Purpose: Golgi phosphoprotein 3 (GOLPH3) has been reported to be involved in various biologic processes. The clinical significance and biologic role of GOLPH3 in breast cancer, however, remains unknown.

Experimental Design: Expression of GOLPH3 in normal breast cells, breast cancer cells, and 6-paired breast cancer and adjacent noncancerous tissues were quantified using real-time PCR and Western blotting. GOLPH3 protein expression was analyzed in 258 archived, paraffin-embedded breast cancer samples using immunohistochemistry. The role of GOLPH3 in breast cancer cell proliferation and tumorigenicity was explored in vitro and in vivo. Western blotting and luciferase reporter analyses were used to investigate the effect of GOLPH3 overexpression and silencing on the expression of cell-cycle regulators and FOXO1 transcriptional activity.

Results: GOLPH3 was significantly upregulated in breast cancer cells and tissues compared with normal cells and tissues. Immunohistochemical analysis revealed high expression of GOLPH3 in 133 of 258 (51.6%) breast cancer specimens. Statistical analysis showed a significant correlation of GOLPH3 expression with advanced clinical stage and poorer survival. Overexpression and ablation of GOLPH3 promoted and inhibited, respectively, the proliferation and tumorigenicity of breast cancer cells in vitro and in vivo. GOLPH3 overexpression enhanced AKT activity and decreased FOXO1 transcriptional activity, downregulated cyclin-dependent kinase (CDK) inhibitor p21Cip1, p27Kip1, and p57Kip2, and upregulated the CDK regulator cyclin D1.

Conclusion: Our results suggest that high GOLPH3 expression is associated with poor overall survival in patients with breast cancer and that GOLPH3 overexpression increases the proliferation and tumorigenicity of human breast cancer cells. Clin Cancer Res; 18(15); 4059–69. ©2012 AACR.

Translational Relevance

Golgi phosphoprotein 3 (GOLPH3), a trans-Golgi matrix membrane protein, plays a role in protein trafficking, receptor recycling, and glycosylation. GOLPH3 has also been linked to tumorigenesis. In this study, we found that GOLPH3 mRNA and protein expression were significantly upregulated in human breast cancer tissues. High levels of GOLPH3 expression were associated with advanced clinical stage and poorly differentiated tumors. GOLPH3 was an independent prognostic factor, along with clinical stage, T classification, M classification, and Ki67 expression. High levels of GOLPH3 expression were associated with poorer overall survival in breast cancer. Furthermore, we showed that overexpression and silencing of GOLPH3 promoted and inhibited, respectively, breast cancer cell proliferation and tumorigenicity in vitro and in vivo. Together, these results suggest that GOLPH3 overexpression promotes progression of human breast cancer, and GOLPH3 has potential as a novel prognostic biomarker and therapeutic target for the treatment of human breast cancer.

Breast cancer is the most common cancer among females and the second leading cause of cancer-related deaths in females. More than 1 million women are diagnosed, and more than 400,000 women die from breast cancer every year (1). Mounting evidences indicate that induction of the neoplastic phenotype is invoked for the early events leading to increased tumorigenicity and metastasis. Thus, it is of great clinical value to identify potential early biomarkers to improve the diagnosis and prognostic assessment of breast cancer.

Golgi phosphoprotein 3 (GOLPH3), also known as GPP34, is a newly identified membrane protein in the trans-Golgi matrix, which plays a role in anterograde and retrograde Golgi trafficking (2–4). Recently, GOLPH3 has been shown to be involved in tumorigenesis. GOLPH3 localizes in human chromosome 5p13, a region that is frequently amplified in multiple solid tumor types (5–7). It has been reported that overexpression of GOLPH3 promotes cell transformation via enhancing the activity of the serine/threonine kinase mTOR (8, 9). GOLPH3 overexpression also drives the transformation of primary cell lines and enhances mouse xenograft tumor growth in vivo. In contrast, silencing of GOLPH3 expression abrogates the transformation and proliferation of tumor cell lines in which GOLPH3 is amplified (9, 10). The clinical significance and biologic role of GOLPH3 in breast cancer, however, remains largely unknown.

In this study, we found that GOLPH3 expression was significantly upregulated in breast cancer cells and tissues, and was associated with the clinical features of breast cancer. Furthermore, we showed that GOLPH3 can promote proliferation and tumorigenicity in breast cancer cells in vitro and in vivo. Moreover, the biologic behavior of GOLPH3 was mechanistically associated with suppression of FOXO1 transcriptional activity via activation of the AKT signaling pathway. Our results show that GOLPH3 functions as an oncoprotein in breast cancer progression and may provide a new therapeutic target for the treatment of human cancer.

Cell lines

Human mammary epithelial cells (HMEC) were established according to a previous report (11). Breast cancer cell lines, including MDA-MB-231, MDA-MB-415, T47D, MCF-10a, BT474, MCF-7, and Bcap37 were cultured in Dulbecco's Modified Eagle's Medium (Gibco) supplemented with 10% FBS (HyClone).

Vectors and retroviral infection

A GOLPH3 expression construct was generated by subcloning PCR-amplified full-length human GOLPH3 cDNA into the pMSCV plasmid. For depletion of GOLPH3, 2 human GOLPH3-targeting siRNA sequences were cloned into pSuper-retro-puro to generate pSuper-retro-GOLPH3-RNAi(s), respectively, and the sequences are RNAi#1: GCATGTTAAGGAAACTCAGCC and RNAi#2: GCAGCGCCTCATCAAGAAAG T (synthesized by Invitrogen). Retroviral production and infection were conducted as previously described (12). Stable cell lines expressing GOLPH3 or GOLPH3 short hairpin RNAs (shRNA) were selected for 10 days with 0.5 μg/mL puromycin. The reporter plasmid for quantitatively detecting the transcriptional activity of FOXO1 was generated in the pGL3-Enhancer plasmid (Promega) as described previously (13).

Patient information and tissue specimens

This study was conducted on a total of 258 paraffin-embedded, archived breast cancer samples, which were histopathologically and clinically diagnosed at the Sun Yat-sen University Cancer Center (Guangdong, China) from 1998 to 2004. Clinical and clinicopathologic classification and stage were determined according to the American Joint Committee on Cancer (AJCC) criteria. The histologic grade was determined according to the Elston–Ellis modification of the Scarff–Bloom–Richardson (SBR) system. For the use of these clinical materials for research purposes, prior consent of the patients and approval from the Institutional Research Ethics Committee were obtained. Clinical information of the samples is summarized in the Supplementary Table S1.

Immunohistochemistry

The immunohistochemical procedure and the scores of GOLPH3 expression were conducted as previously reported (14). Immunohistochemical (IHC) staining was quantitatively analyzed with the AxioVision Rel.4.6 computerized image analysis system assisted with the automatic measurement program (Carl Zeiss). Briefly, the stained sections were evaluated at ×200 magnification and 10 representative staining fields of each section, containing more than 95% tumor cells, were analyzed to verify the mean optical density (MOD), which represented the strength of staining signals as measured per positive pixel. The MOD data were statistically analyzed using the t test to compare the average MOD difference between different groups of tissues; P < 0.05 was considered significant.

Western blotting

Western blotting was carried out according to the standard methods as described previously (11), by using anti-GOLPH3, anti-p16Ink4A, anti-p21Cip1, anti-p27Kip1, anti-p57Kip2, anti-cyclin A, anti-cyclin E, anti-cyclin D1, anti-p-Rb, anti-Rb, anti-p-AKT, anti-AKT, anti-p-GSK-3β, anti-GSK-3β, anti-p-FOXO1, anti-FOXO1, anti-Ki67, anti-p-FOXO3a, anti-FOXO3a, anti-p-FOXO4, anti-FOXO4, anti-P84, and anti-EF-1α antibodies (Cell Signaling). The membranes were stripped and reprobed with an anti-α-tubulin mouse monoclonal antibody (Sigma) as a loading control.

MTT assay

Cells were seeded in 96-well plates at initial density of 2 × 103 per well. At each time point, cells were stained with 100 μL sterile MTT dye (0.5 mg/mL, Sigma) for 4 hours at 37°C, followed by removal of the culture medium and addition of 100 μL of dimethyl sulfoxide (Sigma). The absorbance was measured at 570 nm, with 655 nm as the reference wavelength. All experiments were carried out in triplicates.

Anchorage-independent growth ability assay

Five hundred cells were trypsinized and suspended in 2 mL complete medium plus 0.3% agar (Sigma). The agar cell mixture was plated on top of a bottom layer with 1% agar complete medium mixture. For about 10 days, viable colonies that were larger than 0.1 mm were counted. The experiment was carried out for each cell line in triplicates.

Colony formation assays

Cells were plated in 6-well plates (5 × 102 cells/plate) and cultured for 10 days. The colonies were stained with 1% crystal violet for 30 seconds after fixation with 10% formaldehyde for 5 minutes.

Flow cytometry

Cells were harvested, washed with cold PBS, and processed for cell-cycle analysis by using flow cytometry. Briefly, the cells were fixed in 75% ethanol and stored at −20°C overnight for later analysis. The fixed cells were centrifuged at 1,000 rpm for 5 minutes and washed with cold PBS twice. RNase A (20 μg/mL final concentration) and propidium iodide staining solution (50 μg/mL final concentration) were added to the cells and incubated for 30 minutes at 37°C in the dark. Fifty thousand cells were analyzed by using a Cytomics FC 500 instrument (Beckman Coulter) equipped with CXP software. Modifit LT 3.1 trial cell cycle analysis software was used to determine the percentage of cells in the different phases of the cell cycle.

Bromodeoxyuridine labeling and immunofluorescence

Cells were plated on coverslips (Fisher No.12-545-80, Fisher Scientific). After 24 hours, cells were incubated with bromodeoxyridine (BrdUrd) for 1 hour and stained with anti-BrdU antibody (Upstate) according to the manufacturer's instruction. Gray level images were acquired under a laser scanning microscope (Axioskop 2 plus; Carl Zeiss Co. Ltd.).

Xenografted tumor model

The xenografted tumor model was constructed as previously reported (15). Female Balb/c nude mice (4–5 weeks of age, 18–20 g) were purchased form the Slac-Jingda Laboratory Animal (Hunan, China) and were housed in barrier facilities on a 12-hour light/dark cycle. All experimental procedures were approved by the Institutional Animal Care and Use Committee of Sun Yat-sen University. The Balb/c nude mice were randomly divided into 4 groups (n = 5/group). A 0.72-mg E2 60-day release pellet (Innovative Research of America) was implanted subcutaneously on the dorsal side of each mouse 1 day before tumor cell implantation to support the growth of the estrogen-dependent MCF-7 cell–derived tumors. For tumor cell implantation, MCF-7-GOLPH3 or MCF-7-GOLPH3-RNAi or their respective control cells (2 × 106) in 0.25 mL of the mixture were injected into the mammary fat pads of female athymic mice. Tumors were examined twice weekly; length, width, and thickness measurements were obtained with calipers and tumor volumes were calculated. On day 23, animals were euthanized and tumors were excised and weighed.

Statistical analysis

All statistical analyses were conducted by using the SPSS version 13.0 statistical software packages. Comparisons between groups for statistical significance were carried out with a 2-tailed paired Student t test. A general linearized model was used to compare tumor volumes within groups at each time point. The relationship between GOLPH3 expression and clinicopathologic characteristics was analyzed by the χ2 test. Survival curves were plotted by the Kaplan–Meier method and compared by using the log-rank test. Survival data were evaluated by using univariate and multivariate Cox regression analyses. A P value of less than 0.05 was considered statistically significant in all cases.

GOLPH3 is upregulated in human breast cancer cells and tissues

Real-time PCR and Western blot analysis showed that GOLPH3 mRNA and protein expression were significantly upregulated in all the 7 tested breast cancer cell lines, compared with normal cells (HMECs; Fig. 1A and Supplementary Fig. S1A). Furthermore, comparative analysis revealed that the expression levels of GOLPH3 in 6 pairs of human breast cancer tissues were higher than those in matched adjacent nontumor tissues (Fig. 1A and B and Supplementary Fig. S1B). These results suggest that GOLPH3 expression is upregulated in breast cancer.

Figure 1.

GOLPH3 expression is upregulated in breast cancer. A, Western blot analysis of GOLPH3 expression in HMECs and 7 breast cancer cell lines (left), and in matched primary breast cancer tissues (T) and adjacent noncancerous tissues (ANT; right). α-Tubulin was used as a loading control. B and C, IHC staining analysis of GOLPH3 protein expression (B) in matched primary breast cancer tissues (T) and adjacent noncancerous tissues (ANT), and (C) in normal breast tissues and breast cancer tissues of different clinical stages. (D) Kaplan–Meier curves with univariate analyses (log-rank) for patients with breast cancer with low GOLPH3-expressing (n = 125) versus high GOLPH3-expressing tumors (n = 133).

Figure 1.

GOLPH3 expression is upregulated in breast cancer. A, Western blot analysis of GOLPH3 expression in HMECs and 7 breast cancer cell lines (left), and in matched primary breast cancer tissues (T) and adjacent noncancerous tissues (ANT; right). α-Tubulin was used as a loading control. B and C, IHC staining analysis of GOLPH3 protein expression (B) in matched primary breast cancer tissues (T) and adjacent noncancerous tissues (ANT), and (C) in normal breast tissues and breast cancer tissues of different clinical stages. (D) Kaplan–Meier curves with univariate analyses (log-rank) for patients with breast cancer with low GOLPH3-expressing (n = 125) versus high GOLPH3-expressing tumors (n = 133).

Close modal

GOLPH3 expression is associated with the clinical features of breast cancer

To investigate the relationship between GOLPH3 expression and the clinical features of breast cancer, GOLPH3 expression was examined in 258 paraffin-embedded, archived breast cancer tissues using IHC staining. The samples included 25 cases of clinical stage I (9.7%), 123 cases of stage II (47.5%), 76 cases of stage III (29.3%), and 34 cases of stage IV (13.1%) breast cancer. Statistical analyses of the IHC staining are summarized in Supplementary Tables S2 and S3. As shown in Fig. 1C, IHC staining showed that GOLPH3 expression in breast cancer increased with advanced clinical stage and in the tumors with poor pathologic differentiation (Supplementary Fig. S2). Statistical analysis revealed that GOLPH3 levels were strongly associated with the clinical stage (P < 0.001), T classification (P = 0.011), N classification (P < 0.001), and M classification (P < 0.001), but inverse correlated with expression of estrogen receptor (ER; P = 0.045; Supplementary Tables S2 and S3).

Moreover, Kaplan–Meier survival curves and the log-rank test survival analysis showed that the overall survival of patients with high levels of GOLPH3 was significantly poorer than patients with low levels of GOLPH3 (P < 0.001; Fig. 1D). In addition, similar results were obtained in stage I–II subgroup patients and stage III–IV subgroup patients (Supplementary Fig. S3). Univariate and multivariate analyses indicated that clinical stage, T classification, M classification, and expression of GOLPH3 and Ki67 were independent prognostic factors (Supplementary Table S4), suggesting that GOLPH3 may be a prognostic factor for survival in patients with breast cancer.

GOLPH3 promotes the proliferation of breast cancer cells

Strikingly, we found that the areas in breast cancer specimens that displayed high levels of GOLPH3 staining showed strong Ki67 staining signals, and areas with low GOLPH3 expression displayed weakly detectable Ki67 expression (Supplementary Fig. S4). Statistical analyses indicated that the expression of GOLPH3 was strongly associated with the expression level of Ki67 (P < 0.001; Supplementary Table S2 and S3), suggesting that GOLPH3 overexpression may contribute to the proliferation of breast cancer cells.

To further investigate the effect of GOLPH3 on proliferation, MCF-7 and MDA-MB-231 breast cancer cells stably overexpressing ectopic GOLPH3 were established (Fig. 2A). As shown in Fig. 2B, the proliferation rate was significantly increased in GOLPH3-overexpressing MCF-7 and MDA-MB-231 cells, as compared with control cells. These results were further confirmed by colony formation assay (Fig. 2C). Furthermore, we found that silencing GOLPH3 also significantly reduced cell proliferation that was analyzed by MTT and colony formation assays (Fig. 2A, B, and D). Collectively, our results suggest that overexpression of GOLPH3 plays important roles in proliferation of breast cancer cells in vitro.

Figure 2.

GOLPH3 is essential for breast cancer cell proliferation and tumorigenicity. A, Western blot analysis of GOLPH3 expression in GOLPH3-transduced and GOLPH3-silenced cells. α-Tubulin was used as a loading control. MTT assays (B) and colony formation assay (C and D) indicated that the growth rates increased in GOLPH3-overexpressing cells and decreased in GOLPH3-silenced cells. Values are mean±SD of 3 independent experiments. *, P < 0.05.

Figure 2.

GOLPH3 is essential for breast cancer cell proliferation and tumorigenicity. A, Western blot analysis of GOLPH3 expression in GOLPH3-transduced and GOLPH3-silenced cells. α-Tubulin was used as a loading control. MTT assays (B) and colony formation assay (C and D) indicated that the growth rates increased in GOLPH3-overexpressing cells and decreased in GOLPH3-silenced cells. Values are mean±SD of 3 independent experiments. *, P < 0.05.

Close modal

GOLPH3 promotes the tumorigenicity of breast cancer cells in vitro and in vivo

The anchorage-independent growth assay revealed that overexpressing GOLPH3 significantly increased, but silencing GOLPH3 decreased, the anchorage-independent growth ability of MCF-7 and MDA-MB-231 cells in soft agar (Fig. 3A and B), indicating that GOLPH3 plays a significant role in the tumorigenicity of breast cancer cells in vitro. We then further examined whether GOLPH3 could promote the tumorigenicity of breast cancer cells in vivo. As shown in Fig. 3C, GOLPH3-transduced tumors grew significantly faster than the control tumors at each different time point (P < 0.001), whereas the tumors formed by GOLPH3-silenced cells grew much slower than control tumors (P < 0.001). The tumors formed by GOLPH3-transduced breast cancer cells were larger and had higher tumor weights than vector control tumors. Conversely, tumors formed by GOLPH3-silenced cells were smaller, in both size and weight, than the tumors formed from shRNA vector control cells (Fig. 3C, left and right). Importantly, IHC assay showed that areas of strong GOLPH3 signals displayed intense cyclin D1 and Ki67 staining, whereas areas with low levels of GOLPH3 expression exhibited lower cyclin D1 and Ki67 staining signals (Supplementary Fig. S5), further supporting the notion that GOLPH3 promotes the proliferation and tumorigenicity of breast cancer cells.

Figure 3.

GOLPH3 promotes the tumorigenicity of breast cancer cells in vitro and in vivo. A and B, anchorage-independent growth assay in GOLPH3-overexpressing cells (A) and GOLPH3-silenced cells (B). Soft agar colony formation (colonies larger than 0.1 mm diameter) was quantified after 10 days of culture. C, xenograft model in nude mice. Indicated cells were inoculated into the fat pad of the nude mice (n = 5/group; left). Representative images of tumor growth (left). Tumor volume growth curves (middle). Mean tumor weights (right) 23 days after inoculation. All data are presented as mean ± SD; P < 0.05.

Figure 3.

GOLPH3 promotes the tumorigenicity of breast cancer cells in vitro and in vivo. A and B, anchorage-independent growth assay in GOLPH3-overexpressing cells (A) and GOLPH3-silenced cells (B). Soft agar colony formation (colonies larger than 0.1 mm diameter) was quantified after 10 days of culture. C, xenograft model in nude mice. Indicated cells were inoculated into the fat pad of the nude mice (n = 5/group; left). Representative images of tumor growth (left). Tumor volume growth curves (middle). Mean tumor weights (right) 23 days after inoculation. All data are presented as mean ± SD; P < 0.05.

Close modal

GOLPH3 regulates the G1–S-phase transition in breast cancer cells

To further investigate the mechanism of GOLPH3-mediated proliferation, the BrdUrd incorporation and flow cytometric assays were conducted. As shown in Fig. 4A and C, overexpression of GOLPH3 in MCF-7 and MDA-MB-231 cells significantly increased the percentage of cells which incorporated BrdUrd, whereas knockdown of GOLPH3 significantly reduced the percentage of BrdUrd-incorporated cells. In parallel, flow cytometry assay indicated that upregulation of GOLPH3 dramatically increased the percentage of S-phase cells and decreased the percentage of cells in the G1–G0 phase. Conversely, silencing of GOLPH3 increased the percentage of cells in the G1–G0 phase and decreased the percentage of S-phase cells (Fig. 4B and D). Taken together, these results suggest that GOLPH3 contributes to the G1–S-phase transition in breast cancer cells.

Figure 4.

GOLPH3 regulates the G1–S-phase transition in breast cancer cells. A, representative micrographs (left) and quantification (right) of BrdUrd-incorporated cells in indicated cells. B, flow cytometric analysis of vector control and GOLPH3-overexpressing cells. C, representative micrographs (left) and quantification (right) of BrdUrd incorporation in vector and GOLPH3 shRNA–infected cells. D, flow cytometric analysis of vector and GOLPH3 shRNA–infected cells. All values are mean ± SD of 3 independent experiments; P < 0.05. DAPI, 4′, 6-diamidino-2-phenylindole.

Figure 4.

GOLPH3 regulates the G1–S-phase transition in breast cancer cells. A, representative micrographs (left) and quantification (right) of BrdUrd-incorporated cells in indicated cells. B, flow cytometric analysis of vector control and GOLPH3-overexpressing cells. C, representative micrographs (left) and quantification (right) of BrdUrd incorporation in vector and GOLPH3 shRNA–infected cells. D, flow cytometric analysis of vector and GOLPH3 shRNA–infected cells. All values are mean ± SD of 3 independent experiments; P < 0.05. DAPI, 4′, 6-diamidino-2-phenylindole.

Close modal

Importantly, we found that the expression levels of p21Cip1, p27Kip1, and p57Kip2 were drastically reduced in GOLPH3-transduced cells, accompanied by significantly increased levels of cyclin D1 and phospho-Rb, compared with control cells (Fig. 5A and C). Consistently, the expression of p21Cip1, p27Kip1, and p57Kip2 were significantly increased, whereas cyclin D1 and phospho-Rb decreased in GOLPH3 silenced cells (Fig. 5B and C). However, GOLPH3 has no effects on the expression of p16Ink4A, cyclin A, and cyclin E (Fig. 5C).

Figure 5.

The effect of GOLPH3 on G1–S-phase regulators. A and B, real-time PCR analysis of p21Cip1, p27Kip1, p57Kip2, and cyclin D1 expression in GOLPH3-transduced cells (A) or GOLPH3 shRNA–infected cells (B). Expression levels were normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). C, Western blot analysis of p16Ink4A, p21Cip1, p27Kip1, p57Kip2, cyclin A, cyclin E, cyclin D1, phosphorylated Rb (p-Rb), and total Rb proteins in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. α-Tubulin was used as a loading control. Values are mean ± SD of 3 independent experiments; *, P < 0.05.

Figure 5.

The effect of GOLPH3 on G1–S-phase regulators. A and B, real-time PCR analysis of p21Cip1, p27Kip1, p57Kip2, and cyclin D1 expression in GOLPH3-transduced cells (A) or GOLPH3 shRNA–infected cells (B). Expression levels were normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). C, Western blot analysis of p16Ink4A, p21Cip1, p27Kip1, p57Kip2, cyclin A, cyclin E, cyclin D1, phosphorylated Rb (p-Rb), and total Rb proteins in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. α-Tubulin was used as a loading control. Values are mean ± SD of 3 independent experiments; *, P < 0.05.

Close modal

GOLPH3 downregulates FOXO1 transactivity and activates AKT signaling pathway

It has been reported that p21Cip1, p27Kip1, p57Kip2, and cyclin D1 are regulated by the transcriptional factor FOXO1 (16, 17). We then examined the effect of GOLPH3 on the regulation of FOXO1 transactivity. As shown in Fig. 6A, FOXO1 reporter activity was significantly decreased in GOLPH3-overexpressing cells and significantly increased in GOLPH3-silenced cells. Importantly, we did not observe significant alterations of ER activity and NF-κB reporter activity in GOLPH3-transduced and GOLPH3-silenced cells (Supplementary Fig. S6A), further suggesting the specific effect of GOLPH3 on the FOXO1 transactivity.

Figure 6.

GOLPH3 downregulates FOXO1 transcriptional activity via activation of the AKT signaling pathway. A, related FOXO1 reporter activity in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. B, Western blot analysis of p-AKT, total AKT, p-GSK-3β, total GSK-3β, phosphorylated FOXO1, and total FOXO1 in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. C, quantification of colony formation assays in indicated cells. D, MTT assays showed that silencing FOXO1 increased the proliferation of GOLPH3 shRNA(s)–infected cells. Values are mean ± SD of 3 independent experiments; *, P < 0.05.

Figure 6.

GOLPH3 downregulates FOXO1 transcriptional activity via activation of the AKT signaling pathway. A, related FOXO1 reporter activity in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. B, Western blot analysis of p-AKT, total AKT, p-GSK-3β, total GSK-3β, phosphorylated FOXO1, and total FOXO1 in GOLPH3-transduced cells or GOLPH3 shRNA–infected cells. C, quantification of colony formation assays in indicated cells. D, MTT assays showed that silencing FOXO1 increased the proliferation of GOLPH3 shRNA(s)–infected cells. Values are mean ± SD of 3 independent experiments; *, P < 0.05.

Close modal

As FOXO1 is a downstream target of the PI3K/AKT signaling pathway (18, 19), the protein levels of phospho-AKT and phospho-FOXO1 were analyzed. As shown in Fig. 6B, the levels of phospho-AKT and phospho-FOXO1 were increased in GOLPH3-overexpressing cells and decreased in GOLPH3-silenced cells. However, the expression levels of phospho-FOXO3a and FOXO4 were only slightly increased in GOLPH3-transduced cells and decreased in GOLPH3-silenced cells (Supplementary Fig. S6B). As expected, the cellular fractionation experiment showed that FOXO1 expression in the nuclei was markedly decreased in GOLPH3-transduced cells and increased in GOLPH3-silenced cells (Supplementary Fig. S6C). Moreover, chromatin immunoprecipitation assay indicated no physical binding of GOLPH3 to FOXO1 promoter regions (Supplementary Fig. S6D). Taken together, these data suggest that GOLPH3 may promote the proliferation of breast cancer cells by downregulation of FOXO1 transcriptional activity via activation of the AKT signaling pathway.

To further determine the role of FOXO1 in GOLPH3-mediated cell proliferation, we silenced the expression of FOXO1 in GOLPH3-silenced cells. As expected, the luciferase activity of the FOXO1 reporter was dramatically decreased in GOLPH3-silenced cells after FOXO1 siRNA(s) transfection (Supplementary Fig. S7A). Combined knockdown of FOXO1 in GOLPH3-silenced cells decreased the expression of p21Cip1, p27Kip1, and p57Kip2 but increased the cyclin D1 expression (Supplementary Fig. S7B and S7C). Moreover, colony formation and MTT assays showed that silencing of FOXO1 restored the growth rate of GOLPH3-silenced MCF-7 and MDA-MB-231 cells (Fig. 6C and D). Collectively, our results suggest that FOXO1 plays an important role in the effect of GOLPH3 on proliferation in breast cancer cells.

The key findings of this study are that GOLPH3 overexpression was significantly associated with progression of human breast cancer and that ectopic expression of GOLPH3 increased the proliferation and tumorigenicity of breast cancer cells. We showed that GOLPH3 promotes cell proliferation by accelerating the G1–S-phase transition through downregulation of FOXO1 transcriptional activity via activation of AKT signaling pathway. This study provides new insights and strong evidences that GOLPH3 overexpression plays important roles in promoting the tumorigenicity and progression of human breast cancer.

Tumorigenesis is a complex multistep process, characterized by uncontrolled cell growth and tumor formation and is largely associated with progressive accumulation of genetic and various epigenetic alterations in genes or proteins that regulate cell proliferation (20, 21). Therefore, identification of the genes and their products that lead to tumorigenesis is critical for providing new diagnostic and prognostic methods and potential therapeutic targets. Previously, it has been reported that GOLPH3 is amplified at the 5p13 region in several types of cancer, including breast cancer (9). The clinical significance and biologic role of GOLPH3 in breast cancer progression, however, remains largely unknown. Herein, we found that GOLPH3 was significantly upregulated in a large cohort of human breast cancer tissues and that GOLPH3 levels significantly correlated with the clinical characteristics of breast cancer, including the clinical stage, tumor–node–metastasis (TNM) classification, and expression of ER and Ki67. Importantly, survival analysis showed that patients with low GOLPH3 expression have better overall survival compared with patients with high levels of GOLPH3, suggesting that GOLPH3 is a predictor for survival of patients with breast cancer.

Mounting evidences have established that the tumor suppressor FOXO1 plays an important role in tumorigenesis via the regulation of a variety of biologic processes, such as proliferation, differentiation, stress responses, and apoptosis through transcriptional induction of the cyclin-dependent kinase (CDK) inhibitors, including p21Cip1, p27Kip1, and p57Kip2 (17, 22, 23). Indeed, deregulation of FOXO1 expression is observed in multiple cancers, including breast cancer, prostate cancer, chronic myelogenous leukemia, glioblastoma, rhabdomyosarcoma, and leukemia (17, 24–28). In the present study, we found that FOXO1 transactivity was decreased in GOLPH3-transduced cells and enhanced in GOLPH3-silenced cells, suggesting that GOLPH3-induced proliferation and tumorigenesis may be due to modulation of FOXO1 activity. Upregulation of GOLPH3 accelerated the G1–S-phase transition, whereas downregulation of GOLPH3 induced G1–S-phase arrest. Furthermore, we showed that the mechanism of GOLPH3-mediated proliferation was linked to alternations in the expression of the cell-cycle inhibitors p21Cip1, p27Kip1, and p57Kip2 as well as the CDK regulator cyclin D1.

AKT is a serine/threonine protein kinase that plays key roles in multiple cellular processes, such as cell proliferation, apoptosis, transcription, cell migration, and glucose metabolism. Activated AKT could stimulate the phosphorylation and impact various downstream targets, including GSK-3β, BAD, IKK, p27, MDM2, and the FOXO family of transcription factors (29). It has been shown that phosphorylation of FOXO1 at Ser256 by AKT leads to the nuclear/cytoplasmic translocation and subsequent degradation of FOXO1 via the ubiquitin–proteasome system (18, 19). Our study shows that the levels of phospho-AKT and phospho-FOXO1 increased in GOLPH3-overexpressing cells and decreased in GOLPH3-silenced cells. The mechanism by which GOLPH3 activates the AKT signaling pathway is under investigation in our laboratory currently.

Metastases are already disseminated to adjacent lymph nodes or distant organs at the time of diagnosis in most patients with breast cancer (30). It has been reported that GOLPH3 connects the Golgi membranes to actin cytoskeleton through binding to myosin MYO18A (31). MYO18A, a novel p21-activated kinase PAK2-binding partner, has been proven to play an important role in the modulation of membrane ruffle formation, focal adhesion turnover, actin filament reorganization, and the migratory ability of epithelial cells (32). Thus, the role of GOLPH3 in breast cancer cell migration and metastasis needs to be further explored.

In summary, our results suggest that GOLPH3 plays an important role in human breast cancer progression. Full understanding of the precise role of GOLPH3 in human cancer may provide the opportunity to develop a novel therapeutic strategy by suppressing expression of GOLPH3 in breast cancer cells. In addition, GOLPH3 has potential as a relevant clinical indicator of disease progression and as a prognostic marker for patient survival in human breast cancer. Translational research on the clinical use of GOLPH3 is required to generate a methodology and evaluate the molecular diagnostic ability of GOLPH3 in breast cancer.

No potential conflicts of interest were disclosed.

This work was supported by the Natural Science Foundation of China (No. 81071780 to L. Song, 81030048 to J. Li, 81000888 to Z. Zeng, 81171892 to X. Wang, and 30900569 to K. Chen) and The Science and Technology Department of Guangdong Province, China (No. S2011020002757 to L. Song).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Glass
AG
,
Lacey
JV
,
Carreon
JD
,
Hoover
RN
. 
Breast cancer incidence, 1980–2006: combined roles of menopausal hormone therapy, screening mammography, and estrogen receptor status
.
J Natl Cancer Inst
2007
;
99
:
1152
61
.
2.
Nakashima
KN
,
Asoh
S
,
Ishibashi
Y
,
Mukai
Y
,
Shidara
Y
,
Oda
H
, et al
MIDAS/GPP34, a nuclear gene product, regulates total mitochondrial mass in response to mitochondrial dysfunction
.
J Cell Sci
2005
;
118
:
5357
67
.
3.
Wu
CC
,
Taylor
RS
,
Lane
DR
,
Ladinsky
MS
,
Weisz
JA
,
Howell
KE
. 
GMx33: a novel family of trans-Golgi proteins identified by proteomics
.
Traffic
2000
;
1
:
963
75
.
4.
Snyder
CM
,
Mardones
GA
,
Ladinsky
MS
,
Howell
KE
. 
GMx33 associates with the trans-Golgi matrix in a dynamic manner and sorts within tubules exiting the Golgi
.
Mol Biol Cell
2006
;
17
:
511
24
.
5.
Gorringe
KL
,
Boussioutas
A
,
Bowtell
DD
. 
Novel regions of chromosomal amplification at 6p21, 5p13, and 12q14 in gastric cancer identified by array comparative genomic hybridization
.
Genes Chromosomes Cancer
2005
;
42
:
247
59
.
6.
Bohm
M
,
Wieland
I
,
Schmidt
C
,
Rubben
H
,
Allhoff
EP
. 
Loss of heterozygosity on chromosome 5p13-12 predicts adverse prognosis in advanced bladder cancer independent of tumor stage and grade
.
J Urol
2002
;
168
:
2655
8
.
7.
Yokoi
S
,
Yasui
K
,
Saito-Ohara
F
,
Koshikawa
K
,
Iizasa
T
,
Fujisawa
T
, et al
A novel target gene, SKP2, within the 5p13 amplicon that is frequently detected in small cell lung cancers
.
Am J Pathol
2002
;
161
:
207
16
.
8.
Abraham
RT
. 
GOLPH3 links the Golgi network to mTOR signaling and human cancer
.
Pigment Cell Melanoma Res
2009
;
22
:
378
9
.
9.
Scott
KL
,
Kabbarah
O
,
Liang
MC
,
Ivanova
E
,
Anagnostou
V
,
Wu
J
, et al
GOLPH3 modulates mTOR signalling and rapamycin sensitivity in cancer
.
Nature
2009
;
459
:
1085
90
.
10.
Scott
KL
,
Chin
L
. 
Signaling from the Golgi: mechanisms and models for Golgi phosphoprotein 3-mediated oncogenesis
.
Clin Cancer Res
2010
;
16
:
2229
34
.
11.
Li
J
,
Zhang
N
,
Song
LB
,
Liao
WT
,
Jiang
LL
,
Gong
LY
, et al
Astrocyte elevated gene-1 is a novel prognostic marker for breast cancer progression and overall patient survival
.
Clin Cancer Res
2008
;
14
:
3319
26
.
12.
Hahn
WC
,
Dessain
SK
,
Brooks
MW
,
King
JE
,
Elenbaas
B
,
Sabatini
DM
, et al
Enumeration of the simian virus 40 early region elements necessary for human cell transformation
.
Mol Cell Biol
2002
;
22
:
2111
23
.
13.
Tang
ED
,
Nunez
G
,
Barr
FG
,
Guan
KL
. 
Negative regulation of the forkhead transcription factor FKHR by Akt
.
J Biol Chem
1999
;
274
:
16741
6
.
14.
Liao
W
,
Feng
Y
,
Li
M
,
Liu
GL
,
Li
MZ
,
Zeng
MS
, et al
Overexression of centromere protein H is significantly associated with breast cancer progression and overall patient survival
.
Chin J Cancer
2011
;
30
:
627
37
.
15.
Lin
C
,
Wu
Z
,
Lin
X
,
Yu
C
,
Shi
T
,
Zeng
Y
, et al
Knockdown of FLOT1 impairs cell proliferation and tumorigenicity in breast cancer through upregulation of FOXO3a
.
Clin Cancer Res
2011
;
17
:
3089
99
.
16.
Evans-Anderson
HJ
,
Alfieri
CM
,
Yutzey
KE
. 
Regulation of cardiomyocyte proliferation and myocardial growth during development by FOXO transcription factors
.
Circ Res
2008
;
102
:
686
94
.
17.
Roeb
W
,
Boyer
A
,
Cavenee
WK
,
Arden
KC
. 
PAX3-FOXO1 controls expression of the p57Kip2 cell-cycle regulator through degradation of EGR1
.
Proc Natl Acad Sci U S A
2007
;
104
:
18085
90
.
18.
Cardozo
T
,
Pagano
M
. 
The SCF ubiquitin ligase: insights into a molecular machine
.
Nat Rev Mol Cell Biol
2004
;
5
:
739
51
.
19.
Huang
H
,
Regan
KM
,
Wang
F
,
Wang
D
,
Smith
DI
,
van Deursen
JM
, et al
Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation
.
Proc Natl Acad Sci U S A
2005
;
102
:
1649
54
.
20.
Ponder
BA
. 
Cancer genetics
.
Nature
2001
;
411
:
336
41
.
21.
Bartek
J
. 
DNA damage response, genetic instability and cancer: from mechanistic insights to personalized treatment
.
Mol Oncol
2011
;
5
:
303
7
.
22.
Nakamura
N
,
Ramaswamy
S
,
Vazquez
F
,
Signoretti
S
,
Loda
M
,
Sellers
WR
. 
Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN
.
Mol Cell Biol
2000
;
20
:
8969
82
.
23.
Seoane
J
,
Le
HV
,
Shen
L
,
Anderson
SA
,
Massague
J
. 
Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation
.
Cell
2004
;
117
:
211
23
.
24.
Hu
MC
,
Lee
DF
,
Xia
W
,
Golfman
LS
,
Ou-Yang
F
,
Yang
JY
, et al
IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a
.
Cell
2004
;
117
:
225
37
.
25.
Dong
XY
,
Chen
C
,
Sun
X
,
Guo
P
,
Vessella
RL
,
Wang
RX
, et al
FOXO1A is a candidate for the 13q14 tumor suppressor gene inhibiting androgen receptor signaling in prostate cancer
.
Cancer Res
2006
;
66
:
6998
7006
.
26.
Ward
EC
,
Hoekstra
AV
,
Blok
LJ
,
Hanifi-Moghaddam
P
,
Lurain
JR
,
Singh
DK
, et al
The regulation and function of the forkhead transcription factor, Forkhead box O1, is dependent on the progesterone receptor in endometrial carcinoma
.
Endocrinology
2008
;
149
:
1942
50
.
27.
Myatt
SS
,
Lam
EW
. 
The emerging roles of forkhead box (Fox) proteins in cancer
.
Nat Rev Cancer
2007
;
7
:
847
59
.
28.
Greer
EL
,
Brunet
A
. 
FOXO transcription factors at the interface between longevity and tumor suppression
.
Oncogene
2005
;
24
:
7410
25
.
29.
Kazi
AA
,
Molitoris
KH
,
Koos
RD
. 
Estrogen rapidly activates the PI3K/AKT pathway and hypoxia-inducible factor 1 and induces vascular endothelial growth factor A expression in luminal epithelial cells of the rat uterus
.
Biol Reprod
2009
;
81
:
378
87
.
30.
Autier
P
,
Hery
C
,
Haukka
J
,
Boniol
M
,
Byrnes
G
. 
Advanced breast cancer and breast cancer mortality in randomized controlled trials on mammography screening
.
J Clin Oncol
2009
;
27
:
5919
23
.
31.
Dippold
HC
,
Ng
MM
,
Farber-Katz
SE
,
Kerr
ML
,
Peterman
MC
,
Sim
R
, et al
GOLPH3 bridges phosphatidylinositol-4- phosphate and actomyosin to stretch and shape the Golgi to promote budding
.
Cell
2009
;
139
:
337
51
.
32.
Hsu
RM
,
Tsai
MH
,
Hsieh
YJ
,
Lyu
PC
,
Yu
JS
. 
Identification of MYO18A as a novel interacting partner of the PAK2/betaPIX/GIT1 complex and its potential function in modulating epithelial cell migration
.
Mol Biol Cell
2010
;
21
:
287
301
.