The cell-cycle regulator Cks1 has recently been implicated in Skp2-mediated ubiquitination of the tumor suppressor protein p27. In this article, we report that Cks1 exerts a Skp2-independent regulation of NF-κB that promotes interleukin-8 (IL-8) expression, which is critical to hepatocellular carcinoma (HCC) growth. Cks1 was upregulated frequently in human HCC tissues and cell lines. Cks1 knockdown in HCC cells elevated p27 levels and decreased tumorigenicity in a manner that was also associated with a strong downregulation of IL-8 expression. IL-8 downregulation was not phenocopied by either RNAi-mediated knockdown of Skp2 or ectopic overexpression of p27. However, attenuation of IL-8 expression itself was sufficient to blunt HCC growth. Mechanistic investigations revealed that IL-8 was controlled at a transcriptional level by Cks1 targeting of the NF-κB regulator IκBα, which led to NF-κB activation and IL-8 expression, through a p27-independent regulation of IκB kinase complex components. Collectively, our findings support the hypothesis that Cks1 supports hepatocarcinogenesis by NF-κB–mediated regulation of IL-8 expression, broadening the function of Cks1 in cancer beyond its role as a Skp2 cofactor in p27 ubiquitination. Cancer Res; 71(21); 6827–35. ©2011 AACR.

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide (1). The major risk factors for HCC include chronic hepatitis from hepatitis B or C virus infection and exposure to carcinogens such as aflatoxin B1 (2). Despite the clinical significance of HCC, we only know the elemental basics of the molecular, cellular, and environmental mechanisms that drive disease pathogenesis, and only limited therapeutic options are available, of which many have negligible clinical benefits (3). Therefore, elucidation of the predominant molecular events underlying hepatocarcinogenesis may help identify new therapeutic targets.

Cks proteins were originally identified as subunits that interact closely with cyclin–cyclin-dependent kinase (CDK) complexes (4, 5). Recently, mammalian Cks1 (CDC28 protein kinase regulatory subunit 1B), located on chromosome 1q21.2, was identified as an essential cofactor of Skp2 in the Skp2-containing Skp1-Cullin1-F-box ubiquitin ligase complex (SCFSkp2)–mediated ubiquitination of the tumor suppressor protein p27 that leads to the proteasomal degradation of p27 (6–8). The CDK inhibitor p27 is a hallmark of many cancers, and its reduced levels are associated with high aggressiveness and poor prognosis in various malignant tumors (9). Similarly, upregulation of Skp2 and/or Cks1 is significantly associated with poor prognosis in some cancers and, often, inversely related to p27 protein levels (10–15). Among these malignancies, HCC showed a frequent and large gain of 1q, and 1q21-22 was identified as the minimum overlapping amplified region containing candidate oncogenes for hepatocarcinogenesis (16). Moreover, some microarray studies that characterized the global gene expression in human HCC tissues and HCC-derived cell lines showed that Cks1 was upregulated in HCC (17, 18). Together, these findings indicate a significant role of Cks1 in hepatocarcinogenesis.

Interleukin-8 (IL-8), a proinflammatory CXC chemokine, plays important roles in angiogenesis, mitosis, tissue remodeling, and tumor progression (19). IL-8 secreted from cancer cells plays either autocrine or paracrine roles in the tumor microenvironment of various malignancies, including prostate, colon, and pancreatic cancers (20). Especially in HCC, IL-8 derived from cancer cells was significantly implicated in invasion and metastasis, rather than in tumor angiogenesis (21), and the preoperative serum IL-8 level was suggested as a biomarker of tumor invasiveness and progression (22). However, the relationship between Cks1 and IL-8 has not yet been reported.

In this study, we assessed Cks1 expression in HCC tissues and investigated the role of Cks1 in hepatocarcinogenesis, using genetic manipulation of Cks1 in HCC cell lines. Furthermore, we identified the mechanism of Cks1 oncogenesis in HCC development through analysis of gene expression profile and the relevant signaling pathways.

Tissue samples

Fifteen pairs of HCC tissues and the corresponding nonneoplastic liver tissues were obtained from patients who underwent surgical resection at the Chonbuk National University Hospital, Jeonju, Korea (Supplementary Table S1), and the Hospital Research Ethics Committee approved the study. Written informed consent was obtained from all patients. Surgically removed tissues were sampled for histologic diagnosis and the remaining tissues were immediately cut into small pieces, snap-frozen in liquid nitrogen, and stored until use. All protocols conformed to the ethical guidelines of the Institutional Review Board.

Cell lines and culture conditions

The human HCC cell lines Huh7, SNU387, and SNU475, in addition to human foreskin fibroblasts, were purchased from the Korea Cell Line Bank (Seoul, Korea). The human HCC cell lines HepG2, Hep3B, and SK-Hep1 as well as the human fibrosarcoma cell line HT1080 were purchased from the American Type Culture Collection. The cell lines were maintained in Dulbecco's Modified Eagle's Medium (DMEM) supplemented with FBS and antibiotics (Invitrogen) in a humidified atmosphere of 5% CO2 at 37°C. Human umbilical vein endothelial cells (HUVEC) were obtained from Lonza and maintained in EGM2 (Lonza). The 293FT was purchased from Invitrogen and maintained according to the manufacturer's instructions. The cell lines have been characterized at the bank by DNA fingerprinting analysis using short tandem repeat (STR) markers. All cell lines were placed under cryostage after they were purchased from the bank and used within 6 months of thawing fresh vials.

Northern blot

Total RNA was isolated from cells or tissues using TRIzol Reagent (Invitrogen) according to the manufacturer's instructions. Equal amounts of total RNA (10 μg/lane for tissues and 20 μg/lane for cells) were resolved on 1% denaturing formaldehyde–agarose gels and transferred to a Hybond-N membrane (GE Healthcare). The IL-8 probe was prepared from Huh7 total RNA by reverse transcription PCR using the following primers: (sense) 5′-ATGACTTCCAAGCTGGCCG-3′ and (antisense) 5′-TTATGAATTCTCAGCCCTC-3′. Probes for Cks1 and p27 were prepared with the primers used for constructing pLenti-C and pLenti-p27, respectively. Probes were labeled using the Prime-It II Random Primer Labeling Kit (Agilent Technologies) and hybridized in ExpressHyb hybridization solution (Clontech) at 60°C for 4 hours. Filters were washed with 2× SSC–0.05% SDS at room temperature for 30 minutes and with 0.1× SSC–0.1% SDS at 50°C for 30 minutes (1× SSC: 0.15 mol/L NaCl, 0.015 mol/L sodium citrate). mRNA expression was quantified by autoradiography. 18S rRNA served as a normalization control; the primers for probe preparation were as follows: (sense) 5′-GTAACCCGTTGAACCCCATT-3′ and (antisense) 5′-CCATCCAATCGGTAGTAGCG-3′.

Microarray study

Total RNA (10 μg), prepared using an RNeasy Mini kit and RNase-free DNase I (Qiagen), was hybridized to the CodeLink human whole genome bioarray (55K; GE Healthcare). We analyzed significant changes in gene expression by Hierarchical Clustering using Significance Analysis of Microarrays (SAM) software.

Reverse transcription PCR

Reverse transcription was conducted with total RNA (2 μg), according to the manufacturer's instructions (SuperScript III Reverse transcriptase; Invitrogen), and the reaction mixture was used for the following PCR (ExTaq polymerase; TakaRa).

Western blot analysis and ELISA

To obtain secreted IL-8 protein, 80% confluent cultures of cells were washed with PBS 3 times and incubated in DMEM for 72 hours in 100-mm culture dishes. Conditioned media were harvested and concentrated using Vivaspin 6 (5 kDa molecular weight cut-off; Sartorius-Stedim Biotech). For cellular proteins, cells were washed with PBS 3 times and lysed with lysis buffer (M-PER; Thermo Scientific) according to the manufacturer's instructions. Protein concentration was measured with a Bio-Rad Protein Assay. Approximately 20 to 30 μg of protein was separated using SDS-PAGE and transferred onto nitrocellulose membranes (Bio-Rad Laboratories). Western blot analysis was carried out using ECL reagents (GE Healthcare). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) served as a loading control. In addition, IL-8 was quantified by ELISA. A total of 3 × 105 cells were plated in each well of a 6-well plate in DMEM containing 10% FBS. After 8-hour incubation at 37°C, cells were washed with PBS 3 times and subsequently incubated in DMEM containing 0.1% FBS for 18 hours. After pretreatment with BAY 11-7082 (a specific inhibitor of IκBα phosphorylation) or dimethyl sufloxide (vehicle) for 30 minutes, cells were further incubated in 10% FBS for 5 hours, and the amount of IL-8 secreted into the media was analyzed with an OptEIA human IL-8 ELISA kit (BD Biosciences) according to the manufacturer's instructions.

Cell proliferation assay

Cultured cells were incubated in DMEM containing 0.1% FBS in 6-well plates (2 × 104 cells per well, in triplicate) for 18 hours at 37°C in 5% CO2, and subsequently cultured for the indicated times in DMEM containing 10% FBS. The number of viable cells was counted using trypan blue exclusion assay.

In vitro invasion assay

Cultured cells were incubated for 18 hours in DMEM containing 0.1% FBS. On the day of the assay, we coated the top of Transwell polycarbonate membrane inserts (6.5-mm diameter and 8-μm pores; Corning) with Matrigel (40 μg/well; BD Biosciences). After adding DMEM containing 10% FBS into the lower wells, single-cell suspensions in DMEM containing 0.1% FBS were placed on the membrane filters (1 × 105 cells/100 μL/well) and incubated for 23 hours at 37°C in 5% CO2. Filters were washed, and the cells on the upper surface were manually removed with cotton swabs. Cells that had invaded and adhered to the lower surface were fixed with methanol for 15 minutes and stained with 0.1% (w/v) crystal violet for 15 minutes. The filters were extracted with 30% acetic acid. The cells that had invaded were indirectly quantified by determining the absorbance at 595 nm.

In vitro colony-forming assay

The cell suspension (2 × 104 cells in 2 mL of DMEM supplemented with 10% FBS and 0.3% agar) was plated onto 60-mm dishes containing 5 mL of DMEM with 10% FBS and 0.5% agar. The dishes were incubated at 37°C in 5% CO2 for 21 or 28 days. Colonies were observed under a phase-contrast microscope.

Promoter activity assay

A total of 1 × 106 Huh7 cells were plated in a 6-well plate on the day of transfection and were incubated for 8 hours. Cells were subsequently cotransfected with 1 μg of a luciferase reporter vector [pNF-κB-Luc (Stratagene), pAP1-Luc (Stratagene), or pIL8-Luc] and 1 μg of RSV-LacZ (a kind gift from Jae-B. Kim, Seoul National University, Seoul, Korea), using Lipofectamine 2000 (Invitrogen). Transfected cells were incubated in fresh medium. After 24 hours, both luciferase and β-galactosidase assays were conducted according to the manufacturer's instructions (Promega). The β-galactosidase assay was conducted to normalize transfection efficiencies. pIL8-Luc was constructed by inserting the minimal IL-8 promoter (∼350 bp) into the pGL3-Basic vector (Promega). The IL-8 promoter region was obtained by PCR using Huh7 genomic DNA prepared by a Wizard SV Genomic DNA Purification System (Promega) and the following primers: (sense) 5′-GATGCTAGCGATAATTCACCAAATTGTGGAG-3′ and (antisense) 5′-GATCTCGAGGTTTACACACAGTGAGATGGT-3′.

Implantation of tumors

Male 5-week-old athymic BALB/c nu/nu nude mice (Charles River Laboratories Japan Inc.) were used for xenograft studies. All mice were fed a commercial diet, given water ad libitum, and subjected to a 12-hour light/12-hour dark cycle. Mice (n = 5/group) were s.c. injected with 5 × 106 cells in the proximal midline of the dorsa. We measured tumor sizes every 2 to 3 days and estimated tumor volumes as width2 × length × 0.52.

Statistical analysis

Data are presented as the mean ± SD or SEM. Statistical significance was calculated using the Student t test. Values of P < 0.05 were considered statistically significant.

Cks1 is overexpressed in human HCC

Cks1 overexpression occurs in various cancers, including oral squamous cell, lung, and gastric carcinomas. To investigate the potential role of Cks1 in hepatocarcinogenesis, we determined the level of Cks1 in HCC tissues and cell lines by Northern blot analysis. Cks1 expression was substantially higher in HCC tissues than in the corresponding nontumor liver tissues in 13 of the 15 pairs examined (Fig. 1A; 5 representative cases; Supplementary Fig. S1 for all cases). Furthermore, Cks1 was overexpressed in human HCC cell lines, including Huh7, HepG2, Hep3B, SK-Hep1, SNU387, and SNU475, relative to the levels in human normal liver tissue and nontumorigenic HUVECs and fibroblasts (Fig. 1B). These results indicate that Cks1 overexpression may be associated with hepatocarcinogenesis.

Figure 1.

Cks1 is overexpressed in human HCC. A, Cks1 expression in human HCC tissues. Primary HCC tissues (T) and the matched surrounding nontumor liver tissues (N) of 15 patients were analyzed for Cks1 expression by Northern blot analysis. Five representative cases are shown (#1–5). B, Cks1 expression in human HCC cell lines, compared with human normal liver tissue and nontumorigenic cells. 18S rRNA served as a loading control.

Figure 1.

Cks1 is overexpressed in human HCC. A, Cks1 expression in human HCC tissues. Primary HCC tissues (T) and the matched surrounding nontumor liver tissues (N) of 15 patients were analyzed for Cks1 expression by Northern blot analysis. Five representative cases are shown (#1–5). B, Cks1 expression in human HCC cell lines, compared with human normal liver tissue and nontumorigenic cells. 18S rRNA served as a loading control.

Close modal

Cks1 correlates with in vitro proliferation rates of HCC cells

To investigate the role of Cks1 in the regulation of cellular proliferation of HCC cells, we conducted ectopic overexpression and knockdown of Cks1 in Huh7 cells using lentiviral expression systems. Northern blot analysis confirmed the stable overexpression or knockdown of Cks1 in Huh7 cells. As shown in Fig. 2A (top), the ectopically overexpressed Cks1 mRNA transcript (V: the 6× histidine-tagged form in the C-terminus) was clearly discernible from the endogenous form. In vitro proliferation assays revealed that Cks1 overexpression significantly enhanced cellular proliferation (Fig. 2A, bottom). In Cks1 knockdown, both short hairpin RNAs (shRNA; shC-#1 and shC-#2) efficiently suppressed Cks1 mRNA expression compared with control cells (Fig. 2B, top), and the resulting Cks1-knockdown Huh7 cells showed significantly lower proliferation ability than control cells (Fig. 2B, bottom). Cks1 was also positively correlated with cellular proliferation of SK-Hep1 cells (Supplementary Fig. S2). These results imply that Cks1 plays an important role in HCC cell proliferation.

Figure 2.

Cks1 correlates with in vitro proliferation rates of Huh7 cells. Construction of (A) Cks1-overexpressing and (B) Cks1-knockdown Huh7 cells and in vitro proliferation assays (V, lentivirally overexpressed form; C, endogenous form). Cks1 level was determined by Northern blot analysis. shC-#1 and shC-#2 indicate shRNA I and II, respectively. For proliferation assay, cells were incubated for 8 days in DMEM containing 10% FBS after starvation in DMEM containing 0.1% FBS for 18 hours at 37°C in 5% CO2. The number of viable cells was counted using trypan blue exclusion assay. The mean ± SD (n = 3) is shown. +, P < 0.05; *, P < 0.0005; **, P < 0.002 (vs. Mock and shMock, respectively).

Figure 2.

Cks1 correlates with in vitro proliferation rates of Huh7 cells. Construction of (A) Cks1-overexpressing and (B) Cks1-knockdown Huh7 cells and in vitro proliferation assays (V, lentivirally overexpressed form; C, endogenous form). Cks1 level was determined by Northern blot analysis. shC-#1 and shC-#2 indicate shRNA I and II, respectively. For proliferation assay, cells were incubated for 8 days in DMEM containing 10% FBS after starvation in DMEM containing 0.1% FBS for 18 hours at 37°C in 5% CO2. The number of viable cells was counted using trypan blue exclusion assay. The mean ± SD (n = 3) is shown. +, P < 0.05; *, P < 0.0005; **, P < 0.002 (vs. Mock and shMock, respectively).

Close modal

Cks1 knockdown increases p27 expression and decreases HCC cell tumorigenicity

Because Cks1 is known as a cofactor of Skp2 in the SCFSkp2-mediated ubiquitination of p27, we examined the effect of Cks1 knockdown on p27 level in Huh7 cells by Western blot analysis. Cks1 knockdown increased p27 levels, indicating a role of Cks1 in the regulation of p27 degradation (Fig. 3A). Although CDK1, 2, and 4 were unaffected by Cks1 knockdown, cyclin E was slightly increased, probably because of the decrease in its SCFSkp2-mediated ubiquitination leading to proteasomal degradation (23). Cks1 knockdown also increased active caspase 3, suggesting that this knockdown may induce cellular apoptosis. To investigate the mechanism of p27 increase by Cks1 knockdown, we treated cells with 10 μmol/L MG132 (a specific proteasome inhibitor) for 7 hours, which clearly abolished the Cks1 knockdown–induced p27 increase in Huh7 cells (Fig. 3B). This indicates that the Cks1 knockdown–induced p27 increase was likely due to decreased proteasomal degradation.

Figure 3.

Cks1 knockdown increases p27 in Huh7 cells. Western blot analysis of cell-cycle–related proteins (A) and p27 in the presence (+) or absence (−) of MG132 (B). GAPDH served as a loading control. shM indicates mock cells; shC7, shC9, and shC11 indicate Cks1-knockdown clones; and MG132, a specific proteasome inhibitor.

Figure 3.

Cks1 knockdown increases p27 in Huh7 cells. Western blot analysis of cell-cycle–related proteins (A) and p27 in the presence (+) or absence (−) of MG132 (B). GAPDH served as a loading control. shM indicates mock cells; shC7, shC9, and shC11 indicate Cks1-knockdown clones; and MG132, a specific proteasome inhibitor.

Close modal

Because p27 is an established tumor suppressor, we next assessed the effects of Cks1 knockdown on Huh7 cell tumorigenicity. In a soft agar assay, Cks1-knockdown clones (shC7, shC9, and shC11) showed substantially impaired colony-forming abilities as compared with control cells (Fig. 4A), indicating that Cks1 contributes to the anchorage-independent growth of Huh7 cells. Furthermore, SK-Hep1 cells exhibited a positive correlation between Cks1 and in vitro colony-forming activity (Supplementary Fig. S3). Cks1 knockdown significantly decreased the in vitro invasion ability of Huh7 cells stimulated by 10% FBS (Fig. 4B). In mouse xenograft tumor experiments, Cks1 knockdown substantially inhibited the growth of solid tumors in vivo by more than 50% as compared with tumors from mock cells (Fig. 4C; Supplementary Fig. S4 for results of immunohistochemical analysis). Thus, Cks1 overexpression may lead to hepatocarcinogenesis by directly enhancing the cellular tumorigenic potential.

Figure 4.

Cks1 knockdown decreases the tumorigenicity of Huh7 cells. A, soft agar assay. In vitro colony-forming activities were compared after 21 days of incubation (n = 3). B, in vitro invasion assay. Invasion was stimulated by 10% FBS for 23 hours after 18-hour starvation in DMEM containing 0.1% FBS at 37°C in 5% CO2. The mean ± SD (n = 3) is shown. *, P < 0.0005 (vs. shMock). C, in vivo tumor growth. Cells were implanted s.c. in the proximal midline of the dorsa of nude mice. The mean ± SEM (n = 5) is shown. *, P < 0.0005; **, P < 0.02 [vs. shM2 (mock clone 2)].

Figure 4.

Cks1 knockdown decreases the tumorigenicity of Huh7 cells. A, soft agar assay. In vitro colony-forming activities were compared after 21 days of incubation (n = 3). B, in vitro invasion assay. Invasion was stimulated by 10% FBS for 23 hours after 18-hour starvation in DMEM containing 0.1% FBS at 37°C in 5% CO2. The mean ± SD (n = 3) is shown. *, P < 0.0005 (vs. shMock). C, in vivo tumor growth. Cells were implanted s.c. in the proximal midline of the dorsa of nude mice. The mean ± SEM (n = 5) is shown. *, P < 0.0005; **, P < 0.02 [vs. shM2 (mock clone 2)].

Close modal

Cks1 regulates IL-8 expression in an Skp2-independent mode

To obtain genome-wide insights on Cks1 function(s) in hepatocarcinogenesis, we conducted a microarray study with mock (shM2) and Cks1-knockdown (shC9) Huh7 clones. Cks1 knockdown affected gene expression by more than 2 times in 1,432 of the total 52,486 gene probes, with 108 increased and 1,324 decreased (Supplementary Tables S2 and S3). Among the affected genes, IL-8 was one of the most downregulated genes (9.8-fold), and this was further confirmed by both Northern and Western blot analyses (Fig. 5A). To investigate whether IL-8 downregulation resulted from the loss of Cks1 function as an Skp2 cofactor, we prepared Skp2-knockdown and p27-overexpressing Huh7 cells, as confirmed by Western blot analysis (Fig. 5B and C, left). However, neither Skp2-knockdown nor p27-overexpressing Huh7 cells showed IL-8 downregulation (Fig. 5B and C, right). This may be partly attributable to the fact that, unlike Cks1 knockdown, either p27 overexpression or Skp2 knockdown was not accompanied by Cks1 decrease, as assessed by Western blot analysis (Supplementary Fig. S5). In contrast to IL-8, osteopontin (OPN), another protein substantially downregulated by Cks1 knockdown in Huh7 cells, showed p27-dependent expression in Huh7 cells, showing a different regulation mechanism from that of IL-8 (Supplementary Fig. S6). These results indicate that Cks1 may regulate IL-8 expression in Huh7 cells through an unknown function other than an Skp2 cofactor in p27 ubiquitination.

Figure 5.

Cks1 regulates IL-8 expression in an Skp2-independent mode in Huh7 cells. A, effect of Cks1 knockdown on IL-8 expression. Northern (left) and Western (right) blot analysis. B and C, effect of p27 overexpression (B) or Skp2 knockdown (C) on IL-8 expression. Each genetic modification was confirmed by Western blot analysis (left). IL-8(s) indicates IL-8 protein secreted into the serum-free culture media (right). The 27-1, 27-2, 27-4, 27-5, and 27-6 indicate p27-overexpressing clones. S2 to S8 indicate Skp2-knockdown clones. 18S rRNA and GAPDH served as loading controls for Northern and Western blotting, respectively.

Figure 5.

Cks1 regulates IL-8 expression in an Skp2-independent mode in Huh7 cells. A, effect of Cks1 knockdown on IL-8 expression. Northern (left) and Western (right) blot analysis. B and C, effect of p27 overexpression (B) or Skp2 knockdown (C) on IL-8 expression. Each genetic modification was confirmed by Western blot analysis (left). IL-8(s) indicates IL-8 protein secreted into the serum-free culture media (right). The 27-1, 27-2, 27-4, 27-5, and 27-6 indicate p27-overexpressing clones. S2 to S8 indicate Skp2-knockdown clones. 18S rRNA and GAPDH served as loading controls for Northern and Western blotting, respectively.

Close modal

IL-8 contributes to the oncogenic activities of Cks1

To investigate the contribution of IL-8 to the Cks1 oncogenesis, we carried out in vitro tumorigenicity assays with IL-8–knockdown Huh7 cells. Stable IL-8–knockdown Huh7 cells were established and confirmed by Western and Northern blot analyses (Fig. 6A). Neither the empty vector (pLL3.7B) nor the nonspecific shRNA-expressing vector (pLL3.7B*) affected IL-8 expression (shM and shM*, respectively), compared with the expression in the parental Huh7 cells. This indicates that the Cks1 knockdown–derived IL-8 reduction was not a nonspecific effect from the shRNA expression process itself in the vector system. IL-8 knockdown significantly inhibited both in vitro proliferation and invasion abilities of Huh7 cells, compared with control cells (Fig. 6B). Further evaluation showed that IL-8 also plays a critical role in the anchorage-independent Huh7 cell growth (Fig. 6C). These results suggest that IL-8 significantly contributes to hepatocarcinogenesis by mediating the oncogenic activities of Cks1.

Figure 6.

IL-8 contributes to the tumorigenicity of Huh7 cells. A, construction of IL-8–knockdown Huh7 cells [Western (top) and Northern blotting (bottom)]. B, in vitro proliferation (top) and invasion (bottom) assays. The mean ± SD (n = 3) is shown. NS, not stimulated. C, soft agar assay. In vitro colony-forming activities were compared after 28 days of incubation (n = 3). shM* indicates nonspecific shRNA-expressing mock cells. shI1–4 indicate IL-8–knockdown clones. +, P < 0.002; *, P < 0.000002; **, P < 0.005 (vs. shM*).

Figure 6.

IL-8 contributes to the tumorigenicity of Huh7 cells. A, construction of IL-8–knockdown Huh7 cells [Western (top) and Northern blotting (bottom)]. B, in vitro proliferation (top) and invasion (bottom) assays. The mean ± SD (n = 3) is shown. NS, not stimulated. C, soft agar assay. In vitro colony-forming activities were compared after 28 days of incubation (n = 3). shM* indicates nonspecific shRNA-expressing mock cells. shI1–4 indicate IL-8–knockdown clones. +, P < 0.002; *, P < 0.000002; **, P < 0.005 (vs. shM*).

Close modal

Cks1 controls IL-8 expression through the unique regulation of the NF-κB pathway

The promoter of the IL-8 gene contains several transcription factor–binding sequences and, among these factors, NF-κB and AP1 play central roles in regulating IL-8 gene expression (24). To investigate the regulatory mechanism of IL-8 expression by Cks1, we determined the effect of Cks1 knockdown on luciferase expression under the control of NF-κB enhancer, AP1 enhancer, or minimal IL-8 promoter including both enhancers and found that Cks1 knockdown significantly decreased IL-8 promoter activity in Huh7 cells, which seemed to be derived mainly by the decreased activity of NF-κB, not AP1 (Fig. 7A; Supplementary Fig. S7). To confirm the involvement of the NF-κB pathway in the regulation of IL-8 expression, we measured IL-8 secretion in the presence or absence of BAY 11-7082 (an inhibitor of IκBα phosphorylation). BAY 11-7082 substantially decreased IL-8 expression in a dose-dependent manner, as determined by ELISA (Fig. 7B). Moreover, Western blot analysis showed that Cks1 knockdown significantly decreased the kinase subunits, both IKKα and IKKβ, and the regulatory subunit, IKKγ, of the IKK complex, whereas p27 overexpression decreased only IKKγ (Fig. 7C). This implies that the Cks1 knockdown–induced IKKγ decrease resulted from the Skp2 dysfunction-induced p27 increase. Furthermore, we examined NF-κB p65 phosphorylation status important for its transcriptional activity, but did not find any differences between control and Cks1-knockdown cells (Supplementary Fig. S8). Next, we compared IκBα phosphorylation, the rate-limiting step in NF-κB activation, between control and Cks1-knockdown cells, which showed that Cks1 knockdown significantly reduced IκBα phosphorylation compared with control cells (Fig. 7D). Therefore, it can be hypothesized that the proteasomal degradation of IκBα via IKK complex-mediated phosphorylation was suppressed by Cks1 knockdown through the downregulation of IKK complex, eventually leading to NF-κB inhibition and, thereby, IL-8 downregulation. These results revealed a novel function of Cks1 in the regulation of the NF-κB pathway.

Figure 7.

Cks1 controls IL-8 expression via NF-κB pathway in Huh7 cells. A, promoter activity assay. shM2 indicates mock clone 2 and shC9 indicates Cks1-knockdown clone 9. Each data point was normalized by the corresponding control data from shM2. B, effect of BAY 11-7082 on IL-8 expression. Cells were treated with BAY 11-7082 (a specific inhibitor of IκBα phosphorylation) for 5 hours at concentrations of 0 (B0), 25 (B25), and 50 μmol/L (B50), and secreted IL-8 was measured by ELISA. The mean ± SD (n = 3) is shown. *, P < 0.002; **, P < 0.01; +, P < 0.000005 (vs. shM2 and B0, respectively). C, effects of Cks1 knockdown (top) or p27 overexpression (bottom) on the components of the NF-κB pathway (Western blotting). D, Western blot analysis of IκBα phosphorylation when stimulated with 10% FBS for 15 minutes after 18-hour starvation in DMEM containing 0.1% FBS.

Figure 7.

Cks1 controls IL-8 expression via NF-κB pathway in Huh7 cells. A, promoter activity assay. shM2 indicates mock clone 2 and shC9 indicates Cks1-knockdown clone 9. Each data point was normalized by the corresponding control data from shM2. B, effect of BAY 11-7082 on IL-8 expression. Cells were treated with BAY 11-7082 (a specific inhibitor of IκBα phosphorylation) for 5 hours at concentrations of 0 (B0), 25 (B25), and 50 μmol/L (B50), and secreted IL-8 was measured by ELISA. The mean ± SD (n = 3) is shown. *, P < 0.002; **, P < 0.01; +, P < 0.000005 (vs. shM2 and B0, respectively). C, effects of Cks1 knockdown (top) or p27 overexpression (bottom) on the components of the NF-κB pathway (Western blotting). D, Western blot analysis of IκBα phosphorylation when stimulated with 10% FBS for 15 minutes after 18-hour starvation in DMEM containing 0.1% FBS.

Close modal

The heterogeneous phenotypic and genetic traits of individuals with HCC and the wide range of risk factors associated with HCC have led to HCC being classified as a complex disease (25). In this article, we present a novel tumor-promoting function of Cks1 in hepatocarcinogenesis.

Cks1 reportedly correlates with proliferation of lymphoid, oral squamous cancer, and prostate cancer cells (26–28). Furthermore, Cks1 overexpression is noted in numerous cancers, including multiple myeloma, lung, gastric, and oral carcinomas (13, 27, 29, 30). Recently, Cks1, along with Skp2 and/or p27, was hypothesized to be a potential prognostic marker for some cancers (12, 27, 31, 32). In the present study, Cks1 expression was higher in human HCC tissues than in adjacent nontumor liver tissues. In addition, Cks1 was correlated with the in vitro HCC cell proliferation, and knocking down of Cks1 in Huh7 cells elicited an increase in the p27 protein. MG132 treatment of HCC cells revealed that Cks1 participates in the proteasomal degradation of p27. Consistent with the fact that p27 is a well-known tumor suppressor, Cks1 knockdown significantly inhibited the tumorigenic potential of Huh7 cells both in vitro and in vivo. The in vitro inhibitory effect of Cks1 knockdown on the tumorigenicity of cancer cells was also reported with prostate cancer (28). Recently, the Skp2–Cks1 complex was reported to be involved in the degradation of several tumor suppressor proteins other than p27 (DUSP1, RASSF1A, p130, etc.) in HCC cells (14, 33). Therefore, it may not be excluded that suppressor proteins other than p27 may also participate in the anti-HCC activities of Cks1 knockdown. Our results indicate that Cks1 overexpression may contribute to hepatocarcinogenesis by enhancing the cellular tumorigenic potential, which may be partly imparted by participating in ubiquitination-mediated proteasomal degradation of p27 as a cofactor of Skp2.

In addition to the CDK- or Skp2-dependent function, Cks1 may also function as a transcription activator for maintaining efficient transcriptional activation in Saccharomyces cerevisiae (34–36). This finding prompted us to obtain genome-wide insights on Cks1 function(s) in HCC cells. This analysis revealed that Cks1 knockdown in Huh7 cells significantly changed the expression of genes known to participate in various cellular processes, including gene transcription, cell death, apoptosis, signal transduction, stress response, immune response, cell cycle, and so on. Notably, genes related to proliferation, angiogenesis, inflammation, invasion, and metastasis were significantly downregulated by Cks1 knockdown, implying that Cks1 may play significant and diverse roles in hepatocarcinogenesis. Among these genes, IL-8 was one of the most downregulated genes in Huh7 cells, according to a microarray study and Northern and Western blot analyses.

Autocrine or paracrine roles of IL-8 in tumor growth, invasion, angiogenesis, and/or metastasis have been reported in various tumor types, such as skin, stomach, prostate, hepatic, and pancreatic cancers (37–40). In Ras-transformed cells, Ras-dependent IL-8 secretion has been shown to contribute to tumor growth by initiating tumor-associated inflammation and neovascularization as a paracrine, and not an autocrine, factor (41). In HCC patients, serum IL-8 levels significantly correlated with tumor size and tumor stage, and its preoperative levels were hypothesized to be a useful biologica marker of tumor invasiveness and progression (22). Akiba and colleagues reported IL-8 expression in HCC tissues whose cancer cells were the main source of IL-8, and hypothesized that IL-8 may have an important function in cancer invasion and metastasis, rather than in tumor angiogenesis (21). In the present study, Cks1 knockdown–induced IL-8 downregulation in Huh7 cells was not apparently associated with either Skp2 dysfunction or p27 accumulation, thereby suggesting a novel function of Cks1 in hepatocarcinogenesis. Moreover, IL-8 knockdown significantly suppressed the tumorigenicity of Huh7 cells, indicating that IL-8 may be an important autocrine factor for HCC cells and a key mediator of the Skp2- and p27-independent oncogenic function of Cks1 in hepatocarcinogenesis.

IL-8 is transcriptionally regulated mainly by NF-κB and AP1 (24). In the present study, Cks1 regulated IL-8 expression in Huh7 cells through NF-κB (not AP1)-mediated transcriptional control, although AP1 is reportedly required for constitutive IL-8 gene expression in hepatoma cells (42). IκB proteolysis, the rate-limiting step in NF-κB activation, is regulated by the IKK complex, which is composed of 2 catalytic subunits, IKKα and IKKβ, and 1 regulatory subunit, IKKγ (43). Li and colleagues reported that IKKβ-deficient mice die at mid-gestation from uncontrolled liver apoptosis, a phenotype similar to that of mice deficient in both NF-κB p65 and NF-κB1 (p50/p105; ref. 44). Constitutive activation of NF-κB is seen in several cancers, including prostate cancer, fibrolamellar HCC, and glioblastoma (45–47). Arsura and colleagues reported that constitutive activation of NF-κB in Ras-transformed rat liver epithelial cells was dependent on IKKα and IKKβ activation (48). In addition, Factor and colleagues described an important role of the IKK complex in the constitutive NF-κB activation, which led to HCC development in double transforming growth factor-α and c-myc transgenic mice (49). In the current study, Cks1 controlled IKKα and IKKβ expression independently of p27, in contrast to the p27-dependent control of IKKγ expression, in HCC cells. The p27-induced IKKγ downregulation failed to elicit IL-8 decrease, indicating that IKKα and IKKβ, rather than IKKγ, may play an important role in the NF-κB–mediated transcriptional regulation of IL-8 in HCC cells. However, additional studies are necessary to further investigate the mechanism of how Cks1 regulates the expression of each IKK subunit and the implication of p27-mediated IKKγ regulation in Cks1 function.

In summary, we showed a significant role of Cks1 in hepatocarcinogenesis and identified IL-8 as a key autocrine mediator of the oncogenic function of Cks1. Cks1 regulated IL-8 expression in an Skp2-independent mode, probably through the specific control of IKKα and IKKβ in the NF-κB pathway. Collectively, our study uncovered a novel function of Cks1 in the NF-κB pathway regulation, which may be different from the Skp2 cofactor function in p27 ubiquitination. Thus, Cks1 may be a promising therapeutic target in HCC management.

No potential conflicts of interest were disclosed.

The authors thank J.-H. Lee and H.-K. Joo for skillful technical assistance in animal experiments.

This study was financially supported in part by the Green Cross Corporation (Grant TD03-001-00).

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.
Schutte
K
,
Bornschein
J
,
Malfertheiner
P
. 
Hepatocellular carcinoma–epidemiological trends and risk factors
.
Dig Dis
2009
;
27
:
80
92
.
2.
Thorgeirsson
SS
,
Grisham
JW
. 
Molecular pathogenesis of human hepatocellular carcinoma
.
Nat Genet
2002
;
31
:
339
46
.
3.
Farazi
PA
,
DePinho
RA
. 
Hepatocellular carcinoma pathogenesis: from genes to environment
.
Nat Rev Cancer
2006
;
6
:
674
87
.
4.
Hayles
J
,
Beach
D
,
Durkacz
B
,
Nurse
P
. 
The fission yeast cell cycle control gene cdc2: isolation of a sequence suc1 that suppresses cdc2 mutant function
.
Mol Gen Genet
1986
;
202
:
291
3
.
5.
Hadwiger
JA
,
Wittenberg
C
,
Mendenhall
MD
,
Reed
SI
. 
The Saccharomyces cerevisiae CKS1 gene, a homolog of the Schizosaccharomyces pombe suc1+ gene, encodes a subunit of the Cdc28 protein kinase complex
.
Mol Cell Biol
1989
;
9
:
2034
41
.
6.
Carrano
AC
,
Eytan
E
,
Hershko
A
,
Pagano
M
. 
SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27
.
Nat Cell Biol
1999
;
1
:
193
9
.
7.
Ganoth
D
,
Bornstein
G
,
Ko
TK
,
Larsen
B
,
Tyers
M
,
Pagano
M
, et al
The cell-cycle regulatory protein Cks1 is required for SCF(Skp2)-mediated ubiquitinylation of p27
.
Nat Cell Biol
2001
;
3
:
321
4
.
8.
Spruck
C
,
Strohmaier
H
,
Watson
M
,
Smith
AP
,
Ryan
A
,
Krek
TW
, et al
A CDK-independent function of mammalian Cks1: targeting of SCF(Skp2) to the CDK inhibitor p27Kip1
.
Mol Cell
2001
;
7
:
639
50
.
9.
Philipp-Staheli
J
,
Payne
SR
,
Kemp
CJ
. 
p27(Kip1): regulation and function of a haploinsufficient tumor suppressor and its misregulation in cancer
.
Exp Cell Res
2001
;
264
:
148
68
.
10.
Kudo
Y
,
Kitajima
S
,
Sato
S
,
Miyauchi
M
,
Ogawa
I
,
Takata
T
. 
High expression of S-phase kinase-interacting protein 2, human F-box protein, correlates with poor prognosis in oral squamous cell carcinomas
.
Cancer Res
2001
;
61
:
7044
7
.
11.
Chiarle
R
,
Fan
Y
,
Piva
R
,
Boggino
H
,
Skolnik
J
,
Novero
D
, et al
S-phase kinase-associated protein 2 expression in non-Hodgkin's lymphoma inversely correlates with p27 expression and defines cells in S phase
.
Am J Pathol
2002
;
160
:
1457
66
.
12.
Shapira
M
,
Ben-Izhak
O
,
Linn
S
,
Futerman
B
,
Minkov
I
,
Hershko
DD
. 
The prognostic impact of the ubiquitin ligase subunits Skp2 and Cks1 in colorectal carcinoma
.
Cancer
2005
;
103
:
1336
46
.
13.
Shaughnessy
J
. 
Amplification and overexpression of CKS1B at chromosome band 1q21 is associated with reduced levels of p27Kip1 and an aggressive clinical course in multiple myeloma
.
Hematology
2005
;
10
(
Suppl 1
):
117
26
.
14.
Calvisi
DF
,
Ladu
S
,
Pinna
F
,
Frau
M
,
Tomasi
ML
,
Sini
M
, et al
SKP2 and CKS1 promote degradation of cell cycle regulators and are associated with hepatocellular carcinoma prognosis
.
Gastroenterology
2009
;
137
:
1816
26
.
e1–10
.
15.
Shen
DY
,
Fang
ZX
,
You
P
,
Liu
PG
,
Wang
F
,
Huang
CL
, et al
Clinical significance and expression of cyclin kinase subunits 1 and 2 in hepatocellular carcinoma
.
Liver Int
2009
;
30
:
119
25
.
16.
Wong
N
,
Chan
A
,
Lee
SW
,
Lam
E
,
To
KF
,
Lai
PB
, et al
Positional mapping for amplified DNA sequences on 1q21-q22 in hepatocellular carcinoma indicates candidate genes over-expression
.
J Hepatol
2003
;
38
:
298
306
.
17.
Okabe
H
,
Satoh
S
,
Kato
T
,
Kitahara
O
,
Yanagawa
R
,
Yamaoka
Y
, et al
Genome-wide analysis of gene expression in human hepatocellular carcinomas using cDNA microarray: identification of genes involved in viral carcinogenesis and tumor progression
.
Cancer Res
2001
;
61
:
2129
37
.
18.
Xu
XR
,
Huang
J
,
Xu
ZG
,
Qian
BZ
,
Zhu
ZD
,
Yan
Q
, et al
Insight into hepatocellular carcinogenesis at transcriptome level by comparing gene expression profiles of hepatocellular carcinoma with those of corresponding noncancerous liver
.
Proc Natl Acad Sci U S A
2001
;
98
:
15089
94
.
19.
Baggiolini
M
,
Clark-Lewis
I
. 
Interleukin-8, a chemotactic and inflammatory cytokine
.
FEBS Lett
1992
;
307
:
97
101
.
20.
Waugh
DJ
,
Wilson
C
. 
The interleukin-8 pathway in cancer
.
Clin Cancer Res
2008
;
14
:
6735
41
.
21.
Akiba
J
,
Yano
H
,
Ogasawara
S
,
Higaki
K
,
Kojiro
M
. 
Expression and function of interleukin-8 in human hepatocellular carcinoma
.
Int J Oncol
2001
;
18
:
257
64
.
22.
Ren
Y
,
Poon
RT
,
Tsui
HT
,
Chen
WH
,
Li
Z
,
Lau
C
, et al
Interleukin-8 serum levels in patients with hepatocellular carcinoma: correlations with clinicopathological features and prognosis
.
Clin Cancer Res
2003
;
9
:
5996
6001
.
23.
Nakayama
K
,
Nagahama
H
,
Minamishima
YA
,
Matsumoto
M
,
Nakamichi
I
,
Kitagawa
K
, et al
Targeted disruption of Skp2 results in accumulation of cyclin E and p27(Kip1), polyploidy and centrosome overduplication
.
EMBO J
2000
;
19
:
2069
81
.
24.
Hoffmann
E
,
Dittrich-Breiholz
O
,
Holtmann
H
,
Kracht
M
. 
Multiple control of interleukin-8 gene expression
.
J Leukoc Biol
2002
;
72
:
847
55
.
25.
Aravalli
RN
,
Steer
CJ
,
Cressman
EN
. 
Molecular mechanisms of hepatocellular carcinoma
.
Hepatology
2008
;
48
:
2047
63
.
26.
Urbanowicz-Kachnowicz
I
,
Baghdassarian
N
,
Nakache
C
,
Gracia
D
,
Mekki
Y
,
Bryon
PA
, et al
ckshs expression is linked to cell proliferation in normal and malignant human lymphoid cells
.
Int J Cancer
1999
;
82
:
98
104
.
27.
Kitajima
S
,
Kudo
Y
,
Ogawa
I
,
Bashir
T
,
Kitagawa
M
,
Miyauchi
M
, et al
Role of Cks1 overexpression in oral squamous cell carcinomas: cooperation with Skp2 in promoting p27 degradation
.
Am J Pathol
2004
;
165
:
2147
55
.
28.
Lan
Y
,
Zhang
Y
,
Wang
J
,
Lin
C
,
Ittmann
MM
,
Wang
F
. 
Aberrant expression of Cks1 and Cks2 contributes to prostate tumorigenesis by promoting proliferation and inhibiting programmed cell death
.
Int J Cancer
2008
;
123
:
543
51
.
29.
Inui
N
,
Kitagawa
K
,
Miwa
S
,
Hattori
T
,
Chida
K
,
Nakamura
H
, et al
High expression of Cks1 in human non-small cell lung carcinomas
.
Biochem Biophys Res Commun
2003
;
303
:
978
84
.
30.
Masuda
TA
,
Inoue
H
,
Nishida
K
,
Sonoda
H
,
Yoshikawa
Y
,
Kakeji
Y
, et al
Cyclin-dependent kinase 1 gene expression is associated with poor prognosis in gastric carcinoma
.
Clin Cancer Res
2003
;
9
:
5693
8
.
31.
Chang
H
,
Qi
X
,
Trieu
Y
,
Xu
W
,
Reader
JC
,
Ning
Y
, et al
Multiple myeloma patients with CKS1B gene amplification have a shorter progression-free survival post-autologous stem cell transplantation
.
Br J Haematol
2006
;
135
:
486
91
.
32.
Liu
Z
,
Fu
Q
,
Lv
J
,
Wang
F
,
Ding
K
. 
Prognostic implication of p27Kip1, Skp2 and Cks1 expression in renal cell carcinoma: a tissue microarray study
.
J Exp Clin Cancer Res
2008
;
27
:
51
.
33.
Calvisi
DF
,
Pinna
F
,
Meloni
F
,
Ladu
S
,
Pellegrino
R
,
Sini
M
, et al
Dual-specificity phosphatase 1 ubiquitination in extracellular signal-regulated kinase-mediated control of growth in human hepatocellular carcinoma
.
Cancer Res
2008
;
68
:
4192
200
.
34.
Morris
MC
,
Kaiser
P
,
Rudyak
S
,
Baskerville
C
,
Watson
MH
,
Reed
SI
. 
Cks1-dependent proteasome recruitment and activation of CDC20 transcription in budding yeast
.
Nature
2003
;
423
:
1009
13
.
35.
Yu
VP
,
Baskerville
C
,
Grunenfelder
B
,
Reed
SI
. 
A kinase-independent function of Cks1 and Cdk1 in regulation of transcription
.
Mol Cell
2005
;
17
:
145
51
.
36.
Holic
R
,
Kukalev
A
,
Lane
S
,
Andress
EJ
,
Lau
I
,
Yu
CW
, et al
Cks1 activates transcription by binding to the ubiquitylated proteasome
.
Mol Cell Biol
2010
;
30
:
3894
901
.
37.
Luca
M
,
Huang
S
,
Gershenwald
JE
,
Singh
RK
,
Reich
R
,
Bar-Eli
M
. 
Expression of interleukin-8 by human melanoma cells up-regulates MMP-2 activity and increases tumor growth and metastasis
.
Am J Pathol
1997
;
151
:
1105
13
.
38.
Miyamoto
M
,
Shimizu
Y
,
Okada
K
,
Kashii
Y
,
Higuchi
K
,
Watanabe
A
. 
Effect of interleukin-8 on production of tumor-associated substances and autocrine growth of human liver and pancreatic cancer cells
.
Cancer Immunol Immunother
1998
;
47
:
47
57
.
39.
Kitadai
Y
,
Takahashi
Y
,
Haruma
K
,
Naka
K
,
Sumii
K
,
Yokozaki
H
, et al
Transfection of interleukin-8 increases angiogenesis and tumorigenesis of human gastric carcinoma cells in nude mice
.
Br J Cancer
1999
;
81
:
647
53
.
40.
Inoue
K
,
Slaton
JW
,
Eve
BY
,
Kim
SJ
,
Perrotte
P
,
Balbay
MD
, et al
Interleukin 8 expression regulates tumorigenicity and metastases in androgen-independent prostate cancer
.
Clin Cancer Res
2000
;
6
:
2104
19
.
41.
Sparmann
A
,
Bar-Sagi
D
. 
Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis
.
Cancer Cell
2004
;
6
:
447
58
.
42.
Iguchi
A
,
Kitajima
I
,
Yamakuchi
M
,
Ueno
S
,
Aikou
T
,
Kubo
T
, et al
PEA3 and AP-1 are required for constitutive IL-8 gene expression in hepatoma cells
.
Biochem Biophys Res Commun
2000
;
279
:
166
71
.
43.
Hacker
H
,
Karin
M
. 
Regulation and function of IKK and IKK-related kinases
.
Sci STKE
2006
;
357
:
re13
.
44.
Li
ZW
,
Chu
W
,
Hu
Y
,
Delhase
M
,
Deerinck
T
,
Ellisman
M
, et al
The IKKbeta subunit of IkappaB kinase (IKK) is essential for nuclear factor kappaB activation and prevention of apoptosis
.
J Exp Med
1999
;
189
:
1839
45
.
45.
Shukla
S
,
MacLennan
GT
,
Fu
P
,
Patel
J
,
Marengo
SR
,
Resnick
MI
, et al
Nuclear factor-kappaB/p65 (Rel A) is constitutively activated in human prostate adenocarcinoma and correlates with disease progression
.
Neoplasia
2004
;
6
:
390
400
.
46.
Li
W
,
Tan
D
,
Zenali
MJ
,
Brown
RE
. 
Constitutive activation of nuclear factor-kappa B (NF-kB) signaling pathway in fibrolamellar hepatocellular carcinoma
.
Int J Clin Exp Pathol
2010
;
3
:
238
43
.
47.
Xie
TX
,
Xia
Z
,
Zhang
N
,
Gong
W
,
Huang
S
. 
Constitutive NF-kappaB activity regulates the expression of VEGF and IL-8 and tumor angiogenesis of human glioblastoma
.
Oncol Rep
2010
;
23
:
725
32
.
48.
Arsura
M
,
Mercurio
F
,
Oliver
AL
,
Thorgeirsson
SS
,
Sonenshein
GE
. 
Role of the IkappaB kinase complex in oncogenic Ras- and Raf-mediated transformation of rat liver epithelial cells
.
Mol Cell Biol
2000
;
20
:
5381
91
.
49.
Factor
V
,
Oliver
AL
,
Panta
GR
,
Thorgeirsson
SS
,
Sonenshein
GE
,
Arsura
M
. 
Roles of Akt/PKB and IKK complex in constitutive induction of NF-kappaB in hepatocellular carcinomas of transforming growth factor alpha/c-myc transgenic mice
.
Hepatology
2001
;
34
:
32
41
.