The p34SEI-1 protein exerts oncogenic effects via regulation of the cell cycle, which occurs through a direct interaction with cyclin-dependent kinase 4. Such regulation can increase the survival of various types of tumor cells. Here, we show that the antiapoptotic function of p34SEI-1 increases tumor cell survival by protecting the X-linked inhibitor of apoptosis protein (XIAP) from degradation. Our findings show that p34SEI-1 inhibits apoptosis. This antiapoptotic effect was eliminated by the suppression of p34SEI-1 expression. We also determined that direct binding of p34SEI-1 to the BIR2 domain prevents ubiquitination of XIAP. Interestingly, p34SEI-1 expression is absent or weak in normal tissues but is strongly expressed in tissues obtained from patients with breast cancer. Furthermore, the expression levels of p34SEI-1 and XIAP seem to be coordinated in human breast cancer cell lines and tumor tissues. Thus, our findings reveal that p34SEI-1 uses a novel apoptosis-inhibiting mechanism to stabilize XIAP. [Cancer Res 2009;69(3):741–6]

The recently identified p34SEI-1-encoding SEI-1 gene positively regulates cell division by binding to cyclin-dependent kinase (CDK4; ref. 1). Previous studies have shown that the SEI-1 gene is highly expressed in carcinomas from pancreatic (2), lung (3), and ovarian tissues (4), suggesting that p34SEI-1 functions as an oncoprotein. Interestingly, recent studies have shown that p34SEI-1 is capable of oncogenic transformation of mouse fibroblasts (5). However, it is not clear whether p34SEI-1 regulates apoptosis through the suppression of proapoptotic proteins or via the activation of antiapoptotic proteins.

Previous studies have established that the prosurvival protein Akt/PKB phosphorylates and subsequently stabilizes the X-linked inhibitor of apoptosis protein (XIAP; ref. 6). Other studies have shown that Notch inhibits apoptosis by directly preventing XIAP ubiquitination (7). These findings suggest that XIAP plays a key role in antiapoptotic functions and cell survival. The XIAP is one of the best-characterized members of the IAP family, which consists of at least eight proteins (8). Specifically, XIAP contains three baculovirus IAP repeat (BIR) domains and one RING-finger domain, which exerts E3 ligase activity (9). The BIR 2 domain directly binds to and inhibits active caspase-3 and caspase-7, whereas the BIR 3 domain associates with active caspase-9 (10). However, XIAP-mediated inhibition of caspase-3 and caspase-7 is abolished by Smac/DIABLO, which binds to the BIR 2 domain of XIAP (11). Previous studies have shown that XIAP is overexpressed in various breast cancer cells (12) and diverse malignant cells (13), indicating that XIAP may prove useful as a target for cancer therapies. With this in mind, various research groups have sought to identify inhibitors of XIAP and its BIR domains.

In this study, we show that p34SEI-1 prevents the ubiquitination and degradation of XIAP through a direct association with the BIR2 domain. Our results reveal that p34SEI-1 use s a novel apoptosis-inhibiting mechanism and that this protein represents a promising target for new antitumor therapies.

Cell culture, plasmids, and transfection. Human breast cancer cell lines as well as the human embryonic kidney line, 293, were cultured in DMEM (Life Technologies Bethesda Research Laboratories). The green fluorescent protein (GFP)-tagged p34SEI-1 plasmid and retroviral vectors were obtained by subcloning p34SEI-1 cDNA (i.e., provided by Dr. Rikiro Fukunaga, Graduate School of Medicine, Osaka University, Osaka, Japan) into the pEGFP-N1 (Invitrogen) and pBabe-retroviral vectors, respectively. Human cDNA encoding XIAP and 10 mutant variations were provided by Dr. C.S. Duckett (University of Michigan Medical School, Ann Arbor, MI). We also constructed three XIAP mutants using the PCR-based Quik Change site-directed mutagenesis kit (14). The XIAPC202A, XIAPC213A, and XIAPK208A constructs (i.e., in which Cys-202, Cys-213, and Lys-208, respectively, were replaced with alanine). Lipofect-AMINE 2000 (Invitrogen) was used for transient transfection.

Cell death analysis. Control cells and cells expressing either SEI-1, a p34SEI-1 small interfering RNA (siRNA) knockdown or a scrambled siRNA control were seeded at 105 cells per 60-mm dish, 24 h before treatment, in the presence of various apoptotic stimuli (i.e., staurosporin, etoposide, cisplatin). The cells were then incubated for 18 h.

Immunoprecipitation and in vivo ubiquitination assays. To analyze the interaction between endogenous p34SEI-1 and XIAP, cell lysates were prepared and immunoprecipitated with either anti–SEI-1 or anti-XIAP antibodies. Immunoprecipitation analysis was performed as previously described (7). In preparation for in vivo ubiquitination assays, cells were transfected with either GFP-tagged SEI-1 or HA-tagged XIAP. After immunoprecipitation with an anti-HA antibody, ubiquitin adducts were detected via immunoblot analysis using an anti-ubiquitin antibody.

RNA interference. Human breast cancer cells were transiently transfected with scrambled siRNA or p34SEI-1 siRNA (i.e., 150 pmol siRNA per 60-mm dish; using p34SEI-1 sequence 5′-CCGAAUUGGACUACCUCAUdTdT-3′). Scrambled control siRNA (i.e., 5′-GAAGCAGUCGCAGUGAAG AdTdT-3′) was obtained from Proligo LLC.

Immunofluorescence and immunohistochemistry analyses of p34SEI-1. For colocalization of XIAP and p34SEI-1, immunofluorescence staining was performed as previously described (15). To investigate the relationship between overexpression of p34SEI-1 and the development of breast cancer, we conducted an immunohistochemical analysis of p34SEI-1 expression in 60 tissue samples from patients with breast cancer. Immunohistochemical analysis was performed as previously described (16).

Immunoblot analysis. Immunoblot analysis was performed as previously described (17). Protein was probed with either anti-GFP, anti-HA, anti–caspase-7, anti-ubiquitin, anti–γ-tubulin (Santa Cruz Biotechnology), anti–cleaved caspase-9, anti–cleaved caspase-3, anti-XIAP (Cell Signaling Technology), or anti-p34SEI-1 (AXXORA LLC) antibody.

p34SEI-1 inhibits the apoptotic response to various stimuli. To determine if the differential induction of apoptosis by various stimuli (e.g., staurosporine, etoposide, and cisplatin) was dependent on the level of p34SEI-1, we first assayed the expression of p34SEI-1 in multiple lines of human breast cancer cells (Fig. 1A). The majority of the breast cancer cells displayed high levels of p34SEI-1 expression, with the exception of the SK-BR 3 and Hs578T cell lines. So we examined whether p34SEI-1–expressing cells were resistant to apoptotic inducer. Hs578T and MCF-7 cells were transfected with GFP-tagged p34SEI-1 or the GFP retroviral vector. Cells expressing p34SEI-1 or control GFP were treated with various stimuli (Fig. 1B,, i and ii). Cells expressing p34SEI-1 were resistant to each of these apoptotic stimuli, whereas the control GFP cells were not resistant. To further explore the antiapoptotic function of p34SEI-1, we examined the effects of p34SEI-1 silencing using siRNAs. The population of dead cells increased in p34SEI-1 siRNA–treated cells, relative to the population in scrambled siRNA–treated cells, after exposure to various stimuli (Fig. 1C,, i and ii), or TRAIL (Supplementary Fig. S1). To examine the mechanisms involved in the apoptosis-inhibiting effect of p34SEI-1, we investigated whether p34SEI-1 might affect the levels of several apoptotic related proteins. Expression of the antiapoptotic proteins cIAP-1 or cIAP-2 was not altered in p34SEI-1-expressing 293 cells (Fig. 1D). However, XIAP expression increased in p34SEI-1-expressing 293 cells (Fig. 1D). In addition, the population of dead cells and cleavage of procaspase-3 decreased in parallel with the up-regulation of XIAP in p34SEI-1-siRNA–treated cells, as well as in p34SEI-1-expressing 293 cells (Supplementary Figs. S2 and S3). Thus, the cleaved forms of caspase-3 and caspase-7 were inhibited via the up-regulation of XIAP, implying that p34SEI-1 inhibits apoptosis by preventing the degradation of XIAP.

Figure 1.

The inhibitory effect of p34SEI-1 on apoptosis. A, expression of p34 in multiple human breast cancer cell line. Breast cancer cell lines were grown in DMEM supplemented with 10% fetal bovine serum. Expression of p34SEI-1 was confirmed by Western blot analysis. B and C, multiple breast cancer cells [i.e., Hs578T (B, i), MCF-7 (B, ii; C, i) and MDA-MB231 (C, ii)] were transfected with either a GFP-tagged p34SEI-1 retroviral vector, a control vector (i.e., EGFP), scrambled siRNA or p34SEI-1-siRNA and treated with various proapoptotic agents [e.g., Hs578T cells were stimulated with 1 μmol/L staurosporin (STS), 400 μmol/L etoposide (eto), 400 μmol/L cisplatin (cis); 293 cells were stimulated with 0.5 μmol/L staurosporin, 400 μmol/L etoposide, 400 μmol/L cisplatin; MCF-7 cells were stimulated with 0.01 or 0.5 μmol/L staurosporin, 5 or 30 μmol/L etoposide, 5 or 30 μmol/L cisplatin; and MDA-MB-231 cells were stimulated with 0.01 μmol/L staurosporin, 3 μmol/L etoposide, or 3 μmol/L cisplatin] for 18 h. Cell viability was determined via trypan blue exclusion using at least 300 to 500 cells from each group. Columns, mean of three independent experiments performed in duplicate; bars, SE. D, 293 cells were infected with GFP-p34SEI-1 or a control vector (i.e., EGFP) for 24 h, followed by the preparation of cell lysates. Expression of the XIAP, c-IAP1, and c-IAP-2 proteins was analyzed using anti-XIAP, anti–c-IAP1, and anti–c-IAP2 antibodies, respectively. The γ-tubulin protein was included as a loading control.

Figure 1.

The inhibitory effect of p34SEI-1 on apoptosis. A, expression of p34 in multiple human breast cancer cell line. Breast cancer cell lines were grown in DMEM supplemented with 10% fetal bovine serum. Expression of p34SEI-1 was confirmed by Western blot analysis. B and C, multiple breast cancer cells [i.e., Hs578T (B, i), MCF-7 (B, ii; C, i) and MDA-MB231 (C, ii)] were transfected with either a GFP-tagged p34SEI-1 retroviral vector, a control vector (i.e., EGFP), scrambled siRNA or p34SEI-1-siRNA and treated with various proapoptotic agents [e.g., Hs578T cells were stimulated with 1 μmol/L staurosporin (STS), 400 μmol/L etoposide (eto), 400 μmol/L cisplatin (cis); 293 cells were stimulated with 0.5 μmol/L staurosporin, 400 μmol/L etoposide, 400 μmol/L cisplatin; MCF-7 cells were stimulated with 0.01 or 0.5 μmol/L staurosporin, 5 or 30 μmol/L etoposide, 5 or 30 μmol/L cisplatin; and MDA-MB-231 cells were stimulated with 0.01 μmol/L staurosporin, 3 μmol/L etoposide, or 3 μmol/L cisplatin] for 18 h. Cell viability was determined via trypan blue exclusion using at least 300 to 500 cells from each group. Columns, mean of three independent experiments performed in duplicate; bars, SE. D, 293 cells were infected with GFP-p34SEI-1 or a control vector (i.e., EGFP) for 24 h, followed by the preparation of cell lysates. Expression of the XIAP, c-IAP1, and c-IAP-2 proteins was analyzed using anti-XIAP, anti–c-IAP1, and anti–c-IAP2 antibodies, respectively. The γ-tubulin protein was included as a loading control.

Close modal

p34 SEI-1 inhibits the ubiquitination of XIAP and prevents subsequent degradation. As shown in Fig. 1, the degradation of XIAP was not induced in p34SEI-1-expressing cells. As protein stability is controlled in part by the ubiquitin-proteasome pathway (18), we used the proteasome inhibitor, MG132, to determine if XIAP degradation required proteasome-mediated protein degradation in control cells and p34SEI-1-expressing cells. In the absence of MG132, expression of XIAP increased significantly in cells expressing p34SEI-1. When cells were treated with MG132, XIAP levels increased slightly upon p34SEI-1 expression (Fig. 2A). We further explored whether XIAP turnover might be modulated by p34SEI-1 in the presence of a protein synthesis inhibitor (cycloheximide). Expression of XIAP increased remarkably and was sustained by the expression of p34SEI-1; however, XIAP expression gradually decreased in control cells after cycloheximide treatment (Fig. 2B,, i). We then explored XIAP levels in MCF7 cells treated with p34SEI-1-siRNA. Expression of XIAP decreased rapidly in p34SEI-1-siRNA–treated cells, relative to scrambled siRNA-treated cells (Fig. 2B,, ii), indicating that p34SEI-1 stabilizes XIAP by preventing proteasome-dependent protein degradation. A previous study showed that XIAP degradation occurs in response to apoptotic stress (19). Thus, we examined whether XIAP degradation induced by apoptotic stimuli might be inhibited in a p34SEI-1-dependent manner. Based on our findings thus far, we treated control cells and p34SEI-1-expressing cells with etoposide. In control cells, endogenous XIAP was ubiquitinated and subjected to proteasomal degradation; however, these effects did not occur in cells expressing p34SEI-1 (Fig. 2C). These results suggest that p34SEI-1 significantly inhibits XIAP ubiquitination in response to apoptotic stress. Ubiquitination of XIAP was also not detected upon expression of p34SEI-1 (Fig. 2D). A recent study reported that XIAP ubiquitination is enhanced by Smac/Diablo (14). Thus, we examined whether p34SEI-1 might inhibit the binding of Smac/Diablo and XIAP and might thus prevent ubiquitination of XIAP by Smac/Diablo (Supplementary Fig. S4). Surprisingly, Smac/Diablo did not bind to XIAP in the presence of p34SEI-1 and thus was unable to promote ubiquitination of XIAP by Smac/Diablo. These results suggest that p34SEI-1 strongly inhibits XIAP ubiquitination and prevents subsequent XIAP degradation.

Figure 2.

The p34SEI-1 protein inhibits the proteasomal degradation of XIAP. A, p34SEI-1-expressing 293 cells were treated with a proteasome inhibitor (i.e., MG132, 10 μmol/L) for 18 h, and the expression of XIAP was determined via immunoblot analysis using an anti-XIAP antibody. B, i, 293 cells were transfected with the GFP-tagged p34SEI-1 plasmid or a control vector for 18 h and treated with cycloheximide (i.e., 50 μg/mL, an inhibitor of protein synthesis) for the indicated times. Cell lysates were prepared at the indicated times and expression of XIAP and GFP-tagged p34SEI-1 was analyzed using anti-XIAP and anti-GFP antibodies, respectively. B, ii, MCF-7 cells were transfected with either p34SEI-1 siRNA or scrambled RNA for 24 h and treated with cycloheximide for the indicated times. Expression levels of XIAP and p34SEI-1 were then determined. C, after 293 cells were transfected with or without p34SEI-1, cells were treated with 400 μmol/L of etoposide for the indicated times in the presence of MG132. Cell lysates were immunoprecipitated (IP) with normal serum (i.e., IgG) or anti-XIAP antibody, and immunoblotted with anti-ubiquitin (i.e., top). Expression of XIAP was detected via immunoblot analysis using anti-XIAP antibodies. D, after the cotransfection of 293 cells with GFP-p34SEI-1 and HA-XIAP, cell lysates were immunoprecipitated with anti-HA antibodies and immunoblotted using anti-ubiquitin (i.e., top). The levels of GFP-p34SEI-1 and HA-XIAP were detected via Western blotting (WB) using anti-GFP and anti-HA antibodies, respectively. Input controls (i.e., using 5% of the cell lysates) were included on the blot.

Figure 2.

The p34SEI-1 protein inhibits the proteasomal degradation of XIAP. A, p34SEI-1-expressing 293 cells were treated with a proteasome inhibitor (i.e., MG132, 10 μmol/L) for 18 h, and the expression of XIAP was determined via immunoblot analysis using an anti-XIAP antibody. B, i, 293 cells were transfected with the GFP-tagged p34SEI-1 plasmid or a control vector for 18 h and treated with cycloheximide (i.e., 50 μg/mL, an inhibitor of protein synthesis) for the indicated times. Cell lysates were prepared at the indicated times and expression of XIAP and GFP-tagged p34SEI-1 was analyzed using anti-XIAP and anti-GFP antibodies, respectively. B, ii, MCF-7 cells were transfected with either p34SEI-1 siRNA or scrambled RNA for 24 h and treated with cycloheximide for the indicated times. Expression levels of XIAP and p34SEI-1 were then determined. C, after 293 cells were transfected with or without p34SEI-1, cells were treated with 400 μmol/L of etoposide for the indicated times in the presence of MG132. Cell lysates were immunoprecipitated (IP) with normal serum (i.e., IgG) or anti-XIAP antibody, and immunoblotted with anti-ubiquitin (i.e., top). Expression of XIAP was detected via immunoblot analysis using anti-XIAP antibodies. D, after the cotransfection of 293 cells with GFP-p34SEI-1 and HA-XIAP, cell lysates were immunoprecipitated with anti-HA antibodies and immunoblotted using anti-ubiquitin (i.e., top). The levels of GFP-p34SEI-1 and HA-XIAP were detected via Western blotting (WB) using anti-GFP and anti-HA antibodies, respectively. Input controls (i.e., using 5% of the cell lysates) were included on the blot.

Close modal

p34SEI-1 binds directly to the BIR 2 domain of XIAP. We confirmed that the ubiquitination of XIAP is inhibited by p34SEI-1 (Fig. 2); however, the mechanism by which this protein prevents ubiquitination remains unclear. Therefore, we examined the interaction between p34SEI-1 and XIAP. We detected GFP-tagged p34SEI-1 in HA-XIAP immunoprecipitates from 293 cells that had been cotransfected with constructs expressing these proteins (Fig. 3A). We also examined the interaction between endogenous XIAP and p34SEI-1 in MCF-7 cells (Fig. 3B). Based on these results, we attempted to map the XIAP domains involved in direct binding to p34SEI-1. Previous studies have shown that XIAP contains three BIR domains and one RING-finger domain, each of which has the potential to perform apoptosis-related functions (20). Hence, we used a construct expressing full-length XIAP (HA-XIAP), as well as constructs containing mutant variations of XIAP domains (HA-BIR1, HA-BIR2, HA-BIR3, and HA-RING; Fig. 3C) to determine which domains of XIAP interact with p34SEI-1. After cotransfecting 293 cells with each construct and GFP-tagged p34SEI-1, we observed GFP-tagged p34SEI-1 in immunoprecipitates containing either HA-XIAP or HA-BIR2. However, p34SEI-1-GFP did not interact with HA-BIR1, BIR3, or RING (Fig. 3D), suggesting that p34SEI-1 interacts specifically with the BIR2 domain.

Figure 3.

The interaction between XIAP and p34SEI-1. A, after 293 cells were transfected with HA-XIAP or GFP- p34SEI-1, cell lysates were used for immunoprecipitation-Western blot analyses using anti-HA (i.e., HA-XIAP) and anti-GFP (i.e., GFP- p34SEI-1) antibodies. B, to analyze the molecular interactions between endogenous p34SEI-1 and XIAP, cell lysates from MCF-7 cells were immunoprecipitated with normal serum (i.e., IgG), anti–p34SEI-1 antibody, or anti-XIAP antibody and immunoblotted with anti-XIAP or anti– p34SEI-1 antibodies. C, the XIAP domain mutants used in this study. D, 293 cells were cotransfected with GFP-p34SEI-1 and wild-type HA-XIAP (i.e., wt), as well as either of the HA-BIR1, HA-BIR2, HA-BIR3, or RING domain mutants. Cell lysates were immunoprecipitated with anti-HA (i.e., HA-XIAP) antibody and blotted with anti-GFP (i.e., GFP-p34SEI-1) antibody. Expression of the wild-type and domain mutants was analyzed via Western blot analyses using an anti-HA antibody.

Figure 3.

The interaction between XIAP and p34SEI-1. A, after 293 cells were transfected with HA-XIAP or GFP- p34SEI-1, cell lysates were used for immunoprecipitation-Western blot analyses using anti-HA (i.e., HA-XIAP) and anti-GFP (i.e., GFP- p34SEI-1) antibodies. B, to analyze the molecular interactions between endogenous p34SEI-1 and XIAP, cell lysates from MCF-7 cells were immunoprecipitated with normal serum (i.e., IgG), anti–p34SEI-1 antibody, or anti-XIAP antibody and immunoblotted with anti-XIAP or anti– p34SEI-1 antibodies. C, the XIAP domain mutants used in this study. D, 293 cells were cotransfected with GFP-p34SEI-1 and wild-type HA-XIAP (i.e., wt), as well as either of the HA-BIR1, HA-BIR2, HA-BIR3, or RING domain mutants. Cell lysates were immunoprecipitated with anti-HA (i.e., HA-XIAP) antibody and blotted with anti-GFP (i.e., GFP-p34SEI-1) antibody. Expression of the wild-type and domain mutants was analyzed via Western blot analyses using an anti-HA antibody.

Close modal

p34SEI-1 is significantly overexpressed in human breast cancer tissues. As the overexpression of p34SEI-1 renders breast cancer cells resistant to apoptotic stimuli, whereas repression of p34SEI-1 has the opposite effect, we assessed p34SEI-1 expression in formalin-fixed and paraffin-embedded samples obtained from surgical resections of 60 patients with breast cancer. As shown in Fig. 4A,, i, expression of p34SEI-1 was faint or absent in normal breast tissue and weak in inflammatory cells. Interestingly, immunopositive staining of p34SEI-1 was observed in breast cancer cells (Fig. 4A,, ii) and significant p34SEI-1 overexpression was detected in 32 (53.3%) of the 60 breast cancer tissue samples (Fig. 4B). Similarly, XIAP expression was very low in normal breast tissue but strong in breast cancer cells (Fig. 4A,, iii and iv). Overexpression of XIAP was detected in 30 (50.0%) of the 60 breast cancer tissue samples (Fig. 4B). We examined the coexpression of p34SEI-1 and XIAP in breast cancer cells and found that XIAP was expressed in 25 (78.1%) of the 32 p34SEI-1-positive samples (Fig. 4C). In addition, the proteins were coexpressed in five of eight human breast cancer cell lines (Supplementary Fig. S6), suggesting that p34SEI-1 and XIAP are significantly overexpressed in the tissues of patients with breast cancer.

Figure 4.

Expression of p34SEI-1 and XIAP in breast tissues containing adjacent histologically normal and invasive ductal carcinoma tissues. A positive immunohistochemical reaction is indicated by staining with diaminobenzidine (i.e., brown staining). Hematoxylin was used as a nuclear counterstain. A, representative images from normal and cancerous tissues. A, i, focal and weak immunoreactivity against p34SEI-1 in normal breast epithelial cells. A, ii, the p34 protein was higly expressed in tumors cells. A, iii, normal breast tissue was immunonegative for XIAP. A, iv, tumor cells showed strong cytoplasmic staining for XIAP. B, approximately half (i.e., 32 of 60) of the tumor samples from patients with breast cancer reacted with the anti-p34SEI-1 antibody, whereas 30 of the tumor samples reacted with the anti-XIAP antibody. C, the statistical correlation between the expression of p34SEI-1 and XIAP in human breast cancer was determined using the χ2 test (i.e., P = 0.000003).

Figure 4.

Expression of p34SEI-1 and XIAP in breast tissues containing adjacent histologically normal and invasive ductal carcinoma tissues. A positive immunohistochemical reaction is indicated by staining with diaminobenzidine (i.e., brown staining). Hematoxylin was used as a nuclear counterstain. A, representative images from normal and cancerous tissues. A, i, focal and weak immunoreactivity against p34SEI-1 in normal breast epithelial cells. A, ii, the p34 protein was higly expressed in tumors cells. A, iii, normal breast tissue was immunonegative for XIAP. A, iv, tumor cells showed strong cytoplasmic staining for XIAP. B, approximately half (i.e., 32 of 60) of the tumor samples from patients with breast cancer reacted with the anti-p34SEI-1 antibody, whereas 30 of the tumor samples reacted with the anti-XIAP antibody. C, the statistical correlation between the expression of p34SEI-1 and XIAP in human breast cancer was determined using the χ2 test (i.e., P = 0.000003).

Close modal

Our findings show that p34SEI-1 confers resistance to various apoptotic stimuli on human breast cancer cell lines and 293 cells (Supplementary Figs. S1, S2, and S3; Fig. 1). Furthermore, our data reveal that the antiapoptotic effects of p34SEI-1 result from the up-regulation of XIAP. A recent study aligned the amino acid sequences of various proteins containing the second BIR domain (21), revealing differences in the sequences of XIAP and other proteins. Thus, we examined the binding specificity of p34SEI-1 using XIAP mutants in which certain amino acid residues that are present only in the second BIR domain of XIAP were replaced with alanine (Supplementary Fig. S5A). Interestingly, p34SEI-1 bound weakly to the XIAP mutants (Supplementary Fig. S5B). These results suggest that the specificity of p34SEI-1 may be determined by specific amino acid sequences that exist only in the second BIR domain of XIAP. However, further studies are necessary to explore the details of this mechanism.

In response to apoptotic stimuli, XIAP undergoes proteasome-mediated degradation in cancer cells (19) and consequently promotes apoptosis and caspase activation. Recent studies have shown that the increased stability of XIAP enhances its antiapoptotic activity (7, 22). In the present study, we found that p34SEI-1 inhibited the ubiquitination of XIAP, which is normally induced by apoptotic stimuli. In turn, this increased XIAP levels in p34SEI-1-expressing cells (Fig. 2). These results suggest that p34SEI-1 expression stabilizes XIAP and subsequently confers resistance to apoptosis. This model is supported by our demonstration of specific binding between p34SEI-1 and XIAP (Fig. 3). Our results also show that p34SEI-1 binds to the BIR2 domain of XIAP (Fig. 3D), but does not interact with other domains of XIAP in the absence of BIR2. However, the relationship between p34SEI-1 and other BIR-interacting partners [e.g., Smac/Diablo (23)] remains unclear. These interactions may be influenced by different affinities of these proteins or by other unknown mechanisms. Further research is needed to determine the effects of p34SEI-1 on XIAP antagonists. However, our findings show that p34SEI-1 stabilizes XIAP by binding to the BIR2 domain.

The p34SEI-1 protein rendered cells resistant to apoptotic stimuli and was highly expressed in 32 of 60 samples of human breast cancer examined in this study (Fig. 4). However, this protein is weakly expressed in normal tissues. Furthermore, a significant relationship between p34SEI-1 and XIAP expression was observed in tissue samples from patients with breast cancer (Fig. 4C). Thus, p34SEI-1 may contribute to tumor cell survival by increasing the stability of XIAP and consequently counteracting various apoptotic stimuli. However, as p34SEI-1 expression was unrelated to lymph node metastasis and clinical stage (Supplementary Tables S1–5), it is likely that p34SEI-1 participates solely in the early stages of breast cancer development. Thus, therapeutic strategies that interfere with the expression or function of p34SEI-1 represent promising targets for anticancer drugs and are particularly attractive adjuvant treatments for breast cancer.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Grant support: Research Center for Women's Diseases, at the Korea Science and Engineering Foundation and by a grant from Sookmyung Women's University (2007).

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.

We thank Dr. Colin S Duckett, of the University of Michigan Medical School, for kindly providing the XIAP plasmids; and Dr. Rikiro Fukunaga, of the Osaka University of Japan, for providing SEI-1 cDNA.

1
Sugimoto M, Nakamura T, Ohtani N, et al. Regulation of CDK4 activity by a novel CDK4-binding protein, p34(SEI-1).
Genes Dev
1999
;
13
:
3027
–33.
2
Hoglund M, Gorunova L, Andren-Sandberg A, Dawiskiba S, Mitelman F, Johansson B. Cytogenetic and fluorescence in situ hybridization analyses of chromosome 19 aberrations in pancreatic carcinomas: frequent loss of 19p13.3 and gain of 19q13.1-13.2.
Genes Chromosomes Cancer
1998
;
21
:
8
–16.
3
Ried T, Petersen I, Holtgreve-Grez H, et al. Mapping of multiple DNA gains and losses in primary small cell lung carcinomas by comparative genomic hybridization.
Cancer Res
1994
;
54
:
1801
–6.
4
Tang TC, Sham JS, Xie D, et al. Identification of a candidate oncogene SEI-1 within a minimal amplified region at 19q13.1 in ovarian cancer cell lines.
Cancer Res
2002
;
62
:
7157
–61.
5
Tang DJ, Hu L, Xie D, et al. Oncogenic transformation by SEI-1 is associated with chromosomal instability.
Cancer Res
2005
;
65
:
6504
–8.
6
Dan HC, Sun M, Kaneko S, et al. Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP).
J Biol Chem
2004
;
279
:
5405
–12.
7
Liu WH, Hsiao HW, Tsou WI, Lai MZ. Notch inhibits apoptosis by direct interference with XIAP ubiquitination and degradation.
EMBO J
2007
;
26
:
1660
–9.
8
Deveraux QL, Reed JC. IAP family proteins-suppressor of apoptosis.
Genes Dev
1999
;
13
:
239
–52.
9
Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis.
Nat Rev Mol Cell Biol
2004
;
5
:
897
–9.
10
Chai J, Shiozaki E, Srinivasula SM, et al. Structural basis of caspase-7 inhibition by XIAP.
Cell
2001
;
104
:
769
–80.
11
Scott FL, Denault JB, Riedl SJ, Shin H, Renatus M, Salvesen GS. XIAP inhibits caspase-3 and -7 using two binding sites: evolutionarily conserved mechanism of IAPs.
EMBO J
2005
;
24
:
645
–55.
12
Bockbrader KM, Tan M, Sun Y. A small molecule Smac-mimic compound induces apoptosis and sensitizes TRAIL- and etoposide-induced apoptosis in breast cancer cells.
Oncogene
2005
;
24
:
7381
–8.
13
Schimmer AD, Dalili S, Batey RA, Riedl SJ. Targeting XIAP for the treatment of malignancy.
Cell Death Differ
2006
;
13
:
179
–88.
14
Yang QH, Du C. Smac/DIABLO selectively reduces the levels of c-IAP-1 and c-IAP-2 but not that of XIAP and Livin in HeLa cells.
J Biol Chem
2004
;
279
:
16963
–70.
15
Hong SW, Jin DH, Hahm ES, et al. Ascorbate(vitamin C) induces cell death through the apoptosis-inducing factor in breast cancer cells.
Oncol Rep
2007
;
18
:
811
–5.
16
Park WS, Oh RR, Park JY. Somatic mutations of the trefoil factor family 1 gene in gastric cancer.
Gastroenterology
2000
;
119
:
691
–8.
17
Jung MS, Jin DH, Chae HD, et al. Bcl-xL and E1–19K proteins inhibit p53-induced irreversible growth arrest and senescence by preventing reactive oxygen species-dependent p38 activation.
J Biol Chem
2004
;
279
:
17765
–71.
18
Roos-Mattjus P, Sistonen L. The ubiquitin-proteasome pathway.
Ann Med
2004
;
36
:
285
–95.
19
Yang Y, Fang S, Jensen JP, Weissman AM, Ashwell JD. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli.
Science
2000
;
288
:
874
–7.
20
Takahashi R, Deveraux Q, Tamm I, et al. A single BIR domain of XIAP sufficient for inhibiting caspases.
J Biol Chem
1998
;
273
:
7787
–90.
21
Verhagen AM, Coulson EJ, Vaux DL. Inhibitor of apoptosis proteins and their relatives: IAPs and other BIRPs. Genome Biol 2001;2: reviews 3009.1-3009.10.
22
Dohi T, Okada K, Xia F, et al. An IAP-IAP complex inhibits apoptosis.
J Biol Chem
2004
;
279
:
34087
–90.
23
Du C, Fang M, Li Y, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition.
Cell
2000
;
102
:
33
–42.