Skip to main content

The Y-linked proto-oncogene TSPY contributes to poor prognosis of the male hepatocellular carcinoma patients by promoting the pro-oncogenic and suppressing the anti-oncogenic gene expression

Abstract

Background

Liver cancer is one of the major causes of cancer death worldwide, with significantly higher incidence and mortality among the male patients. Although sex hormones and their receptors could contribute to such sex differences, the story is incomplete. Genes on the male-specific region of the Y chromosome could play a role(s) in this cancer. TSPY is the putative gene for the gonadoblastoma locus on the Y chromosome (GBY) that is ectopically expressed in a subset of male hepatocellular carcinomas (HCCs). Although various studies showed that TSPY expression is associated with poor prognosis in the patients and its overexpression promotes cell proliferation of various cancer cell lines, it remains unclear how TSPY contributes to the clinical outcomes of the HCC patients. Identifying the downstream genes and pathways of TSPY actions would provide novel insights on its contribution(s) to male predominance in this deadly cancer.

Results

To determine the effects of TSPY on HCC, a TSPY transgene was introduced to the HCC cell line, HuH-7, and studied with RNA-Seq transcriptome analysis. The results showed that TSPY upregulates various genes associated with cell-cycle and cell-viability, and suppresses cell-death related genes. To correlate the experimental observations with those of clinical specimens, transcriptomes of male HCCs with high TSPY expression were analyzed with reference to those with silent TSPY expression from the Cancer Genome Atlas (TCGA). The comparative analysis identified 49 genes, which showed parallel expression patterns between HuH-7 cells overexpressing TSPY and clinical specimens with high TSPY expression. Among these 49 genes, 16 likely downstream genes could be associated with survival rates in HCC patients. The major upregulated targets were cell-cycle related genes and growth factor receptor genes, including CDC25B and HMMR, whose expression levels are negatively correlated with the patient survival rates. In contrast, PPARGC1A, SLC25A25 and SOCS2 were downregulated with TSPY expression, and possess favorable prognoses for HCC patients.

Conclusion

We demonstrate that TSPY could exacerbate the oncogenesis of HCC by differentially upregulate the expression of pro-oncogenic genes and downregulate those of anti-oncogenic genes in male HCC patients, thereby contributing to the male predominance in this deadly cancer.

Background

The testis specific protein Y-encoded (TSPY) is a tandemly repeated gene (> 30 times) located within the gonadoblastoma locus on the Y chromosome (GBY), which predisposes XY patients with disorders of sexual development (DSDs) to gonadoblastoma development [1,2,3]. TSPY encodes a protein harboring a SET/NAP domain, initially identified in the SET oncoprotein and nucleosome assembly proteins (NAPs) [4, 5]. SET/NAP proteins play important roles in transcriptional regulation, chromatin assembly, histone chaperone, and cell-cycle regulation [6,7,8,9,10]. Previously, we demonstrated that TSPY interacts with the cyclin-B/CDK1 complex, eukaryotic translation elongation factor 1A (EEF1A), and androgen receptor (AR) via its SET/NAP domain, and enhances the respective functions of these binding partners [11,12,13]. Overexpression of TSPY accelerates cell proliferation in various cancer cell lines [11, 13, 14]. Further, TSPY directly binds to the chromatin at its own exon-1 region and amplifies its own expression in a positive-feedback manner [15], suggesting the potential role of TSPY as a transcriptional regulator. Others showed that TSPY could suppress TP53 functions and enhance cell proliferation by inhibiting USP7, a protective deubiquitinase for TP53 [16]. TSPY is predominantly expressed at the early-stages of male germ cells, i.e. gonocytes and prespermatogonia at fetal testis [17] and spermatogonia and spermatocytes in adult testis [18]. It is postulated to participate in mitotic proliferation and meiotic division of male germ cells [19]. Importantly, TSPY is frequently expressed in germ cell tumors including gonadoblastoma, seminoma, and the carcinoma-in situ/intratubular germ cell neoplasia unclassified (CIS/ITHCNU), the precursor for all germ cell tumors [20,21,22,23,24]. Since TSPY is the primary gene located within GBY locus and is expressed in gonadoblastoma, TSPY is the putative gene for this oncogenic locus and could predispose dysfunctional germ cells to tumor development in dysgenetic gonads. Indeed, transgenic mouse studies showed that ectopic expression of TSPY in ovaries resulted in gonadoblastoma-like structures in female mice [25]. Significantly, TSPY is also expressed in various types of somatic cancer; including prostate cancer, lung cancer, and hepatocellular carcinoma (HCC) [26,27,28,29,30]. Accordingly, TSPY could also promote oncogenic initiation and/or progression of somatic cancers in male patients.

Liver cancer causes more than 700,000 cancer deaths each year worldwide [31, 32]. Significantly the incidence and mortality of HCC is much higher in males than females, with greater than threefold difference [32,33,34]. Both sex hormones and/or their receptors and the sex chromosome genes have been postulated to contribute to such sex differences, the mechanisms of which, however, could be quite complex [35,36,37,38,39,40]. As a proto-oncogene on the Y chromosome, TSPY is expressed in a subclass of male HCC cases (30–40%), frequently associated with the global hypomethylation of the genomic DNA, including its own promoter sequence [27, 28]. However, beside its general functions in cell proliferation and growth, the exact mechanism(s) by which TSPY exerts on the oncogenic processes in HCC patients remains unknown.

In order to determine the effects of TSPY expression on HCC, we overexpressed TSPY in the HCC cell line, HuH-7, and analyzed the differential gene expression patterns between HuH-7 cells expressing and non-expressing the TSPY transgene using transcriptome analysis, and correlated the results with those derived from comparative study between the male HCC cases with high TSPY expression and non-expression (silent) from the Cancer Genome Atlas (TCGA) [27, 41]. We identified 16 genes that were potentially regulated by TSPY and their differential expression could be correlated with the prognoses of the HCC patients. Among these genes, TSPY upregulates cell-cycle regulators and components of cell-division machinery, e.g. BUB1, CDC25B, CDC45, CENPA, PRC1, PRIM1, RRM2, and SPC24, as well as growth factor receptors, e.g. ADGRD1 and HMMR, which are associated with unfavorable prognosis and poor survivals of the patients while the expression of PPARGC1A, SLC25A25 and SOCS2 are inversely correlated with TSPY expression but are associated with favorable prognosis and better survival of the patients. Our results suggest that ectopic expression of the Y-located TSPY could promote the expression of pro-oncogenic genes and suppression of anti-oncogenic genes in HCC, thereby contributing to the male predominance of this deadly cancer.

Methods

Cell culture and lentiviruses

HuH-7 cells, a human hepatocellular carcinoma cell line, were cultured in DMEM medium containing 10% tet system-approved fetal bovine serum (FBS; Clontech) and antibiotics cocktail (100 U/mL penicillin and 100 μg/mL Streptomycin). Lentiviruses for the expression of TSPY and EGFP with tet-ON system were prepared as described previously [13, 42]. Cells were transduced with lentiviral particles containing the expression vectors, FUW-tetO-TSPY-ires-EGFP or FUW-tetO-EGFP with pFUW-M2rtTA. The ires sequences in the FUW-tetO-TSPY-ires-EGFP vector enable ribosome complex-binding and simultaneous expression of both TSPY and EGFP genes in a bicistronic cassette. The transduced cells were cultured in the absence of doxycycline (Dox) until analyses. The transduced cells were cultured in the presence of 1 μg/mL Dox (Sigma-Aldrich) to induce the expression of both TSPY and EGFP (Fig. 1a). For cell proliferation analyses, cells were seeded at 2000 cells/well in 96-well plates and cultured in the presence of 0.5 μg/mL Dox. The cell viability was monitored at the indicated time points using the CellTiter 96 Aqueous One Cell Proliferation Assay kit (Promega), according to the instructions from the manufacturer.

Fig. 1
figure 1

Overexpression of TSPY in HuH-7 cells using the Tet-ON system. a Schematic diagram of the Tet-ON system, in which the expression levels of TSPY and EGFP are upregulated by doxycycline (Dox) treatment via recruitment of the rtTA transcription activator onto the Tet-responsive promoter. b Confirmation of doxycycline-induced TSPY expression by western-blot analysis. β-actin was tested as a reference. c Immunofluorescence analysis showed that TSPY (red) and co-expressed EGFP (green) were expressed in the HuH-7-tetON-TSPY cells cultured with and without Dox. DNA was visualized by DAPI staining (blue). d Cell proliferation assay showing HuH-7-tetON-TSPY cells (red line) proliferated faster than HuH-7-tetO-EGFP cells (green line) in the presence of 1 µg/mL Dox

Western blot

Western-blot was performed as described previously [43], using anti-TSPY mouse monoclonal antibody (clone 17, generated in our laboratory) and anti-β-actin mouse monoclonal antibody (clone AC-15, Sigma-Aldrich). Immunoreactive signals were visualized by IRDye680RD conjugated anti-mouse IgG antibody, and recorded by the Odyssey system (LI-COR, Lincoln, NE).

Immunofluorescence

Immunofluorescence was performed as described previously [11]. Briefly, cells were fixed with 4% paraformaldehyde-PBS solution for 5 min, and permeabilized by methanol treatment. After blocking with 3% bovine serum albumin (Sigma)-PBS solution for 1 h, the slides were incubated with primary antibodies at 4 °C overnight. The primary antibodies used for immunofluorescence were anti-GFP goat IgG (Abcam) and anti-TSPY mouse monoclonal antibody (clone 17). The immunoreactive signals were visualized by Alexa Fluor 488 conjugated anti-goat IgG antibody (Invitrogen) and Alexa Fluor 594 conjugated anti-mouse IgG antibody respectively. Nuclear DNA was stained with 4′,6-diamidino-2-phenylindole (DAPI) (Roche Applied Science). Fluorescent images were recorded with a Nikon Eclipse Ti fluorescence microscope and image acquisition workstation (Nikon instrument Inc., Melville, NY).

RNA preparation and RNA-Seq transcriptome analysis of the transduced HuH-7 cells

Total RNA was isolated from the cells cultured in a 6-well plate (9.6 cm2 surface area) at 24 h after Dox-induction using TRIZOL-Plus RNA purification kit (ThermoFisher Scientific, NY). One μg total RNA from each triplicate sample was used for the library preparation using KAPA Stranded mRNA-Seq kit (Kapa Biosystems, MA). Libraries were indexed (bar-coded) with NEBNext multiplex primers for Illumina (New England Biolabs, CA). The libraries were subjected to 75 single-end read cycles of sequencing on the NextSeq 500 (Illumina, CA). All procedures were performed according to the manufacturer’s instructions.

The sequence reads were mapped onto the Ensembl GRCh37 (hg19) human reference genome using TopHat (version 1.0.1) [44], after quality assessment by FastQC program (version 0.11.4) [45]. The mapped reads were summarized and calculated to the count reads, which were then quantitated as the expression levels using the featureCounts (version 1.5.1) program [46]. Normalization and differential gene expression analysis were performed using an R package TCC [47].

Gene expression enrichment analysis, functional network analyses and pathway analyses were performed with Ingenuity Pathways Analysis (IPA) (Ingenuity Systems, build version 463341M) and DAVID bioinformatics resources [48]. A schematic diagram of cell cycle pathway was downloaded from KEGG website (https://www.genome.jp/kegg/) with permission [49].

Data set and data mining analysis of HCC specimens from TCGA

The RNA-Seq gene expression data and associated clinical information of HCC cases at the Cancer Genome Atlas (TCGA) data portal were downloaded from the UCSC Xena Browser [50]. The datasets included 50 non-tumor samples (28 male cases and 22 female cases) and 371 tumor samples (250 male cases and 121 female cases). The expression levels were calculated as RSEM normalized read count [51]. The survival information of the respective HCC patients was obtained from the Human Protein Atlas (HPA) data portal [52], except for classification of the high TSPY-expressing patients and the TSPY-silent patients. Statistical analyses were performed with the Prism6 program (GraphPad Software, Inc., La Jolla, CA).

Results

TSPY-overexpression enhances cell proliferation in HuH-7 cells

The effects of TSPY in HCC were examined in the HCC HuH-7 cells, which were transduced with Tet-On lentiviral vectors overexpressing TSPY-ires-EGFP and EGFP alone under doxycycline-induced conditions, hereby designated as HuH-7-tetON-TSPY and HuH-7-tetON-EGFP respectively (Fig. 1a). HuH-7-tetON-EGFP cells served as a control for all experiments in the study. Western-blot analyses confirmed the expression of TSPY and EGFP being appropriately induced by Dox in the culture media (Fig. 1b). Immunocytochemical analyses showed that the TSPY protein was localized in both cytoplasm and nuclei (Fig. 1c), consistently with the observations in clinical HCC samples [28]. Cell proliferation assays showed that overexpression of TSPY promoted cell proliferation in HuH-7 cells (Fig. 1d), as previously reported on other cancer cell types [13, 14, 16].

TSPY upregulated various cell-cycle related genes and suppressed tumor suppressor genes in HuH-7 cells

To explore the mechanisms by which TSPY regulates the molecular events in HuH-7 cells, we had performed a transcriptome analysis using the Illumina RNA-Seq platform. The mRNAs were isolated from respective HuH-7 cells (n = 3) overexpressing TSPY and EGFP alone at 24 h after Dox administration, and analyzed in biological triplicates with RNA-Seq strategy [13]. The differentially expressed genes (DEGs) between HuH-7-tetON-TSPY and HuH-7-tetON-EGFP cells were identified with false discovery rate (FDR) < 0.05 by the TCC analysis, Student’s t-test P-value < 0.05, expression level of log2[expression read count] > 3.32, and Log2[fold change] > 0.6, resulting in a total of 1884 DEGs consisting of 1139 up-regulated genes and 745 down-regulated genes (Fig. 2a and Additional file 1: Table S1). The DEGs were subsequently mapped onto the KEGG pathway, representing molecular interaction and relation network, using the DAVID gene-annotation enrichment analysis software [48, 49]. The results showed that the pathways of metabolic pathways, steroid biosynthesis, glycosaminoglycan degradation, cell cycle, and bile secretion, were enriched at Benjamini–Hochberg FDR < 0.05, suggesting that these pathways could be affected by TSPY overexpression in HuH-7 cells (Fig. 2b).

Fig. 2
figure 2

Transcriptome and DAVID analyses of the HuH-7-tetOn-TSPY cells. a MA-plots showing the differentially expressed genes (DEGs) between HuH-7-tetOn-TSPY cells and HuH-7-tetOn-EGFP cells. Genes plotted within the areas of Log2[expression level] > 3.32 and |log2[fold change]| > 0.6 (red) were analyzed as differentially expressed genes (DEGs). b DAVID gene-annotation enrichment analysis identified top 5 KEGG pathways enriched in DEGs with FDR < 0.05; FDR = false discovery rate or corrected P-value. c Mapping of the 26 DEGs on the KEGG cell-cycle pathway [49]. Red indicates the upregulated genes and green indicates the downregulated genes in the Dox-treated HuH-7-tetON-TSPY cells

Using a set of DEGs derived from a more stringent expression level, we further analyzed the TSPY effects with the Ingenuity Pathways Analysis (IPA) program [53, 54]. Our results showed that the biological functions remarkably affected by TSPY overexpression were pathways associated with cell-cycle and cell viability (Additional file 2: Figure S1a and S1b). Noticeably, the pathways of necrosis and cell-death were inhibited (Additional file 2: Figure S1b, blue columns) while the pathways associated with cellular interphase processes and cell-survival were enhanced (Additional file 2: Figure S1b, red columns), as indicated by the |z-scores of − and + |> 2 for significance in inhibition and activation respectively. The IPA hepatotoxicity analysis demonstrated that the TSPY-induced DEGs were significantly associated with liver cancer development (Additional file 2: Figure S1c). Altogether, both gene annotation/ontology programs consistently suggested that the pathways of cell-cycle and cell viability could be mostly affected by TSPY overexpression in HuH-7 cells. To further illustrate the various cell cycle genes being affected by TSPY, we mapped the associated DEGs onto the KEGG cell-cycle pathway [48, 49]. Our results showed that the tumor suppressors CDKN1A (also known as Cip1 and p21) and GADD45 were downregulated while various positive cell-cycle regulators and components for DNA replication, including cyclin-B, cyclin-D, CDK1, CDC25B, and MCMs, were upregulated by TSPY-overexpression (Fig. 2c).

Overall, the transcriptome analysis of the HuH-7 cells overexpressing TSPY showed that TSPY could promote cell proliferation and viability by directly or indirectly augmenting a pro-oncogenic gene expression pattern consistent with the results of cell-proliferation assay (Fig. 1d).

High expression of TSPY is associated with poor survival rate in male HCC patients

To correlate the effects of TSPY on the gene expression patterns and clinical outcomes in the HCC patients, we had examined the transcriptomes and the corresponding clinical data of the HCC samples from the Cancer Genome Atlas (TCGA) [27, 41]. Among the 250 male HCC cases, the top 34 cases expressed TSPY at the highest level (expression read count > 50) and were classified as the male TSPY-high group, and 180 cases without TSPY expression were classified as the male TSPY-silent group (Fig. 3a). The survival data showed that the survival rate of the male TSPY-high group was significantly lower than that of male TSPY-silent group (Log-rank test P-value = 0.0005) (Fig. 3b), suggesting that TSPY could be directly or indirectly involved in the progression of HCC and survival of the patients. Further and importantly, the survival rate of the male TSPY-high group was significantly lower than that of female group (Log-rank test P-value = 0.0164), while there was not significant difference between male TSPY-silent group and female group (Log-rank test P-value = 0.0671) (Fig. 3b).

Fig. 3
figure 3

Identification of the TSPY downstream genes associated with clinical outcomes in primary HCC. a Based on the TSPY expression level, male HCC cases were classified into the TSPY-high group (n = 36, expression count > 50) and the TSPY-silent group (n = 180, expression count = 0). b Survival curves showing that the survival rates of the male TSPY-high group (red), male TSPY-silent group (blue), and female group (gray). Log-rank test P-values are indicated. c Volcano plot representing the DEGs between TSPY-high and TSPY-silent HCC groups (red plots). d The diagram shows the workflow to identify the downstream genes regulated by TSPY in HCC tissues. The genes whose expression levels consistently correlated with the TSPY expression level in both HuH-7 cells and clinical TSPY-high HCC tissue samples were selected as described in the body text. Sixteen genes (red), whose expression patterns correlated with that of TSPY, were identified to be associated with clinical outcomes.

Correlation of the differentially expressed genes between the TSPY overexpression in HuH-7 cells and TSPY-high group male HCC patients

To identify the common DEGs associated with both TSPY overexpression in HuH-7 cells and TSPY high expression in HCC patients, we had analyzed the differential gene expression patterns of HuH-7 cells overexpressing TSPY versus EGFP and those of TSPY-high group versus TSPY-silent group in clinical HCC tissue samples. The datamining of the TCGA datasets identified 692 DEGs with a cut-off differential expression level at |log2[difference]| > 0.6 and a P value < 0.05 between the male TSPY-high group and the male TSPY-silent group of HCC (Fig. 3c and Additional file 3: Table S2). By comparing these DEGs in HCCs to those derived from HuH-7 overexpressing TSPY (Fig. 2a), 49 DEGs were identified to share similar expression patterns, i.e. either up or down, between HuH-7 overexpressing TSPY and TSPY-high group in clinical HCC specimens (Fig. 3d and Additional file 4: Table S3). To further identify those potentially play important roles in HCC, these 49 genes were further analyzed with respect to the correlation of their expression patterns with the mortality of HCC patients (Fig. 3d) [52]. Such additional analysis identified 16 DEGs whose expression levels were associated with the survival rates of HCC patients (Fig. 3d and Table 1). Three genes, PPARGC1A, SLC25A25 (also known as MCSC), and SOCS2, associated with favorable prognoses and good patient survivals were downregulated by TSPY. PPARGC1A and SOCS2 are considered to be tumor suppressors in liver cancer [55,56,57,58,59], while SLC25A members are involved in mitochondrial Ca2+ signaling important for hepatocyte physiology [60, 61]. The expression patterns of the remainder 13 genes were unfavorable for patient survival and were upregulated by TSPY (Table 1, and Figs. 3, 4, Additional file 5: Figure S2, and Additional file 6: Figure S3). These pro-oncogenic genes included various cell-cycle related genes, e.g. BUB1, CDC25B, CDC45, CENPA, PRC1, PRIM1, RRM2, and SPC24, that participate in DNA replication and G2/M-phases progression respectively (Table 1 and Additional file 7: Table S4), which are consistent with our previous observations that TSPY accelerates G2/M-transition [14] and enhances the mitotic cyclin B-CDK1 phosphorylation activities [12, 62]. In addition, growth factor receptors, e.g. ADGRD1 (also known as GPR133) and HMMR (also known as RHAMM), and components of DNA repair machinery, e.g. EXO1 and RAD51, were also upregulated by TSPY and negatively associated with patient survival (Table 1 and Figs. 3, 4, Additional file 5: Figure S2, and Additional file 6: Figure S3). These results suggest that the ectopically expressed TSPY could exacerbate oncogenesis and progression in HCC by stimulating downstream unfavorable (pro-oncogenic) but repressing favorable (anti-oncogenic) gene expression in the tumors.

Table 1 Sixteen downstream genes potentially regulated by TSPY and associated with prognosis in HCC patients
Fig. 4
figure 4

Representative expression patterns and associated with survival rates of TSPY downstream genes in HCC. a TSPY stimulated the expression levels of CDC25B, HMMR, and PPARGC1A, in HuH-7 cells. The results of transcriptome analysis (n = 3) were presented with the P-values by Student t-tests. b–d Left panels, expression levels of TSPY and selected downstream genes, e.g. CDC25B, HMMR, and PPARGC1A, among female non-tumor liver (NT), female HCC, male non-tumor liver (NT), male TSPY-silent HCC (TS(−)), and male TSPY-high HCC (TS(++)), in the TCGA transcriptome dataset. Y-axis indicates the expression level as RSEN normalized count. Right panels, survival analyses of the TCGA data, comparing the high-expresser HCC patients (red lines) and the low-expresser HCC patients (blue lines) for the respective genes. Both male and female cases were included in these analyses. Log-rank test P-values are indicated. Abbreviations; **t-test P-value < 0.005; *P-value < 0.05; nd, P-value > 0.05. See Additional file 5: Figure S2 and Additional file 6: Figure S3 for same analyses for all 16 potential TSPY downstream genes

TSPY promotes pro-oncogenic gene expression and poor survival of male HCC patients

Comparison of the gene expression levels among respective TSPY-related groups showed that the expression levels of the unfavorable/pro-oncogenic genes, e.g. ADGRD1, CDC25B, CDC45, EXO1, HMMR, PRIM1, RAD51, RRM2, and SPC24, were significantly higher in the male TSPY-high group HCCs than female HCCs, while the expression level of the favorable/anti-oncogenic gene, e.g. PPARGC1A, was significantly lower in the male TSPY-high group HCCs than the female HCCs (Fig. 4 and Additional file 6: Figure S3). However, there was no significant difference in the expression levels of the 16 downstream genes between the male TSPY-silent group HCCs and female HCCs, except EGLN3 and SPC24 (Fig. 4 and Additional file 6: Figure S3). These results suggest that TSPY could differentially affect the expression levels of pro-oncogenic and unfavorable genes in male HCCs, as compared to those in female HCCs. Indeed, as described above, the survival data indicate that, while there was no significant difference between female HCC group and the male TSPY-silent HCC group, the survival ratio of the male TSPY-high HCC group was significantly lower than that of female HCC group (Log-rank test P-value = 0.0164) with 3 years survival ratio of 35% in the male TSPY-high HCC group and 60–70% in female HCC and the male TSPY-silent HCC groups respectively (Fig. 3b). These observations support the hypothesis that TSPY could be a male-specific genetic risk factor and its aberrant and epigenetic activation contributes to the oncogenic processes and the male predominance of HCC.

Discussion

Recent studies have provided significant evidence demonstrating the sex differences in various diseases, including cardiovascular, inflammatory and neurodegenerative diseases, as well as numerous types of cancer [38, 63,64,65,66,67,68,69]. Currently, the mechanisms contributing to such sex differences are still largely unknown. Liver cancer is one of the most male predominant cancers with men having approximately three to sixfolds higher incidence and mortality than women, depending on the geographic locations [32,33,34]. The sex hormones and their receptors have been postulated to play key roles in such sex difference in HCC [39, 40, 70, 71]. Female hormones, i.e. estrogen and prolactin, have been proposed to suppress HCC development by reducing the activities of inflammatory pathways, such as IL-6 and TNF signaling cascades in liver [36, 37, 72]. In contrast, male hormones, i.e. androgens, could stimulate hepatitis B virus replication and expression of the HBV viral oncoprotein HBx via androgen receptor actions, resulting in exacerbation of HCC development [73,74,75,76,77]. Since it is estimated that 50–80% of HCC patients could be associated with HBV-infection, the androgen-dependent upregulation of the viral oncoprotein HBx might contribute to the male predominance of HCC. Notably, sex differences in HCC still observed between men and women after menopause age, and various clinical trials targeting androgen receptor showed limited effects on HCC progression [33, 78]. Hence, in addition to sex hormones and their receptors, genetic factors could also be important for the male predominance of HCC. Such genetic factors include single nucleotide polymorphisms (SNPs) on autosomal genes correlating with the sex difference of HCC [79, 80]. Further, we demonstrated that an X-encoded tumor suppressor TSPX, the homologue of TSPY on the X chromosome, could bind to and promote the proteasomal degradation of the viral oncoprotein HBx [81]. Since men have only one X-chromosome, inactivation and/or mutation of such tumor suppressor gene could increase the risk of HBx-medicated HCC in men [38, 81].

TSPY is a male specific proto-oncogene on the Y chromosome and is ectopically expressed in a subclass of male HCC cases (30–40% of cases) [27, 28, 30]. Although such TSPY expression could be related to its pro-oncogenic functions, such as acceleration of cell proliferation, stimulation of protein synthesis and cell growth, suppression of TP53 functions, and exacerbation on androgen receptor transactivation [11,12,13,14, 16], its downstream molecular effects have not been fully explored. We previously identified the TSPY co-expression network genes that are frequently upregulated together with TSPY in multiple somatic cancers, including HCC, and are associated with global DNA hypomethylation [27]. Using transcriptome analyses on the HuH-7 HCC cells overexpressing TSPY and those of clinical HCC specimens from the TCGA database, we identified 16 genes that were likely regulated directly or indirectly by TSPY in HCC cells and contributed to poor prognosis of HCC patients (Table 1, and Fig. 4 and Additional file 5: Figure S2). These TSPY-regulated genes include numerous cell-cycle related genes and growth factor receptors (Table 1 and Fig. 5). Importantly, the expression levels of the unfavorable genes, e.g. ADGRD1, CDC25B, CDC45, EXO1, HMMR, PRIM1, RAD51, RRM2, and SPC24, were significantly higher in the male TSPY-high group HCCs than TSPY-silent/female group HCCs, while the expression level of the favorable gene, e.g. PPARGC1A, was significantly lower in the male TSPY-high group HCCs than female HCCs (Fig. 4 and Additional file 6: Figure S3). Previous studies demonstrated the oncogenic properties of ADGRD1, CDC25B, HMMR, PRIM1, RAD51, RRM2, and SPC24, in HCC and/or other somatic cancers [82,83,84,85,86,87,88,89,90,91] (Additional file 7: Table S4). Indeed, the small molecule inhibitors against CDC25 and RRM2 significantly inhibited cell proliferation in HuH-7 cells, and the TSPY-overexpression relieved the effects of these inhibitors (Additional file 8: Figure S4). Our results strongly suggest that TSPY is a male-specific genetic factor that exacerbates HCC oncogenesis by stimulating the expression of these downstream pro-oncogenic genes, thereby contributing to the male predominance of HCC. Such postulation is supported by the fact that the survival rate of the male TSPY-high HCC group was significantly lower than those of male TSPY-silent and female HCC groups (Fig. 3b).

Fig. 5
figure 5

Schematic diagram illustrating the functions of TSPY and TSPY-downstream genes in cell cycle events, particularly mitosis, in HCC. From top-left; CDC45, RRM2 and PRIM1 participate in DNA replication at S-phase; CDC25B activates cyclin-B/CDK1 complex and promotes M-phase entry. TSPY binds cyclin-B/CDK1 complex and enhances its activity. BUB1, CENPA, and SPC24 interact with the condensed chromatin and participate in spindle assembly checkpoint and chromatin segregation respectively. HMMR promotes spindle pole formation; PRC1 binds microtubules and modulates cytokinesis. HMMR also activates the signaling cascades of cell-proliferation via extracellular hyaluronan

HMMR was originally identified as the receptor for hyaluronan (HA), a component of extracellular matrix, and it is frequently upregulated in various cancer types, including colon cancer, bladder cancer, prostate cancer, and liver cancer [83, 92,93,94,95]. The high HMMR expression correlates with cancer growth, metastasis, and poor prognosis in these types of cancer [83, 92,93,94,95]. The level of extracellular HA is elevated according to the progression of liver fibrosis, which is a pathological precondition for HCC [96]. The intracellular HMMR participates in mitotic spindle pole formation and cytokinesis [97]. The identification of HMMR as a downstream gene upregulated by TSPY suggests that it could be involved in the initiation and progression of HCC via its stimulation of the HA-HMMR signaling cascade, in addition to oncogenic activities as noted above.

Conclusion

The present study shows that TSPY is a male-specific genetic risk factor, whose ectopic expression could contribute partially to the male predominance of HCC. The ectopically expressed TSPY upregulates the expression of genes involved in cell-cycle progression, especially G2/M-phase, in HCC. In addition, TSPY may promote liver fibrosis and expedite an oncogenic transition to HCC via activation of the HA-HMMR signaling cascade, among others. Future studies on the mechanisms of TSPY-mediated differential regulation of its downstream genes will provide novel insights on its oncogenic actions, prognostic values and potential therapeutic targets for effective clinical treatments of HCC.

References

  1. Salo P, Kaariainen H, Petrovic V, Peltomaki P, Page DC, de la Chapelle A. Molecular mapping of the putative gonadoblastoma locus on the Y chromosome. Genes Chromosom Cancer. 1995;14(3):210–4.

    Article  CAS  PubMed  Google Scholar 

  2. Tsuchiya K, Reijo R, Page DC, Disteche CM. Gonadoblastoma: molecular definition of the susceptibility region on the Y chromosome. Am J Hum Genet. 1995;57(6):1400–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Skaletsky H, Kuroda-Kawaguchi T, Minx PJ, Cordum HS, Hillier L, Brown LG, Repping S, Pyntikova T, Ali J, Bieri T, et al. The male-specific region of the human Y chromosome is a mosaic of discrete sequence classes. Nature. 2003;423(6942):825–37.

    Article  CAS  PubMed  Google Scholar 

  4. Schnieders F, Dork T, Arnemann J, Vogel T, Werner M, Schmidtke J. Testis-specific protein, Y-encoded (TSPY) expression in testicular tissues. Hum Mol Genet. 1996;5(11):1801–7.

    Article  CAS  PubMed  Google Scholar 

  5. Zhang JS, Yang-Feng TL, Muller U, Mohandas TK, de Jong PJ, Lau YF. Molecular isolation and characterization of an expressed gene from the human Y chromosome. Hum Mol Genet. 1992;1(9):717–26.

    Article  CAS  PubMed  Google Scholar 

  6. Lau YF. Gonadoblastoma, testicular and prostate cancers, and the TSPY gene. Am J Hum Genet. 1999;64(4):921–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chae YC, Kim KB, Kang JY, Kim SR, Jung HS, Seo SB. Inhibition of FoxO1 acetylation by INHAT subunit SET/TAF-Ibeta induces p21 transcription. FEBS Lett. 2014;588(17):2867–73.

    Article  CAS  PubMed  Google Scholar 

  8. D’Arcy S, Martin KW, Panchenko T, Chen X, Bergeron S, Stargell LA, Black BE, Luger K. Chaperone Nap1 shields histone surfaces used in a nucleosome and can put H2A-H2B in an unconventional tetrameric form. Mol Cell. 2013;51(5):662–77.

    Article  PubMed  CAS  Google Scholar 

  9. Loyola A, Almouzni G. Histone chaperones, a supporting role in the limelight. Biochim Biophys Acta. 2004;1677(1–3):3–11.

    Article  CAS  PubMed  Google Scholar 

  10. Seo SB, McNamara P, Heo S, Turner A, Lane WS, Chakravarti D. Regulation of histone acetylation and transcription by INHAT, a human cellular complex containing the set oncoprotein. Cell. 2001;104(1):119–30.

    Article  CAS  PubMed  Google Scholar 

  11. Kido T, Lau YF. The human Y-encoded testis-specific protein interacts functionally with eukaryotic translation elongation factor eEF1A, a putative oncoprotein. Int J Cancer. 2008;123(7):1573–85.

    Article  CAS  PubMed  Google Scholar 

  12. Li Y, Lau YF. TSPY and its X-encoded homologue interact with cyclin B but exert contrasting functions on cyclin-dependent kinase 1 activities. Oncogene. 2008;27(47):6141–50.

    Article  CAS  PubMed  Google Scholar 

  13. Li Y, Zhang DJ, Qiu Y, Kido T, Lau YC. The Y-located proto-oncogene TSPY exacerbates and its X-homologue TSPX inhibits transactivation functions of androgen receptor and its constitutively active variants. Hum Mol Genet. 2017;26(5):901–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Oram SW, Liu XX, Lee TL, Chan WY, Lau YF. TSPY potentiates cell proliferation and tumorigenesis by promoting cell cycle progression in HeLa and NIH3T3 cells. BMC Cancer. 2006;6:154.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Kido T, Lau YF. The Y-located gonadoblastoma gene TSPY amplifies its own expression through a positive feedback loop in prostate cancer cells. Biochem Biophys Res Commun. 2014;446(1):206–11.

    Article  CAS  PubMed  Google Scholar 

  16. Shen Y, Tu W, Liu Y, Yang X, Dong Q, Yang B, Xu J, Yan Y, Pei X, Liu M, et al. TSPY1 suppresses USP7-mediated p53 function and promotes spermatogonial proliferation. Cell Death Dis. 2018;9(5):542.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Honecker F, Stoop H, de Krijger RR, Chris Lau YF, Bokemeyer C, Looijenga LH. Pathobiological implications of the expression of markers of testicular carcinoma in situ by fetal germ cells. J Pathol. 2004;203(3):849–57.

    Article  CAS  PubMed  Google Scholar 

  18. Kido T, Lau YF. A Cre gene directed by a human TSPY promoter is specific for germ cells and neurons. Genesis. 2005;42(4):263–75.

    Article  CAS  PubMed  Google Scholar 

  19. Lau YF, Li Y, Kido T. Role of the Y-located putative gonadoblastoma gene in human spermatogenesis. Syst Biol Reprod Med. 2011;57(1–2):27–34.

    Article  CAS  PubMed  Google Scholar 

  20. Honecker F, Stoop H, Mayer F, Bokemeyer C, Castrillon DH, Lau YF, Looijenga LH, Oosterhuis JW. Germ cell lineage differentiation in non-seminomatous germ cell tumours. J Pathol. 2006;208(3):395–400.

    Article  CAS  PubMed  Google Scholar 

  21. Kersemaekers AM, Honecker F, Stoop H, Cools M, Molier M, Wolffenbuttel K, Bokemeyer C, Li Y, Lau YF, Oosterhuis JW, et al. Identification of germ cells at risk for neoplastic transformation in gonadoblastoma: an immunohistochemical study for OCT3/4 and TSPY. Hum Pathol. 2005;36(5):512–21.

    Article  CAS  PubMed  Google Scholar 

  22. Lau Y, Chou P, Iezzoni J, Alonzo J, Komuves L. Expression of a candidate gene for the gonadoblastoma locus in gonadoblastoma and testicular seminoma. Cytogenet Cell Genet. 2000;91(1–4):160–4.

    Article  CAS  PubMed  Google Scholar 

  23. Li Y, Tabatabai ZL, Lee TL, Hatakeyama S, Ohyama C, Chan WY, Looijenga LH, Lau YF. The Y-encoded TSPY protein: a significant marker potentially plays a role in the pathogenesis of testicular germ cell tumors. Hum Pathol. 2007;38(10):1470–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Li Y, Vilain E, Conte F, Rajpert-De Meyts E, Lau YF. Testis-specific protein Y-encoded gene is expressed in early and late stages of gonadoblastoma and testicular carcinoma in situ. Urol Oncol. 2007;25(2):141–6.

    Article  CAS  PubMed  Google Scholar 

  25. Kido T, Schubert S, Schmidtke J, Lau YF. Expression of the human TSPY gene in the brains of transgenic mice suggests a potential role of this Y chromosome gene in neural functions. J Genet Genom. 2011;38:181–92.

    Article  CAS  Google Scholar 

  26. Kido T, Hatakeyama S, Ohyama C, Lau YF. Expression of the Y-encoded TSPY is associated with progression of prostate cancer. Genes (Basel). 2010;1(2):283–93.

    Article  CAS  Google Scholar 

  27. Kido T, Lau YC. Identification of a TSPY co-expression network associated with DNA hypomethylation and tumor gene expression in somatic cancers. J Genet Genom. 2016;43(10):577–85.

    Article  Google Scholar 

  28. Kido T, Lo RC, Li Y, Lee J, Tabatabai ZL, Ng IO, Lau YF. The potential contributions of a Y-located protooncogene and its X homologue in sexual dimorphisms in hepatocellular carcinoma. Hum Pathol. 2014;45(9):1847–58.

    Article  CAS  PubMed  Google Scholar 

  29. Lau YF, Lau HW, Komuves LG. Expression pattern of a gonadoblastoma candidate gene suggests a role of the Y chromosome in prostate cancer. Cytogenet Genome Res. 2003;101(3–4):250–60.

    Article  CAS  PubMed  Google Scholar 

  30. Yin YH, Li YY, Qiao H, Wang HC, Yang XA, Zhang HG, Pang XW, Zhang Y, Chen WF. TSPY is a cancer testis antigen expressed in human hepatocellular carcinoma. Br J Cancer. 2005;93(4):458–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. World Health Organization. https://www.who.int/cancer/resources/keyfacts/en/. Accessed 23 Nov 2018.

  32. Global Burden of Disease Cancer C, Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, Allen C, Alsharif U, Alvis-Guzman N, Amini E, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016: a systematic analysis for the global burden of disease study. JAMA Oncol. 2018;4(11):1553–68.

    Article  Google Scholar 

  33. Liu P, Xie SH, Hu S, Cheng X, Gao T, Zhang C, Song Z. Age-specific sex difference in the incidence of hepatocellular carcinoma in the United States. Oncotarget. 2017;8(40):68131–7.

    PubMed  PubMed Central  Google Scholar 

  34. Wong MC, Jiang JY, Goggins WB, Liang M, Fang Y, Fung FD, Leung C, Wang HH, Wong GL, Wong VW, et al. International incidence and mortality trends of liver cancer: a global profile. Sci Rep. 2017;7:45846.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cheung OK, Cheng AS. Gender differences in adipocyte metabolism and liver cancer progression. Front Genet. 2016;7:168.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Hartwell HJ, Petrosky KY, Fox JG, Horseman ND, Rogers AB. Prolactin prevents hepatocellular carcinoma by restricting innate immune activation of c-Myc in mice. Proc Natl Acad Sci USA. 2014;111(31):11455–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM, Karin M. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 2007;317(5834):121–4.

    Article  CAS  PubMed  Google Scholar 

  38. Clocchiatti A, Cora E, Zhang Y, Dotto GP. Sexual dimorphism in cancer. Nat Rev Cancer. 2016;16(5):330–9.

    Article  CAS  PubMed  Google Scholar 

  39. Ma WL, Lai HC, Yeh S, Cai X, Chang C. Androgen receptor roles in hepatocellular carcinoma, fatty liver, cirrhosis and hepatitis. Endocr Relat Cancer. 2014;21(3):R165–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yeh SH, Chen PJ. Gender disparity of hepatocellular carcinoma: the roles of sex hormones. Oncology. 2010;78(Suppl 1):172–9.

    Article  CAS  PubMed  Google Scholar 

  41. Tomczak K, Czerwinska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol (Pozn). 2015;19(1A):A68–77.

    Google Scholar 

  42. Hockemeyer D, Soldner F, Cook EG, Gao Q, Mitalipova M, Jaenisch R. A drug-inducible system for direct reprogramming of human somatic cells to pluripotency. Cell Stem Cell. 2008;3(3):346–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kido T, Sun Z, Lau YC. Aberrant activation of the human sex-determining gene in early embryonic development results in postnatal growth retardation and lethality in mice. Sci Rep. 2017;7(1):4113.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25(9):1105–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. FastQC: a quality control tool for high throughput sequence data. http://www.bioinformatics.babraham.ac.uk/projects/fastqc/. Accessed 28 Mar 2017.

  46. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30.

    Article  CAS  PubMed  Google Scholar 

  47. Sun J, Nishiyama T, Shimizu K, Kadota K. TCC: an R package for comparing tag count data with robust normalization strategies. BMC Bioinform. 2013;14:219.

    Article  Google Scholar 

  48. Huang DW, Sherman BT, Tan Q, Kir J, Liu D, Bryant D, Guo Y, Stephens R, Baseler MW, Lane HC, et al. DAVID bioinformatics resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 2007;35(Web Server issue):W169–75.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45(D1):D353–61.

    Article  CAS  PubMed  Google Scholar 

  50. Goldman M, Craft B, Kamath A, Brooks AN, Zhu J, Haussler D. The UCSC Xena platform for cancer genomics data visualization and interpretation. bioEziv. 2018. https://doi.org/10.1101/326470.

    Article  Google Scholar 

  51. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323.

    Article  CAS  Google Scholar 

  52. Uhlen M, Zhang C, Lee S, Sjostedt E, Fagerberg L, Bidkhori G, Benfeitas R, Arif M, Liu Z, Edfors F, et al. A pathology atlas of the human cancer transcriptome. Science. 2017;357(6352):eaan2507.

    Article  PubMed  CAS  Google Scholar 

  53. Kramer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics. 2014;30(4):523–30.

    Article  PubMed  CAS  Google Scholar 

  54. Li Y, Zheng M, Lau YF. The sex-determining factors SRY and SOX9 regulate similar target genes and promote testis cord formation during testicular differentiation. Cell Rep. 2014;8(3):723–33.

    Article  CAS  PubMed  Google Scholar 

  55. Liu R, Zhang H, Zhang Y, Li S, Wang X, Wang X, Wang C, Liu B, Zen K, Zhang CY, et al. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha acts as a tumor suppressor in hepatocellular carcinoma. Tumour Biol. 2017;39(4):1010428317695031.

    Article  PubMed  Google Scholar 

  56. Wang X, Liao Z, Bai Z, He Y, Duan J, Wei L. MiR-93-5p Promotes cell proliferation through down-regulating PPARGC1A in hepatocellular carcinoma cells by bioinformatics analysis and experimental verification. Genes (Basel). 2018;9(1):51.

    Article  CAS  Google Scholar 

  57. Qiu X, Zheng J, Guo X, Gao X, Liu H, Tu Y, Zhang Y. Reduced expression of SOCS2 and SOCS6 in hepatocellular carcinoma correlates with aggressive tumor progression and poor prognosis. Mol Cell Biochem. 2013;378(1–2):99–106.

    Article  CAS  PubMed  Google Scholar 

  58. Cui M, Sun J, Hou J, Fang T, Wang X, Ge C, Zhao F, Chen T, Xie H, Cui Y, et al. The suppressor of cytokine signaling 2 (SOCS2) inhibits tumor metastasis in hepatocellular carcinoma. Tumour Biol. 2016;37(10):13521–31.

    Article  CAS  PubMed  Google Scholar 

  59. Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee JM, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018;67(6):2254–70.

    Article  CAS  PubMed  Google Scholar 

  60. Mashima H, Ueda N, Ohno H, Suzuki J, Ohnishi H, Yasuda H, Tsuchida T, Kanamaru C, Makita N, Iiri T, et al. A novel mitochondrial Ca2+-dependent solute carrier in the liver identified by mRNA differential display. J Biol Chem. 2003;278(11):9520–7.

    Article  CAS  PubMed  Google Scholar 

  61. Pathak T, Trebak M. Mitochondrial Ca(2+) signaling. Pharmacol Ther. 2018;192:112–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lau YF, Li Y, Kido T. Gonadoblastoma locus and the TSPY gene on the human Y chromosome. Birth Defects Res C Embryo Today. 2009;87(1):114–22.

    Article  CAS  PubMed  Google Scholar 

  63. Casimir GJ, Mulier S, Hanssens L, Zylberberg K, Duchateau J. Gender differences in inflammatory markers in children. Shock. 2010;33(3):258–62.

    Article  CAS  PubMed  Google Scholar 

  64. Gillies GE, Pienaar IS, Vohra S, Qamhawi Z. Sex differences in Parkinson’s disease. Front Neuroendocrinol. 2014;35(3):370–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Maan AA, Eales J, Akbarov A, Rowland J, Xu X, Jobling MA, Charchar FJ, Tomaszewski M. The Y chromosome: a blueprint for men’s health? Eur J Hum Genet. 2017;25(11):1181–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mosca L, Barrett-Connor E, Wenger NK. Sex/gender differences in cardiovascular disease prevention: what a difference a decade makes. Circulation. 2011;124(19):2145–54.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Rathod KS, Kapil V, Velmurugan S, Khambata RS, Siddique U, Khan S, Van Eijl S, Gee LC, Bansal J, Pitrola K, et al. Accelerated resolution of inflammation underlies sex differences in inflammatory responses in humans. J Clin Invest. 2017;127(1):169–82.

    Article  PubMed  Google Scholar 

  68. Regitz-Zagrosek V, Kararigas G. Mechanistic pathways of sex differences in cardiovascular disease. Physiol Rev. 2017;97(1):1–37.

    Article  PubMed  Google Scholar 

  69. Pike CJ. Sex and the development of Alzheimer’s disease. J Neurosci Res. 2017;95(1–2):671–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kalra M, Mayes J, Assefa S, Kaul AK, Kaul R. Role of sex steroid receptors in pathobiology of hepatocellular carcinoma. World J Gastroenterol. 2008;14(39):5945–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li Z, Tuteja G, Schug J, Kaestner KH. Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell. 2012;148(1–2):72–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Naugler WE, Karin M. The wolf in sheep’s clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med. 2008;14(3):109–19.

    Article  CAS  PubMed  Google Scholar 

  73. Chiu CM, Yeh SH, Chen PJ, Kuo TJ, Chang CJ, Chen PJ, Yang WJ, Chen DS. Hepatitis B virus X protein enhances androgen receptor-responsive gene expression depending on androgen level. Proc Natl Acad Sci USA. 2007;104(8):2571–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang SH, Yeh SH, Chen PJ. The driving circuit of HBx and androgen receptor in HBV-related hepatocarcinogenesis. Gut. 2014;63(11):1688–9.

    Article  CAS  PubMed  Google Scholar 

  75. Wu MH, Ma WL, Hsu CL, Chen YL, Ou JH, Ryan CK, Hung YC, Yeh S, Chang C. Androgen receptor promotes hepatitis B virus-induced hepatocarcinogenesis through modulation of hepatitis B virus RNA transcription. Sci Transl Med. 2010;2(32):32ra5.

    Article  CAS  Google Scholar 

  76. Yang WJ, Chang CJ, Yeh SH, Lin WH, Wang SH, Tsai TF, Chen DS, Chen PJ. Hepatitis B virus X protein enhances the transcriptional activity of the androgen receptor through c-Src and glycogen synthase kinase-3beta kinase pathways. Hepatology. 2009;49(5):1515–24.

    Article  CAS  PubMed  Google Scholar 

  77. Zhu R, Zhang JS, Zhu YZ, Fan J, Mao Y, Chen Q, Zhu HG. HBx-induced androgen receptor expression in HBV-associated hepatocarcinoma is independent of the methylation status of its promoter. Histol Histopathol. 2011;26(1):23–35.

    CAS  PubMed  Google Scholar 

  78. Tian YE, Xie XU, Lin Y, Tan G, Zhong WU. Androgen receptor in hepatocarcinogenesis: recent developments and perspectives. Oncol Lett. 2015;9(5):1983–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Falleti E, Fabris C, Toniutto P, Fontanini E, Cussigh A, Bitetto D, Fumolo E, Fornasiere E, Bragagnini W, Pinato DJ, et al. Interleukin-6 polymorphisms and gender: relationship with the occurrence of hepatocellular carcinoma in patients with end-stage liver disease. Oncology. 2009;77(5):304–13.

    Article  CAS  PubMed  Google Scholar 

  80. Hamaoka K, Nagoshi S, Sugawara K, Naiki K, Uchida Y, Inao M, Nakayama N, Fujiwara K, Mochida S. SNPs in the promoter region of the osteopontin gene as a possible host factor for sex difference in hepatocellular carcinoma development in patients with HCV. Hepatol Int. 2013;7(2):683–92.

    Article  PubMed  Google Scholar 

  81. Kido T, Ou JH, Lau YF. The X-linked tumor suppressor TSPX interacts and promotes degradation of the hepatitis B viral protein HBx via the proteasome pathway. PLoS ONE. 2011;6(7):e22979.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bayin NS, Frenster JD, Kane JR, Rubenstein J, Modrek AS, Baitalmal R, Dolgalev I, Rudzenski K, Scarabottolo L, Crespi D, et al. GPR133 (ADGRD1), an adhesion G-protein-coupled receptor, is necessary for glioblastoma growth. Oncogenesis. 2016;5(10):e263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. He X, Liao W, Li Y, Wang Y, Chen Q, Jin J, He S. Upregulation of hyaluronan-mediated motility receptor in hepatocellular carcinoma predicts poor survival. Oncol Lett. 2015;10(6):3639–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lee WH, Chen LC, Lee CJ, Huang CC, Ho YS, Yang PS, Ho CT, Chang HL, Lin IH, Chang HW, et al. DNA primase polypeptide 1 (PRIM1) involves in estrogen-induced breast cancer formation through activation of the G2/M cell cycle checkpoint. Int J Cancer. 2019;144(3):615–30.

    Article  CAS  PubMed  Google Scholar 

  85. Shao J, Xu Z, Peng X, Chen M, Zhu Y, Xu L, Zhu H, Yang B, Luo P, He Q. Gefitinib synergizes with irinotecan to suppress hepatocellular carcinoma via antagonizing Rad51-Mediated DNA-repair. PLoS ONE. 2016;11(1):e0146968.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Tilghman J, Wu H, Sang Y, Shi X, Guerrero-Cazares H, Quinones-Hinojosa A, Eberhart CG, Laterra J, Ying M. HMMR maintains the stemness and tumorigenicity of glioblastoma stem-like cells. Cancer Res. 2014;74(11):3168–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Tsai MS, Kuo YH, Chiu YF, Su YC, Lin YW. Down-regulation of Rad51 expression overcomes drug resistance to gemcitabine in human non-small-cell lung cancer cells. J Pharmacol Exp Ther. 2010;335(3):830–40.

    Article  CAS  PubMed  Google Scholar 

  88. Yan X, Chua MS, He J, So SK. Small interfering RNA targeting CDC25B inhibits liver tumor growth in vitro and in vivo. Mol Cancer. 2008;7:19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Zhong Y, Yang J, Xu WW, Wang Y, Zheng CC, Li B, He QY. KCTD12 promotes tumorigenesis by facilitating CDC25B/CDK1/Aurora A-dependent G2/M transition. Oncogene. 2017;36(44):6177–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lee B, Ha SY, Song DH, Lee HW, Cho SY, Park CK. High expression of ribonucleotide reductase subunit M2 correlates with poor prognosis of hepatocellular carcinoma. Gut Liver. 2014;8(6):662–8.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Zhu P, Jin J, Liao Y, Li J, Yu XZ, Liao W, He S. A novel prognostic biomarker SPC24 up-regulated in hepatocellular carcinoma. Oncotarget. 2015;6(38):41383–97.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Niedworok C, Kretschmer I, Rock K, Vom Dorp F, Szarvas T, Hess J, Freudenberger T, Melchior-Becker A, Rubben H, Fischer JW. The impact of the receptor of hyaluronan-mediated motility (RHAMM) on human urothelial transitional cell cancer of the bladder. PLoS ONE. 2013;8(9):e75681.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Rizzardi AE, Vogel RI, Koopmeiners JS, Forster CL, Marston LO, Rosener NK, Akentieva N, Price MA, Metzger GJ, Warlick CA, et al. Elevated hyaluronan and hyaluronan-mediated motility receptor are associated with biochemical failure in patients with intermediate-grade prostate tumors. Cancer. 2014;120(12):1800–9.

    Article  CAS  PubMed  Google Scholar 

  94. Mele V, Sokol L, Kolzer VH, Pfaff D, Muraro MG, Keller I, Stefan Z, Centeno I, Terracciano LM, Dawson H, et al. The hyaluronan-mediated motility receptor RHAMM promotes growth, invasiveness and dissemination of colorectal cancer. Oncotarget. 2017;8(41):70617–29.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Jordan AR, Racine RR, Hennig MJ, Lokeshwar VB. The role of CD44 in disease pathophysiology and targeted treatment. Front Immunol. 2015;6:182.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Gudowska M, Gruszewska E, Panasiuk A, Cylwik B, Flisiak R, Swiderska M, Szmitkowski M, Chrostek L. Hyaluronic acid concentration in liver diseases. Clin Exp Med. 2016;16(4):523–8.

    Article  CAS  PubMed  Google Scholar 

  97. Maxwell CA, McCarthy J, Turley E. Cell-surface and mitotic-spindle RHAMM: moonlighting or dual oncogenic functions? J Cell Sci. 2008;121(Pt 7):925–32.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

TK and Y-FCL conceived the idea of TSPY as an oncogene in liver cancer. TK performed the experimental and data analyses; Y-FCL and TK co-wrote the manuscript. Both authors read and approved the final manuscript.

Acknowledgements

We thank Dr. Yunmin Li for technical assistance.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Transcriptome data of HuH-7 cells will be submitted to the Gene Expression Omnibus database, and the accession numbers posted upon acceptance of the manuscript. All other reagents will be available upon request from the authors after the manuscript is published.

Consent for publication

All authors have read and consented for the publication of this manuscript.

Ethics approval and consent to participate

Not applicable.

Funding

This work is partially supported by a grant from the Department of Defense Peer-Reviewed Cancer Program to YFCL (CA150248) and a Merit-Review Grant IO1 BX004446 from the Department of Veterans Affairs. YFCL is a Research Career Scientist of the Department of Veterans Affairs.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yun-Fai Chris Lau.

Additional files

Additional file 1: Table S1.

Differentially expressed genes between TSPY-overexpressing and EGFP-control HuH-7 cells.

Additional file 2: Figure S1.

Results of Ingenuity Pathway Analysis (IPA) using a stringent cutoff on expression level of DEGs.

Additional file 3: Table S2.

Differentially expressed genes between the male TSPY-high HCCs and the male TSPY-silent HCCs.

Additional file 4: Table S3.

DEGs commonly correlated with the TSPY expression level in both HuH-7 cells and clincal male HCC samples.

Additional file 5: Figure S2.

Survival rates of the high-expresser patients (red lines) and the low-expresser patients (blue lines) for the 16TSPY downstream genes.

Additional file 6: Figure S3.

Comparison of the gene expression levels of the 16 TSPY downstream genes among female non-tumor liver (NT), female HCC, male non-tumor liver (NT), male TSPY-silent HCC (TS(−)), and male TSPY-high HCC (TS(++)) groups.

Additional file 7: Table S4.

Detailed description of 16 genes potentially regulated by TSPY and associated with prognoses in HCC patients.

Additional file 8: Figure S4.

Small molecule inhibitors for CDC25 and RRM2, the downstream pro-oncogenic molecules of TSPY, inhibited cell proliferation in hepatocellular carcinoma cell line HuH-7.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kido, T., Lau, YF.C. The Y-linked proto-oncogene TSPY contributes to poor prognosis of the male hepatocellular carcinoma patients by promoting the pro-oncogenic and suppressing the anti-oncogenic gene expression. Cell Biosci 9, 22 (2019). https://doi.org/10.1186/s13578-019-0287-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13578-019-0287-x

Keywords