTTTGTG120
Sp100 AGCTACAACCACAGTCCCCT TCCTGTCCTTTTCCGTCTTCTAA111
Zbp1 GACGGACAGACGTGGAAGATC TTGACCGGATTGTGCTGACA110
Gys1 CGCTGGAAGGGTGAGCTTT GAAGTGGGCAACCACATACG156
Mtss1 ATGGAGGCTGTGATCGAGAAG TCCGGCTTTGTTTATGAAGTCTT114
Selenbp1 AGCCAGGTCATCCACAGGTT ACTTCGTGCTGTCCCCAAAG100
Ndrg1 CACACAACATTTTGCTGTCTGC GCCAACTGATCCATTGAGGGG100
Cyclin B1 TGGCCTCACAAAGCACATGA GCTGTGCCAGCGTGCTAATC77
Cyclin D1 TAGGCCCTCAGCCTCACTC CCACCCCTGGGATAAAGCAC80
Cyclin E1 AATTGGGGCAATAGAGAAGAGGT TGGAGCTTATAGACTTCGCACA161
β-catenin GGCAACCCTGAGGAAGAAGA CACTGGTGACCCAAGCATTTT597
Gapdh2 TTCAACGGCACAGTCAAGG TGAAGTCGCAGGAGACAACC694
SPATA12 CGCGGATCCATGTCCAGTTCTGCTCTGACT CCCAAGCTTGCAGGATTATTATTGATTACAG607

[i] The primers of Gapdh1 were used for quantitative RT-PCR, while those for Gapdh2 were used for RT-PCR.

Statistical analysis

Statistical analysis was carried out using the Student’s t-test. P-values ≤0.05 were considered to indicate statistically significant results.

Results

Global gene expression induced by SPATA12 in GC-1 spg cells

Global gene expression levels induced by SPATA12 in GC-1 spg cells from the microarray analysis are represented in Fig. 1. The differentially expressed genes with fold changes of ≥2 or ≤0.5 (P≤0.05) were analyzed using t-test and P-value and clustered with the software package Cluster 3.0. Our data showed that the expression of 286 out of 32,256 genes was altered after SPATA12 was expressed. Of these, 182 genes were upregulated and 104 genes were downregulated specifically.

Verification of the differentially expressed genes from the microarray data

To test the reliability of the microarray data, the expression patterns of 10 selected genes (including 6 upregulated genes and 4 downregulated genes) (Table II) were examined by quantitative real-time RT-PCR. The overall profile of gene expression by real-time RT-PCR analysis was similar to that revealed by the microarray data for all the selected genes (Figs. 2 and 3).

Table II.

Selected genes with differential expression between GC-1 spg and GC-1 spg-SPATA12 cells.

Table II.

Selected genes with differential expression between GC-1 spg and GC-1 spg-SPATA12 cells.

Term descriptionSymbolRatioGene nameGene function
Upregulated genesCcl532.0033Chemokine (C-C motif) ligand 5Cytokine activity; chemokine activity; chemoattractant activity; immune regulation
Rtp411.7258Receptor transporter protein 4Unknown
Zbp18.7427Z-DNA binding protein 1Left-handed Z-DNA binding; RNA binding; double-stranded RNA adenosine deaminase activity; innate immune response
Fbxo396.7244F-box protein 39Cancer/testis antigen
Sp1004.1610SP100 nuclear antigenTumorigenesis; immunity; gene regulation
Wnt10a2.1549Wingless related MMTV integration site 10aSignal transducer activity; receptor binding; embryogenesis; carcinogenesis
Downregulated genesGys10.4613Glycogen synthase 1 (muscle)Catalytic activity; glycogen (starch) synthase activity; protein binding; transferase activity; transferase activity, transferring glycosyl groups
Mtss10.3599Metastasis suppressor 1Unknown
Ndrg10.2371N-myc downstream regulated gene 1Stress responses; hormone responses; cell growth; cell differentiation; p53-mediated caspase activation and apoptosis
Selenbp10.1536Selenium binding protein 1Selenium binding; selenium-dependent role in ubiquitination/deubiquitination-mediated protein degradation
Gene ontology (GO) analysis of the microarray data

To gain insight into the potential functional consequences of the SPATA12-induced expression in GC-1 spg cells, GO analysis was applied to distribute genes into groups according to biological process, molecular function and cellular component, respectively.

Functional analysis using GO of the biological process group revealed that genes related to immune response, antigen processing and presentation, positive regulation of T cell-mediated cytotoxicity, transcription regulation, and defense response to bacterium were upregulated in the SPATA12-transfected GC-1 spg cells (Fig. 4A and B). This suggests that based on the GO terms these upregulated genes combined with SPATA12 may play important roles in the physiology of the immune response. In contrast, genes related to carbohydrate metabolism, lipid metabolism, cholesterol biosynthesis and glycolysis were downregulated in the SPATA12-transfected cells, which indicates that based on these GO terms these genes affected by SPATA12 may be involved in metabolic processes.

In the cellular component group (Fig. 4C and D), GO analysis showed that the major differentially expressed genes, including 61 upregulated and 22 downregulated genes, were located in the nucleus. These data indicate that SPATA12 may play roles in the nucleus through interacting with genes associated in these GO categories. This observation was consistent with our previous bioinformatics analysis report, which predicted that SPATA12 probably functions as a testis-specific nuclear protein involved in spermatogenesis (21).

We also noted that these genes (molecular function group) (Fig. 4E and F), either upregulated or downregulated in SPATA12-transfected cells, were mostly involved in protein binding, ATP/GTP binding, zinc ion binding or DNA/RNA binding, indicating that the function of SPATA12 may be related with the activity of ‘binding’.

Pathway analysis and the expression change in β-catenin signaling induced by SPATA12

The analysis of the differential expression of genes led us to wonder whether these genes may represent related pathways in which SPATA12 is involved. Thus, biological pathway analysis was applied to characterize the unique gene networks associated with SPATA12 in germ cells. Most genes associated with immune-related pathways such as antigen processing and presentation, cell adhesion, T cell receptor signaling pathway, and development-related pathways such as MAPK, Jak-STAT and Wnt, were significantly overexpressed which provides evidence that both immune responses and developmental processes may be associated with various functions of SPATA12 (Table III). Changes in the expression of Wnt signaling-related genes such as β-catenin supported this possibility. Semi-quantitative RT-PCR results showed that the expression of β-catenin was obviously downregulated (Fig. 5) and its downstream target cell cycle gene cyclin E1 was strongly decreased (Fig. 6) in GC-1 spg cells transfected with SPATA12.

Table III.

Selected pathways related with SPATA12 identified by biological pathway analysis.

Table III.

Selected pathways related with SPATA12 identified by biological pathway analysis.

Pathway nameTotalGene (ratio)
Antigen processing and presentation12H2-Q10 (2.36), H2-Q7 (2.79), H2-T9 (2.49), H2-T3 (2.01), H2-Q6 (2.02), Tap1 (2.3442), Psme2 (2.4576), H2-K1 (2.28), H2-Q1 (2.92), Psme2b-ps (2.4576), B2m (2.5449), H2-T23 (2.24)
Cell adhesion molecules (CAMs)9H2-Q10 (2.36), H2-Q7 (2.79), H2-T9 (2.49), H2-T3 (2.01), H2-Q6 (2.02), Itgb7 (2.73), H2-K1 (2.28), H2-Q1 (2.92), H2-T23 (2.24)
Type I diabetes mellitus8H2-T23 (2.24), H2-Q1 (2.92), H2-K1 (2.28), H2-Q6 (2.02), H2-T3 (2.01), H2-T9 (2.49), H2-Q7 (2.79), H2-Q10 (2.36)
Toll-like receptor signaling pathway5Myd88 (2.0637), Cxcl10 (5.5093), Stat1 (5.04), Cd14 (2.0947), Ccl5 (32.0033)
MAPK signaling pathway4Ddit3 (2.0538), Cd14 (2.0947), Dusp6 (2.9646), Map4k2 (0.4768)
Biosynthesis of steroids3Sqle (0.4314), Pmvk (0.4707), Fdft1 (0.4985)
ECM-receptor interaction3Col6a1 (0.4919), Col5a3 (0.4825), Itgb7 (2.7347)
Jak-STAT signaling pathway3Isgf3g (3.57), Stat1 (5.04), Stat2 (2.55)
Cytokine-cytokine receptor interaction3Ccl5 (32.0033), Vegfa (0.4651), Cxcl10 (5.5093)
Natural killer cell mediated cytotoxicity2H2-T23 (2.24), H2-K1 (2.28)
T cell receptor signaling pathway1Pdk1 (0.4713)
Wnt signaling pathway1Wnt10a (2.15)

Discussion

High-density cDNA microarrays provide an important tool to study the global patterns of gene expression. For example, it has been used to understand interactions in a given cell, tissue and organism under normal and diseased states (22,23), or it has been used for gene discovery and function (24,25). Spermatogenesis is a complex process of cell development and differentiation that requires the highly regulated expression of multiple genes (26). Characterization and functional analysis of new testis-specific genes related to spermatogenesis is of momentous physiological and pathological significance in order to understand the molecular mechanisms of spermatogenesis. Herein, cDNA microarray was used to identify the upregulated or downregulated genes affected by SPATA12 and obtain a global overview on the expression patterns of these genes, aimed at acquiring a further understanding of the function of SPATA12 and the possible pathways in which SPATA12 is involved.

We identified 182 upregulated genes and 104 downregulated genes with a fold change of ≥2 or ≤0.5 (P≤0.05) in expression. Through quantitative real-time RT-PCR, we confirmed the expression of 10 (Ccl5, Fbxo39, Wnt10a, Rtp4, Sp100, Zbp1, Gys1, Mtss1, Selenbp1 and Ndrg1) of the differentially expressed genes. These genes along with their GO categories identified as being differentially expressed following induction by SPATA12 may be of significant biological interest.

Gene ontology studies provide biologically meaningful information regarding genes including the cellular location, molecular function and biological process. Fig. 4 lists each GO category and the specific number of genes in their respective category that were differentially expressed following induction by SPATA12. Biological pathway analysis identified several related pathways such as antigen processing and presentation, cell adhesion, MAPK, Jak-STAT and Wnt. The differential expression of these signaling pathway-related genes may also show that SPATA12 is associated with immune responses in germ cell development.

Alterations in the expression of Wnt/β-catenin signaling-related genes support this possibility. Wnt/β-catenin signaling is one of the most important developmental signaling pathways that control cell fate decisions and tissue patterning during early and late embryonic development. β-catenin is a key component of the Wnt/β-catenin signaling pathway (2730) and was previously reported to be expressed in the plasma membrane and cytoplasm of germ cells during testis development. Suppression of Wnt/β-catenin signaling is necessary for the normal development of primordial germ cells since stabilization of β-catenin in germ cells was found to delay cell cycle progression resulting in germ cell deficiency (31). Here, we showed that expression of SPATA12 by transfection downregulated β-catenin in GC-1 spg cells. Studies in the literature reported that the reduction in β-catenin level was accompanied by inhibition of its transactivation potential and downregulation of downstream target genes, such as Myc, cyclin D1 and cyclin E1 (28,32,33). In the present study, our data suggest that SPATA12 negatively regulates cell cycle-related gene cyclin E1, which indicates that SPATA12 inhibits cell proliferation via downregulation of β-catenin in GC-1 spg cells. These results were shown to be of particular relevance between SPATA12 and the β-catenin signaling pathway.

Taken together, the present study demonstrated alterations in the gene expression profile of GC-1 spg cells transfected with SPATA12. The functional classification of these genes and their expression profiles provide useful information to understand the transcriptional regulation of SPATA12. A number of genes, GO categories and biological pathways such as immune responses may be associated with the function of SPATA12. Moreover, our study showed that SPATA12 may interact with the β-catenin signaling pathway and SPATA12 could negatively regulate β-catenin signaling during spermatogenesis.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (nos. 30872763, 81270735) and the Fundamental Research Funds for the Central Universities of China (no. 531107040314).

References

1. 

O’Flynn O’Brien KL, Varghese AC and Agarwal A: The genetic causes of male factor infertility: a review. Fertil Steril. 93:1–12. 2010.

2. 

Ferlin A, Raicu F, Gatta V, Zuccarello D, Palka G and Foresta C: Male infertility: role of genetic background. Reprod Biomed Online. 14:734–745. 2007. View Article : Google Scholar : PubMed/NCBI

3. 

Coussens M, Maresh JG, Yanagimachi R, Maeda G and Allsopp R: Sirt1 deficiency attenuates spermatogenesis and germ cell function. PLoS One. 3:e15712008. View Article : Google Scholar : PubMed/NCBI

4. 

Takeuchi A, Mishina Y, Miyaishi O, Kojima E, Hasegawa T and Isobe K: Heterozygosity with respect to Zfp148 causes complete loss of fetal germ cells during mouse embryogenesis. Nat Genet. 33:172–176. 2003. View Article : Google Scholar : PubMed/NCBI

5. 

Zhou CX, Zhang YL, Xiao L, et al: An epididymis-specific beta-defensin is important for the initiation of sperm maturation. Nat Cell Biol. 6:458–464. 2004. View Article : Google Scholar : PubMed/NCBI

6. 

Roy A, Yan W, Burns KH and Matzuk MM: Tektin3 encodes an evolutionarily conserved putative testicular microtubules-related protein expressed preferentially in male germ cells. Mol Reprod Dev. 67:295–302. 2004. View Article : Google Scholar

7. 

Takahashi T, Tanaka H, Iguchi N, et al: Rosbin: a novel homeobox-like protein gene expressed exclusively in round spermatids. Biol Reprod. 70:1485–1492. 2004. View Article : Google Scholar : PubMed/NCBI

8. 

Kissel H, Georgescu MM, Larisch S, Manova K, Hunnicutt GR and Steller H: The Sept4 septin locus is required for sperm terminal differentiation in mice. Dev Cell. 8:353–364. 2005. View Article : Google Scholar : PubMed/NCBI

9. 

Zhu H, Zhu JX, Lo PS, et al: Rescue of defective pancreatic secretion in cystic-fibrosis cells by suppression of a novel isoform of phospholipase C. Lancet. 362:2059–2065. 2003. View Article : Google Scholar : PubMed/NCBI

10. 

Dan L, Lifang Y and Guangxiu L: Expression and possible functions of a novel gene SPATA12 in human testis. J Androl. 28:502–512. 2007. View Article : Google Scholar : PubMed/NCBI

11. 

Liu ZW, Lin YT, Liu XM, Yu WW, Zhang YS and Li D: Experimental study of inhibition of tumor cell proliferation by a novel gene SPATA12. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 37:222–227. 2012.(In Chinese).

12. 

Ji L, Minna JD and Roth JA: 3p21.3 tumor suppressor cluster: prospects for translational applications. Future Oncol. 1:79–92. 2005. View Article : Google Scholar : PubMed/NCBI

13. 

Riquelme E, Tang M, Baez S, Diaz A, Pruyas M, Wistuba II and Corvalan A: Frequent epigenetic inactivation of chromosome 3p candidate tumor suppressor genes in gallbladder carcinoma. Cancer Lett. 250:100–106. 2007. View Article : Google Scholar : PubMed/NCBI

14. 

Kim HJ, Joo HJ, Kim YH, et al: Systemic analysis of heat shock response induced by heat shock and a proteasome inhibitor MG132. PLoS One. 6:e202522011. View Article : Google Scholar : PubMed/NCBI

15. 

Korkor MT, Meng FB, Xing SY, Zhang MC, Guo JR, Zhu XX and Yang P: Microarray analysis of differential gene expression profile in peripheral blood cells of patients with human essential hypertension. Int J Med Sci. 8:168–179. 2011. View Article : Google Scholar : PubMed/NCBI

16. 

Roy Choudhury D, Small C, Wang Y, Mueller PR, Rebel VI, Griswold MD and McCarrey JR: Microarray-based analysis of cell-cycle gene expression during spermatogenesis in the mouse. Biol Reprod. 83:663–675. 2010.PubMed/NCBI

17. 

King HC and Sinha AA: Gene expression profile analysis by DNA microarrays: promise and pitfalls. JAMA. 286:2280–2288. 2001. View Article : Google Scholar : PubMed/NCBI

18. 

Zeng S and Gong Z: Expressed sequence tag analysis of expression profiles of zebrafish testis and ovary. Gene. 294:45–53. 2002. View Article : Google Scholar : PubMed/NCBI

19. 

Xiao P, Tang A, Yu Z, Gui Y and Cai Z: Gene expression profile of 2058 spermatogenesis-related genes in mice. Biol Pharm Bull. 31:201–206. 2008. View Article : Google Scholar : PubMed/NCBI

20. 

Lo LJ, Zhang ZH, Hong N, Peng JR and Hong YH: 3640 unique EST clusters from the medaka testis and their potential use for identifying conserved testicular gene expression in fish and mammals. PLoS One. 3:e39152008. View Article : Google Scholar : PubMed/NCBI

21. 

LI D and LU GX: Identification and expression of a novel human testis-specific gene by digital differential display. Chin Med J. 117:1791–1796. 2004.PubMed/NCBI

22. 

Sridhar K, Ross DT, Tibshirani R, Butte AJ and Greenberg PL: Relationship of differential gene expression profiles in CD34+ myelodysplastic syndrome marrow cells to disease subtype and progression. Blood. 114:4847–4858. 2009. View Article : Google Scholar : PubMed/NCBI

23. 

Creekmore AL, Silkworth WT, Cimini D, Jensen RV, Roberts PC and Schmelz EM: Changes in gene expression and cellular architecture in an ovarian cancer progression model. PLoS One. 6:e176762011. View Article : Google Scholar : PubMed/NCBI

24. 

Mattison J, Weyden L, Hubbard T and Adams DJ: Cancer gene discovery in mouse and man. Biochim Biophys Acta. 1796:140–161. 2009.PubMed/NCBI

25. 

Chen F, Zhu HH, Zhou LF, et al: Genes related to the very early stage of ConA-induced fulminant hepatitis: a gene-chip-based study in a mouse model. BMC Genomics. 11:2402010. View Article : Google Scholar : PubMed/NCBI

26. 

Eddy EM: Male germ cell gene expression. Recent Prog Horm Res. 57:103–128. 2002. View Article : Google Scholar : PubMed/NCBI

27. 

Davidson G: The cell cycle and Wnt. Cell Cycle. 9:1667–1668. 2010. View Article : Google Scholar : PubMed/NCBI

28. 

Chang H, Gao F, Guillou F, Taketo MM, Huff V and Behringer RR: Wt1 negatively regulates β-catenin signaling during testis development. Development. 135:1875–1885. 2008.

29. 

Olmeda D, Castel S, Vilaró S and Cano A: Beta-catenin regulation during the cell cycle: implications in G2/M and apoptosis. Mol Biol Cell. 14:2844–2860. 2003. View Article : Google Scholar : PubMed/NCBI

30. 

Peifer M and Polakis P: Wnt signalling in oncogenesis and embryogenesis: a look outside the nucleus. Science. 287:1606–1609. 2000. View Article : Google Scholar : PubMed/NCBI

31. 

Kimura T, Nakamura T, Murayama K, et al: The stabilization of beta-catenin leads to impaired primordial germ cell development via aberrant cell cycle progression. Dev Biol. 300:545–553. 2006. View Article : Google Scholar : PubMed/NCBI

32. 

Tetsu O and McCormick F: Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 398:422–426. 1999. View Article : Google Scholar : PubMed/NCBI

33. 

Botrugno OA, Fayard E, Annicotte JS, et al: Synergy between LRH-1 and beta-catenin induces G1 cyclin-mediated cell proliferation. Mol Cell. 15:499–509. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February 2013
Volume 31 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin Y, Liu Z, Liu X, Zhang Y, Rong Z and Li D: Microarray-based analysis of the gene expression profile in GC-1 spg cells transfected with spermatogenesis associated gene 12. Int J Mol Med 31: 459-466, 2013
APA
Lin, Y., Liu, Z., Liu, X., Zhang, Y., Rong, Z., & Li, D. (2013). Microarray-based analysis of the gene expression profile in GC-1 spg cells transfected with spermatogenesis associated gene 12. International Journal of Molecular Medicine, 31, 459-466. https://doi.org/10.3892/ijmm.2012.1225
MLA
Lin, Y., Liu, Z., Liu, X., Zhang, Y., Rong, Z., Li, D."Microarray-based analysis of the gene expression profile in GC-1 spg cells transfected with spermatogenesis associated gene 12". International Journal of Molecular Medicine 31.2 (2013): 459-466.
Chicago
Lin, Y., Liu, Z., Liu, X., Zhang, Y., Rong, Z., Li, D."Microarray-based analysis of the gene expression profile in GC-1 spg cells transfected with spermatogenesis associated gene 12". International Journal of Molecular Medicine 31, no. 2 (2013): 459-466. https://doi.org/10.3892/ijmm.2012.1225