T-lymphokine–activated killer cell–originated protein kinase (TOPK) is overexpressed in highly proliferating tumors such as leukemias and myelomas, and seems to play a key role in tumorigenesis or metastasis. However, the precise role and regulatory mechanism explaining the effects of TOPK on tumor cells still remain elusive. Here, we reported that TOPK regulates UVB-induced c-Jun-NH2-kinase 1 (JNK1) activity, and is essential for H-Ras–induced activator protein-1 activity and cell transformation. We showed that TOPK associated with and phosphorylated JNK1 following UVB irradiation in vitro or in vivo. Moreover, UVB-induced JNK1 activity was greatly augmented in mouse epidermal JB6 Cl41 cells that stably expressed TOPK cDNA. On the other hand, JNK1 activity was markedly attenuated by stable expression of small interfering RNA against TOPK in malignant melanoma RPMI 7951 cells. Interestingly, TOPK interacted with JNK-interacting protein 1 and caused an elevation of JNK-interacting protein 1 scaffolding activity, thereby enhancing JNK1 activity. Furthermore, JNK1 was required for TOPK-mediated activator protein-1 transcriptional activity and transformed foci induced by UVB or H-Ras. Taken together, these findings showed that TOPK positively modulated UVB-induced JNK1 activity and played a pivotal role in JNK1-mediated cell transformation induced by H-Ras. These studies might also provide a novel molecular mechanism for the role of TOPK in UVB-mediated skin carcinogenesis. [Cancer Res 2007;67(11):5186–94]

Mitogen-activated protein kinases (MAPK), including extracellular signal–regulated kinases, c-Jun-NH2-kinases (JNK), and p38 MAPKs, function as critical regulatory effectors in many cellular processes, including cell proliferation, migration, survival, and death (14). Particularly, JNKs, also known as stress-activated MAPKs, contain the dual phosphorylation motif Thr-Pro-Tyr (5), and have been shown to bind and phosphorylate c-Jun on Ser63 and Ser73 in response to UV irradiation (6, 7). JNKs are activated by many diverse stimuli such as proinflammatory cytokines [e.g., tumor necrosis factor (TNF) and interleukin 1], UV irradiation, and tumor promoters (813). Activated JNKs can phosphorylate numerous substrates, including several transcription factors such as activator protein-1 (AP-1), thereby affecting gene expression and subsequent physiologic cellular functions (14). The JNK signaling pathway consists of JNKs/stress-activated MAPKs, MAP2Ks (e.g., MKK4 or MKK7), and MAP3Ks, which include the mixed lineage kinase (MLK) 3 or MAPK kinase kinase 1. Some of these kinases form signaling complexes mediated by scaffold proteins, such as the JNK-interacting protein 1 (JIP1; refs. 1517). Various scaffold proteins, including JIP1, were suggested to form complexes with MAPK signaling components, thereby creating a functional signaling module (18). JIP1 physically associates with JNKs, MKK7, and members of the MLK group of MAP3Ks functioning to stimulate JNK activity (16).

T-lymphokine–activated killer cell–originated protein kinase (TOPK), a MAPKK-like protein kinase, is a serine/threonine kinase that is highly expressed in many tumors such as leukemias, lymphomas, and myelomas (1921). TOPK is also up-regulated in testicular tissue and is believed to participate in spermatogenesis (22). Structurally, TOPK comprises kinase subdomains, the conserved dual specificity active site sequence (D-X-K-X-X-N) spanning amino acids 174 to 179, and a COOH-terminal ETDV motif that binds PDZ domains, which might associate with the tumor suppressor protein hDlg (20). TOPK is known to be phosphorylated during mitosis and is a substrate of Cdc2/cyclin B (20, 23). Furthermore, p38 MAPK has also been reported to be a substrate of TOPK (19). More recently, TOPK was shown to phosphorylate histone H3 at Ser10in vitro and in vivo, and TOPK kinase activity seems to be involved in its oncogenic function (24). However, the detailed signaling pathways and biological functions associated with TOPK, particularly in tumor cells, still remain to be determined.

Here, we identified TOPK as a novel upstream activator of JNKs. We showed that TOPK regulates JNK activity in response to UVB irradiation and further showed that TOPK has a major role in H-ras oncogene–induced cell transformation. We also provided evidence showing that TOPK could associate with and phosphorylate JNK1 and that TOPK regulation of JNK1 activity enhanced the ability of JNKs to mediate H-Ras–induced cell transformation.

Cells and reagents. Mouse epidermal skin (JB6 Cl41), human epithelial kidney (HEK293), human malignant melanoma (RPMI 7951), and mouse embryo fibroblasts (3T3) were purchased from American Type Culture Collection. JNK wild-type murine embryonic fibroblasts (MEF), JNK1 knockout (JNK1−/−) MEFs, and JNK2 knockout (JNK2−/−) MEFs were kind gifts from Dr. Roger J. Davis (University of Massachusetts Medical School, Worcester, MA). JB6 Cl41 cells and RPMI 7951 cells stably transfected with TOPK cDNA and TOPK small interfering RNA (siRNA), respectively, were selected with G418, identified, and established as described (12). Cells were cultured in DMEM or MEM supplemented with 5 or 10% fetal bovine serum, 2 mmol/L of l-glutamine, and 1% penicillin/streptomycin. Transfection of cells was done using jetPEI (Qbiogene) according to the instructions of the manufacturer. 12-O-Tetradecanoylphorbol-13-acetate (TPA) or TNFα was purchased from Sigma and the DNA ligation kit and glutathione-Sepharose 4B were from Takara Bio, Inc. and Amersham Biosciences, respectively. Antibodies against HA, V5, Myc, or FLAG epitopes were purchased from Santa Cruz Biotechnology, Inc., Invitrogen, BD Biosciences, Inc., or Sigma, respectively. Antibodies against c-Jun, phospho-c-Jun (Ser63), JNK, phospho-JNKs (Thr183/Tyr185), and TOPK were from Cell Signaling Technology, Inc. Texas red– or FITC-conjugated IgG was purchased from Invitrogen and active TOPK or MKK7β1 was from Invitrogen or Upstate, respectively.

Plasmids. The plasmids for the expression of pcDNA3.1/V5-tagged JNK1, JNK2, or JNK3 and deletion mutants of V5-tagged JNK1or JNK3 were described previously (10). The plasmid pcDNA3/HA-TOPK encoding TOPK cDNA was a gift from Dr. Yasuhito Abe (Ehime University School of Medicine, Ehime, Japan). The glutathione S-transferase (GST)-TOPK construct was generated by cloning the PCR product into the BamHI/EcoRI sites of pGEX-5X1 (Amersham) and the cDNA insert was verified by sequencing (Genewiz, Inc.). Expression vectors for MKK7, JIP1, and MLK3 were kindly provided by Dr. Roger J. Davis, Dr. Lawrence B. Holzman (University of Michigan Medical School, Ann Arbor, MI), and Dr. James R. Woodgett (Ontario Cancer Institute, Toronto, Canada), respectively. Expression vectors encoding GST-c-Jun (1–79) and c-Jun dominant-negative TAM-67 were gifts from Dr. Lynn E. Heasley (University of Colorado Health Sciences Center, Denver, CO) and Dr. Michael J. Birrer (National Cancer Institute, Bethesda, MD), respectively.

In vitro GST pull down. The plasmids for expression of pcDNA3.1/V5-JNK1, 2, 3, or deletion mutants of JNK1 or JNK3 were translated in vitro with L-[35S]methionine using the TNT Quick coupled transcription/translation system (Promega). Briefly, GST-TOPK proteins immobilized on glutathione-Sepharose beads (Amersham Biosciences) were incubated at 4°C for 3 h with 300 μg of each cell lysate or 35S-labeled translated JNK1, JNK2, JNK3, or deletion mutants. The bound proteins were washed extensively, boiled in sample buffer, and separated by SDS-PAGE and subsequently analyzed by immunoblotting with appropriate antibodies or by autoradiography.

Kinase assay. The in vitro kinase assays and GST-c-Jun pull-down kinase assays were done as described previously (12). Briefly, active TOPK or MKK7 was incubated at 30°C for 30 min with 1 μg of GST-JNK1 and 10 μCi of γ-32p(ATP). The reaction mixture was separated with SDS-PAGE and exposed to X-ray film for autoradiography. The JNK kinase assays were done using c-Jun immobilized on glutathione-Sepharose beads. Each 300 μg of cell lysate was incubated at 4°C for 2 h with 20 μL of GST-c-Jun beads, washed extensively, and subjected to immunoblotting with phospho-c-Jun (Ser63). For the TOPK kinase assay, HEK293 cells were transfected with control vector or pcDNA3/HA-TOPK. At 48 h after transfection, cells were irradiated with UVB (4 kJ/m2), and cell lysates were subjected to immunoprecipitation using protein G Sepharose beads with anti-HA followed by a kinase assay with GST-JNK1 as substrate.

Immunoblot analysis and immunoprecipitation. Immunoblotting or immunoprecipitation was done as described (25). To check the level of phosphorylation of JNKs in TOPK-expressing cells, TOPK siRNA cells, or their respective control cells after exposure to UVB (4 kJ/m2), immunoblotting was carried out using a phospho-JNK antibody recognizing Thr183/Tyr185 residues (Cell Signaling Technology).

Immunofluorescence assay. To determine the localization of endogenous JNK or TOPK in vivo, 70% confluent RPMI 7951 cells were starved in serum-free medium for 24 h, and then were or were not irradiated with UVB (4 kJ/m2). An immunofluorescence assay was done as described (12). Briefly, cells were fixed in 4% paraformaldehyde and incubated with anti–phospho-JNK and FITC-conjugated IgG or anti-TOPK and Texas red–conjugated IgG. The phosphorylated JNK or TOPK was stained for green or red fluorescence, respectively. Nuclei were stained with Hoechst and samples were analyzed with a fluorescence microscope system (Leica).

Reporter gene assays. For reporter gene assays, transient transfections were carried out using jetPEI (Qbiogene) and different combinations of each expression construct together with the AP-1 reporter gene. Cotransfection with 0.5 μg of the pRL-SV40 gene (Promega) for each transfection was done and Renilla luciferase activity was normalized for transfection efficiency. At 48 h after transfection, cells were harvested and luciferase activity was assessed using the Luminoskan Ascent (Thermo Electron, Corp.).

Focus-forming assay. A transformation assay of 3T3 Swiss cells was carried out according to standard protocols (26). Briefly, 3T3 Swiss cells were plated in six-well plates at a density of 2 × 105 cells 24 h before transfection. Cells were transiently transfected with various combinations of plasmids, H-RasG12V (100 ng), pcDNA3.1/V5-tagged JNK1 (1 μg), pcDNA3/HA-TOPK (1 μg), or JNK1 siRNA (1 μg). At 48 h posttransfection, cells were transferred to 100 mm plates, and then cultured in DMEM with 5% bovine calf serum for 2 weeks. Foci were fixed with methanol, stained with 0.4% crystal violet, and then counted under a microscope.

TOPK associates with and phosphorylates JNK1. Signal transduction cascades related to TOPK still remain undiscovered. Here, we found that TOPK associated with JNK. To confirm that JNK directly interacted with TOPK, we cotransfected HEK293 cells with HA-TOPK and V5-JNK1, V5-JNK2, or V5-JNK3 constructs and then immunoprecipitation experiments were done. Results indicated that ectopically expressed JNK1 or JNK3, but not JNK2, bound to TOPK (Fig. 1A), indicating that JNK1 or JNK3 could associate with TOPK in vivo. We also carried out GST pull-down assays to further confirm the interaction of JNK and TOPK in vitro. GST or GST-TOPK, coupled to glutathione-Sepharose beads, was incubated with 35S-methionine–labeled JNKs using an in vitro transcription and translation system (Promega). Consistent with the immunoprecipitation results, the GST pull-down assay showed that JNK1 or JNK3 bound to TOPK in vitro, supporting a direct interaction between TOPK and JNK (Fig. 1B,, top). To identify the domains that are required for the association of the two kinases, a GST pull-down assay was done, using constructs of several deletion mutants of JNK1 or JNK3. Results revealed that fragments containing the glycine-rich region of JNK1 or JNK3 bound to TOPK, implying that this portion of the NH2-terminal region is important for binding of JNK1 (Fig. 1B,, middle) or JNK3 (Fig. 1B,, bottom) to TOPK. We next investigated whether the MAPKK-like TOPK could phosphorylate JNK. A kinase assay using GST-JNK1 revealed that active TOPK phosphorylated JNK1 in vitro (Fig. 1C,, top, lane 4). Active MKK7 phosphorylation of JNK1 was used as a positive control (Fig. 1C,, top, lane 5). To further verify that this TOPK-mediated JNK1 phosphorylation occurred in vivo, we examined the possibility that UVB irradiation (4 kJ/m2) elicits phosphorylation of TOPK and subsequent JNK1 phosphorylation. Results revealed that UVB irradiation induced the phosphorylation of serine and threonine (Ser/Thr) residues on TOPK (Fig. 1C,, middle), and that activated TOPK phosphorylated JNK1 (Fig. 1C,, bottom). We then examined the effect of the TOPK-JNK1 association on JNK1 activity using GST-c-Jun as substrate and found that JNK1 activity was enhanced by the presence of the recombinant His-tagged TOPK protein (data not shown). These results suggested that the TOPK interaction with JNK1 might stimulate JNK1 activity to phosphorylate c-Jun. To next determine the cellular location of phosphorylated JNK1 and TOPK, immunofluorescence staining was done and results showed that following UVB, phosphorylated JNK1 was translocated into the nucleus where it colocalized with TOPK (Fig. 1D). Collectively, these data indicated that TOPK associates with and phosphorylates JNK1 in vitro and in vivo.

Figure 1.

TOPK directly associates with JNKs and phosphorylates JNK1. A, HEK293 cells were transfected with 1 μg of pcDNA3/HA-TOPK and pcDNA3/V5–tagged JNK1, JNK2, or JNK3, respectively. At 48 h after transfection, individual cell lysates (500 μg) were used for immunoprecipitation (IP) with anti-HA and immunoprecipitates were subjected to immunoblotting (IB) with anti-V5 or anti-HA. Cell lysates were also subjected to immunoblotting with anti-V5 (bottom) to confirm the expression levels. B, GST or GST-TOPK proteins (2 μg) immobilized on glutathione-Sepharose beads were incubated with in vitro–translated 35S-methionine–labeled JNK1, JNK2, or JNK3 (top). After washing, the bound proteins were subjected to 10% SDS-PAGE and analyzed by autoradiography. Top right, 2% input of translated JNK1, JNK2, or JNK3. GST-TOPK proteins on beads (2 μg) were incubated with 35S-methionine–labeled deletion mutants of JNK1 (middle) or JNK3 (bottom). C, an in vitro kinase assay (top) was done with 100 ng each of active TOPK (Invitrogen) or MKK7β1 (Upstate), or 200 ng of purified GST-TOPK and substrate, GST-JNK1 (1 μg), and followed by SDS-PAGE and detection by autoradiography. Serum-starved RPMI 7951 cells were treated or not treated with UVB (4 kJ/m2) and harvested after 1 h, and then subjected to immunoprecipitation (IP) with anti-TOPK and subsequent immunoblotting with anti–phospho-Ser/Thr or with anti-TOPK (middle). To do the immunocomplex kinase assay, RPMI 7951 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK and irradiated with UVB (4 kJ/m2) and harvested after 1 h. Cell lysates were subjected to immunoprecipitation and subsequent kinase assay (bottom). D, serum-starved RPMI 7951 cells were or were not irradiated with UVB (4 kJ/m2), fixed, and then stained for phosphorylated JNK (green) or TOPK (red). The images were observed by immunofluorescence microscopy. Localization of phosphorylated JNK1 and TOPK or merged images are shown with (bottom) or without (top) UVB. The nuclei were stained with Hoechst and representative photos of at least three independent experiments are shown. Scale bar, 10 μm.

Figure 1.

TOPK directly associates with JNKs and phosphorylates JNK1. A, HEK293 cells were transfected with 1 μg of pcDNA3/HA-TOPK and pcDNA3/V5–tagged JNK1, JNK2, or JNK3, respectively. At 48 h after transfection, individual cell lysates (500 μg) were used for immunoprecipitation (IP) with anti-HA and immunoprecipitates were subjected to immunoblotting (IB) with anti-V5 or anti-HA. Cell lysates were also subjected to immunoblotting with anti-V5 (bottom) to confirm the expression levels. B, GST or GST-TOPK proteins (2 μg) immobilized on glutathione-Sepharose beads were incubated with in vitro–translated 35S-methionine–labeled JNK1, JNK2, or JNK3 (top). After washing, the bound proteins were subjected to 10% SDS-PAGE and analyzed by autoradiography. Top right, 2% input of translated JNK1, JNK2, or JNK3. GST-TOPK proteins on beads (2 μg) were incubated with 35S-methionine–labeled deletion mutants of JNK1 (middle) or JNK3 (bottom). C, an in vitro kinase assay (top) was done with 100 ng each of active TOPK (Invitrogen) or MKK7β1 (Upstate), or 200 ng of purified GST-TOPK and substrate, GST-JNK1 (1 μg), and followed by SDS-PAGE and detection by autoradiography. Serum-starved RPMI 7951 cells were treated or not treated with UVB (4 kJ/m2) and harvested after 1 h, and then subjected to immunoprecipitation (IP) with anti-TOPK and subsequent immunoblotting with anti–phospho-Ser/Thr or with anti-TOPK (middle). To do the immunocomplex kinase assay, RPMI 7951 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK and irradiated with UVB (4 kJ/m2) and harvested after 1 h. Cell lysates were subjected to immunoprecipitation and subsequent kinase assay (bottom). D, serum-starved RPMI 7951 cells were or were not irradiated with UVB (4 kJ/m2), fixed, and then stained for phosphorylated JNK (green) or TOPK (red). The images were observed by immunofluorescence microscopy. Localization of phosphorylated JNK1 and TOPK or merged images are shown with (bottom) or without (top) UVB. The nuclei were stained with Hoechst and representative photos of at least three independent experiments are shown. Scale bar, 10 μm.

Close modal

TOPK participates in UVB-induced JNK activation. We next investigated whether TOPK was involved in TPA-, UV- or TNFα-induced JNK activation. For this purpose, HEK293 cells were transiently transfected with HA-TOPK, and then were treated with TPA, UVB, or TNFα. Results of GST-c-Jun pull-down kinase assays showed that TOPK expression resulted in a strong increase in JNK activity to phosphorylate c-Jun in response to UVB irradiation (4 kJ/m2; Fig. 2A). Also, TNFα treatment of cells transfected with TOPK exhibited an increase in JNK activity. However, TOPK expression did not affect TPA-induced JNK activity, indicating that TOPK might contribute specifically to JNK activation signaling pathways depending on the stimulus. We also confirmed that TOPK expression enhanced UVB-induced JNK activity in JB6 Cl41 cells (Fig. 2B). In another experiment, epidermal JB6 Cl41 cells, which have almost no TOPK endogenous expression, and malignant melanoma RPMI 7951 cells, which express high levels of TOPK, were stably transfected with TOPK cDNA or TOPK siRNA, respectively. The results of a kinase assay revealed that the TOPK-expressing JB6 cells exhibited dramatically increased UVB-induced JNK activity (Fig. 2C), whereas knockdown of TOPK by siRNA in RPMI cells resulted in substantially decreased activity (Fig. 2D). These data suggested that TOPK contributes to JNK activation and phosphorylation of c-Jun in response to UVB.

Figure 2.

TOPK contributes to the UVB-induced JNK activation pathway. A, HEK293 cells were transfected with 1 μg of control vector pcDNA3 or pcDNA3/HA-TOPK. Then, 48 h after transfection, cells were treated with UVB (4 kJ/m2), TPA (20 ng/mL), or TNFα (40 ng/mL) and harvested after 1 h (UVB, TPA) or 20 min (TNFα). Top, cell extracts (300 μg) were subjected to immunoprecipitation using GST-c-Jun (1–79) immobilized on glutathione-Sepharose for a GST-c-Jun pull-down kinase assay and subsequent immunoblotting with anti–phospho-c-Jun (Ser63). Bottom, the level of GST-c-Jun fusion proteins. B, JB6 Cl41 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK. At 48 h posttransfection, cells were irradiated with UVB (4 kJ/m2) and harvested after 1 h and a GST-c-Jun pull-down kinase assay was done using each cell lysate as in (A). C, JB6 Cl41 cells stably expressing TOPK or control vector were exposed to UVB (4 kJ/m2) and then cell lysates were used for the GST-c-Jun pull-down kinase assay. Bottom, expression level of TOPK. D, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were irradiated with UVB (4 kJ/m2) and then the cell lysates were used for the GST-c-Jun pull-down kinase assay. Bottom, effectiveness of TOPK siRNA in suppressing TOPK expression. Blots shown are representative of at least three independent experiments.

Figure 2.

TOPK contributes to the UVB-induced JNK activation pathway. A, HEK293 cells were transfected with 1 μg of control vector pcDNA3 or pcDNA3/HA-TOPK. Then, 48 h after transfection, cells were treated with UVB (4 kJ/m2), TPA (20 ng/mL), or TNFα (40 ng/mL) and harvested after 1 h (UVB, TPA) or 20 min (TNFα). Top, cell extracts (300 μg) were subjected to immunoprecipitation using GST-c-Jun (1–79) immobilized on glutathione-Sepharose for a GST-c-Jun pull-down kinase assay and subsequent immunoblotting with anti–phospho-c-Jun (Ser63). Bottom, the level of GST-c-Jun fusion proteins. B, JB6 Cl41 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK. At 48 h posttransfection, cells were irradiated with UVB (4 kJ/m2) and harvested after 1 h and a GST-c-Jun pull-down kinase assay was done using each cell lysate as in (A). C, JB6 Cl41 cells stably expressing TOPK or control vector were exposed to UVB (4 kJ/m2) and then cell lysates were used for the GST-c-Jun pull-down kinase assay. Bottom, expression level of TOPK. D, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were irradiated with UVB (4 kJ/m2) and then the cell lysates were used for the GST-c-Jun pull-down kinase assay. Bottom, effectiveness of TOPK siRNA in suppressing TOPK expression. Blots shown are representative of at least three independent experiments.

Close modal

TOPK-mediated JNK1 activation induced by UVB requires phosphorylation of Thr183/Tyr185 on JNK1. We next determined whether TOPK regulated JNK activity through its phosphorylation of the Thr183/Tyr185 residues of JNK, a posttranslational modification that is required for JNK activation in response to UVB irradiation. Immunoblot analysis revealed that UVB-induced phosphorylation of Thr183/Tyr185 was increased by ectopic expression of TOPK (Fig. 3A). In addition, JNK phosphorylation was also increased in cells stably expressing TOPK compared with cells expressing empty vector (Fig. 3B), whereas the phosphorylation was markedly impaired in TOPK knockdown cells (Fig. 3C). Interestingly, phosphorylation of c-Jun induced by UVB irradiation was augmented in TOPK-expressing cells (Fig. 3B), suggesting that TOPK-mediated UVB-induced JNK activation culminates in the phosphorylation of c-Jun. Taken together, these data indicated that TOPK plays a key role in UVB-induced JNK activation and phosphorylation, and c-Jun phosphorylation.

Figure 3.

TOPK mediates UVB-induced JNK activation through phosphorylation of JNK1/2. A, HEK293 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK. At 48 h after transfection, cells were treated or not treated with UVB (4 kJ/m2) and harvested 1 h later. Immunoblotting with anti–phospho-JNKs (Cell Signaling) was done using each cell lysate. Expression of total JNK proteins was determined (bottom). B, JB6 Cl41 cells stably expressing TOPK or control vector were exposed to UVB (4 kJ/m2), harvested 1 h later, and then subjected to immunoblotting with anti–phospho-JNK or anti–phospho-c-Jun. Expression of total JNKs and c-Jun proteins was also determined. C, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were irradiated with UVB (4 kJ/m2), harvested after 1 h, and then the cell lysates were used for immunoblotting with anti–phospho-JNKs. Bottom, expression of total endogenous JNKs. Blots shown are representative of at least three independent experiments.

Figure 3.

TOPK mediates UVB-induced JNK activation through phosphorylation of JNK1/2. A, HEK293 cells were transfected with 1 μg of control vector or pcDNA3/HA-TOPK. At 48 h after transfection, cells were treated or not treated with UVB (4 kJ/m2) and harvested 1 h later. Immunoblotting with anti–phospho-JNKs (Cell Signaling) was done using each cell lysate. Expression of total JNK proteins was determined (bottom). B, JB6 Cl41 cells stably expressing TOPK or control vector were exposed to UVB (4 kJ/m2), harvested 1 h later, and then subjected to immunoblotting with anti–phospho-JNK or anti–phospho-c-Jun. Expression of total JNKs and c-Jun proteins was also determined. C, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were irradiated with UVB (4 kJ/m2), harvested after 1 h, and then the cell lysates were used for immunoblotting with anti–phospho-JNKs. Bottom, expression of total endogenous JNKs. Blots shown are representative of at least three independent experiments.

Close modal

TOPK associates with JIP1 and modulates JIP1 scaffolding activity. JIP1 was shown to be closely involved in the regulation of JNK and p38 kinase activities, and especially JNKs (27). We thus investigated whether TOPK regulation of JNKs might also affect JIP1 scaffolding activity. HEK293 cells were transfected with HA-TOPK and Myc-JIP1 constructs and an immunoprecipitation experiment was done. Results revealed that TOPK associated with JIP1 in vivo (Fig. 4A). To further confirm the interaction of TOPK and JIP1, total lysates from cells transfected with Myc-JIP1 were incubated with GST or GST-TOPK. Data showed that TOPK directly interacted with JIP1 in vitro, indicating that a TOPK association with JIP1 could be implicated in JIP1 function (Fig. 4B). To assess whether TOPK regulates JIP1-mediated JNK activity, we next examined the effect of TOPK on JIP1 scaffolding activity. JIP1 has been shown to physically interact with JNKs, MKK7, and members of the MLK group of MAP3Ks, resulting in an enhancement of the JNK signaling pathway (17, 27). As expected, coexpression of JIP1 together with MLK3, MKK7, and JNK1 increased JIP1-mediated JNK1 activity (Fig. 4C). However, JIP1 scaffolding activity was also substantially elevated with the coexpression of TOPK together with JIP1, MLK3, MKK7, and JNK1. Furthermore, immunoblot analysis using anti–phospho-JNK1/2 revealed that coexpression of TOPK together with MLK3, MKK7, JNK1, and JIP1 strongly increased JNK phosphorylation (Fig. 4D), implying that the interaction between TOPK and MLK3 or MKK7 might be critical for JIP1 scaffolding activity. These data suggested that the interaction of TOPK with JIP1 might play a key role in the regulation of JNK activity.

Figure 4.

TOPK interacts with JIP1 and enhances JIP1 scaffolding activity. A, HEK293 cells were transfected with the indicated combinations of plasmids Myc-JIP1 (1 μg) or HA-TOPK (1 μg). At 48 h posttransfection, cell lysates (500 μg) were subjected to immunoprecipitation (IP) with anti-HA and subsequent immunoblotting (IB) with anti-Myc (top) or anti-HA (bottom). B, GST or GST-TOPK beads were incubated with 300 μg of total lysates of cells transfected with Myc-JIP1 (1 μg), and followed by immunoblotting with anti–c-Myc. The input indicates the amount of cell lysate loaded. C, HEK293 cells were transfected with various combinations of 0.5 μg of expression vectors for FLAG-MLK3, FLAG-MKK7, V5-JNK1, Myc-JIP1, and HA-TOPK. Cell lysates (300 μg) were then subjected to the GST-c-Jun pull-down kinase assay. Bottom, the level of GST-c-Jun. D, HEK293 cells were transfected with the indicated combinations of 0.5 μg of plasmids FLAG-MLK3, FLAG-MKK7, V5-JNK1, Myc-JIP1, and HA-TOPK. Cell lysates (20 μg) were then subjected to immunoblotting with anti–phospho-JNKs.

Figure 4.

TOPK interacts with JIP1 and enhances JIP1 scaffolding activity. A, HEK293 cells were transfected with the indicated combinations of plasmids Myc-JIP1 (1 μg) or HA-TOPK (1 μg). At 48 h posttransfection, cell lysates (500 μg) were subjected to immunoprecipitation (IP) with anti-HA and subsequent immunoblotting (IB) with anti-Myc (top) or anti-HA (bottom). B, GST or GST-TOPK beads were incubated with 300 μg of total lysates of cells transfected with Myc-JIP1 (1 μg), and followed by immunoblotting with anti–c-Myc. The input indicates the amount of cell lysate loaded. C, HEK293 cells were transfected with various combinations of 0.5 μg of expression vectors for FLAG-MLK3, FLAG-MKK7, V5-JNK1, Myc-JIP1, and HA-TOPK. Cell lysates (300 μg) were then subjected to the GST-c-Jun pull-down kinase assay. Bottom, the level of GST-c-Jun. D, HEK293 cells were transfected with the indicated combinations of 0.5 μg of plasmids FLAG-MLK3, FLAG-MKK7, V5-JNK1, Myc-JIP1, and HA-TOPK. Cell lysates (20 μg) were then subjected to immunoblotting with anti–phospho-JNKs.

Close modal

UVB-induced AP-1 activity is mediated by the TOPK-JNK1-c-Jun signaling cascade. UV is suggested to induce JNK activation that results in the phosphorylation of c-Jun, which plays a pivotal role in UV-induced AP-1 activation (28). To explore the role of TOPK in UVB-induced AP-1 activity, we next assessed the UVB-induced AP-1 promoter–driven luciferase activity in JB6 Cl41 cells expressing TOPK or RPMI 7951 cells expressing TOPK siRNA. The UVB-induced AP-1 transcriptional activity was increased by ∼2-fold in cells expressing TOPK compared with the level of increase of control cells expressing an empty vector (Fig. 5A), whereas the activity was diminished in TOPK siRNA cells compared with control siRNA cells (Fig. 5B). We next determined the effect of the TOPK-JNK1 association on UVB-induced AP-1 activity. Coexpression of TOPK and JNK1 increased the UVB-induced JNK1-c-Jun–mediated transcriptional activity of AP-1. In contrast, expression of a dominant-negative mutant of c-Jun, TAM-67, strongly attenuated AP-1 activity (Fig. 5C). These results indicated that TOPK is implicated in the UVB-induced JNK1-c-Jun–dependent signaling pathway leading to AP-1 activation.

Figure 5.

The TOPK-JNK1-c-Jun signaling pathway mediates UVB-induced AP-1 activity. A, JB6 Cl41 cells stably transfected with HA-TOPK or control vector were transfected with the AP-1–driven luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg). At 24 h after transfection, cells were serum-starved for another 24 h, and then were or were not irradiated with UVB (4 kJ/m2) and harvested after 6 h. The firefly luciferase activity was normalized against Renilla luciferase activity. B, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were transfected as in (A). At 24 h posttransfection, cells were treated or not treated with UVB and luciferase activity was measured as in (A). C, JB6 Cl41 cells were transfected with the indicated combinations of 1 μg of plasmids HA-TOPK, V5-JNK1, c-Jun, or c-Jun dominant-negative (TAM-67) together with 1 μg of the AP-1-luciferase reporter gene and 0.1 μg of the pRL-SV40 gene. At 24 h posttransfection, cell lysates were assessed for luciferase activity. Columns, means of three independent experiments done in duplicate; bars, SE (A–C). Significant differences were evaluated using the Student's t test (*, P < 0.01 and **, P < 0.05).

Figure 5.

The TOPK-JNK1-c-Jun signaling pathway mediates UVB-induced AP-1 activity. A, JB6 Cl41 cells stably transfected with HA-TOPK or control vector were transfected with the AP-1–driven luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg). At 24 h after transfection, cells were serum-starved for another 24 h, and then were or were not irradiated with UVB (4 kJ/m2) and harvested after 6 h. The firefly luciferase activity was normalized against Renilla luciferase activity. B, RPMI 7951 cells stably transfected with TOPK siRNA or control siRNA were transfected as in (A). At 24 h posttransfection, cells were treated or not treated with UVB and luciferase activity was measured as in (A). C, JB6 Cl41 cells were transfected with the indicated combinations of 1 μg of plasmids HA-TOPK, V5-JNK1, c-Jun, or c-Jun dominant-negative (TAM-67) together with 1 μg of the AP-1-luciferase reporter gene and 0.1 μg of the pRL-SV40 gene. At 24 h posttransfection, cell lysates were assessed for luciferase activity. Columns, means of three independent experiments done in duplicate; bars, SE (A–C). Significant differences were evaluated using the Student's t test (*, P < 0.01 and **, P < 0.05).

Close modal

JNK is required for TOPK-mediated H-Ras–induced cell transformation. The oncogenic H-ras is known to enhance the ability of JNKs to phosphorylate c-Jun and to influence cell transformation and tumorigenesis (29, 30). We therefore investigated whether TOPK was involved in JNK1-mediated AP-1 activity induced by H-Ras. AP-1 transcriptional activities were evaluated in several cell lines. As expected, expression of H-RasG12V increased AP-1 activity, and coexpression of JNK1 or TOPK together with H-RasG12V resulted in a greater increase in AP-1 activity (Fig. 6A). Moreover, AP-1 transcriptional activity was even more augmented by coexpression of H-RasG12V, JNK1, and TOPK, suggesting that TOPK and JNK1 function as mediators in H-RasG12V signaling leading to AP-1 activation. The role of TOPKs' association with JNK1 in AP-1 activation was next examined in TOPK-expressing cells or JNK1 knockout (JNK1−/−) MEFs. Results indicated that H-RasG12V–induced AP-1 activity was impaired in JNK1−/− MEFs compared with wild-type MEFs (Fig. 6B). More importantly, coexpression of H-RasG12V and TOPK did not greatly augment AP-1 activity in JNK1−/− MEFs compared with wild-type MEFs. In contrast, AP-1 activity in JNK2−/− MEFs was similarly increased by the expression of H-RasG12V and TOPK compared with wild-type MEFs, indicating that JNK1, but not JNK2, mediated the H-RasG12V-TOPK signaling pathway leading to AP-1. Similarly, H-RasG12V–induced AP-1 activity was greatly increased in TOPK-expressing cells, compared with control cells (Fig. 6C). But again, coexpression of H-RasG12V and JNK1 caused an even greater augmentation in the activity, indicating that TOPK also mediated the H-RasG12V signaling pathway associated with AP-1 activation. Taken together, these results suggested that TOPK and JNK1 were critical effectors for H-RasG12V signaling to AP-1. Oncogenic H-ras has been shown to strongly augment JNK activity and c-Jun phosphorylation, suggesting that JNKs act as important mediators of tumorigenesis (10). We hypothesized that regulation of JNK activity by TOPK might affect the ability of JNKs to mediate H-Ras–induced cell transformation. Thus, we transfected 3T3 cells with H-RasG12V and/or various combinations of TOPK and JNK1, and then subjected these cells to a focus-forming assay. Results indicated that transformed foci were increased in cells transfected with H-RasG12V compared with mock-transfected cells (Fig. 6D,, top, plate 1 versus 2). As expected, coexpression of JNK1 together with H-RasG12V caused a further increase in foci formation (Fig. 6D,, top, plate 2 versus 3). Interestingly, the number of foci was substantially augmented by coexpression of TOPK and H-RasG12V (Fig. 6D,, top, plate 2 versus 4); and furthermore, coexpression of TOPK and JNK1 with H-RasG12V elicited even more foci (Fig. 6D,, middle, plate 1 versus top, plates 2, 3, and 4). Importantly, the number of transformed foci induced by H-RasG12V and TOPK or JNK1 was substantially diminished in cells cotransfected with combinations of H-RasG12V, TOPK siRNA, or JNK1 siRNA (Fig. 6D , middle, plates 2, 3, and 4). Collectively, these results suggested that TOPK might function as a key effector in H-Ras signaling, leading to cell transformation and tumorigenesis, and that JNK1 is an important component of H-Ras-TOPK signaling to cell transformation.

Figure 6.

JNK1 is required for TOPK-mediated cell transformation induced by H-Ras. A, Swiss 3T3 cells were transfected with the indicated combinations of plasmids H-RasG12V (0.1 μg), HA-TOPK (1 μg), V5-JNK1 (1 μg), and the AP-1-luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg). At 24 h posttransfection, luciferase activities were then analyzed. B, the plasmids, H-RasG12V (0.1 μg), HA-TOPK (1 μg), and the AP-1-luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg) were expressed as indicated in wild-type MEFs, JNK1−/− MEFs, or JNK2−/− MEFs. At 24 h after transfection, cells were subjected to analysis for luciferase activities. C, JB6 Cl41 cells stably transfected with HA-TOPK (JB6-TOPK) or control vector (JB6) were transfected with the indicated combinations of plasmids, H-RasG12V (0.1 μg), V5-JNK1 (1 μg), and AP-1-luciferase reporter gene (1 μg) together with pRL-SV40 gene (0.1 μg). At 24 h after transfection, luciferase activities were measured. D, Swiss 3T3 cells cultured in six-well plates were transiently transfected with various combinations of H-RasG12V (0.1 μg), V5-JNK1 (1 μg), HA-TOPK (1 μg), or JNK1 siRNA (1 μg) as indicated. At 48 h after transfection, cells were transferred to 100-mm plates, and then cultured for an additional 2 wks with DMEM containing 5% donor bovine calf serum. Foci were analyzed by microscope and Image Plus Pro software (v. 4). The effectiveness of TOPK siRNA or JNK siRNA to inhibit TOPK or JNK expression, respectively, is shown (bottom). For all experiments, transfection of empty vector pcDNA3 served as the mock control. Columns, means of three independent experiments done in duplicate; bars, SE (A–C). Significant differences were evaluated using the Student's t test (*, P < 0.01 and **, P < 0.05).

Figure 6.

JNK1 is required for TOPK-mediated cell transformation induced by H-Ras. A, Swiss 3T3 cells were transfected with the indicated combinations of plasmids H-RasG12V (0.1 μg), HA-TOPK (1 μg), V5-JNK1 (1 μg), and the AP-1-luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg). At 24 h posttransfection, luciferase activities were then analyzed. B, the plasmids, H-RasG12V (0.1 μg), HA-TOPK (1 μg), and the AP-1-luciferase reporter gene (1 μg) together with the pRL-SV40 gene (0.1 μg) were expressed as indicated in wild-type MEFs, JNK1−/− MEFs, or JNK2−/− MEFs. At 24 h after transfection, cells were subjected to analysis for luciferase activities. C, JB6 Cl41 cells stably transfected with HA-TOPK (JB6-TOPK) or control vector (JB6) were transfected with the indicated combinations of plasmids, H-RasG12V (0.1 μg), V5-JNK1 (1 μg), and AP-1-luciferase reporter gene (1 μg) together with pRL-SV40 gene (0.1 μg). At 24 h after transfection, luciferase activities were measured. D, Swiss 3T3 cells cultured in six-well plates were transiently transfected with various combinations of H-RasG12V (0.1 μg), V5-JNK1 (1 μg), HA-TOPK (1 μg), or JNK1 siRNA (1 μg) as indicated. At 48 h after transfection, cells were transferred to 100-mm plates, and then cultured for an additional 2 wks with DMEM containing 5% donor bovine calf serum. Foci were analyzed by microscope and Image Plus Pro software (v. 4). The effectiveness of TOPK siRNA or JNK siRNA to inhibit TOPK or JNK expression, respectively, is shown (bottom). For all experiments, transfection of empty vector pcDNA3 served as the mock control. Columns, means of three independent experiments done in duplicate; bars, SE (A–C). Significant differences were evaluated using the Student's t test (*, P < 0.01 and **, P < 0.05).

Close modal

TOPK, a serine/threonine MAP2K, has been suggested to be activated in response to TPA, and Cdc2/cyclin B was reported to phosphorylate Thr9 on TOPK in vitro (19, 20). Also, p38 MAPK and c-Myc have been considered to be potential substrates of TOPK (19, 31). A possible association between Raf and TOPK was also found by yeast two-hybrid screening analysis (32). These previous studies implied that TOPK might be closely involved in the MAPK signaling pathways. However, unequivocal signal transduction cascades involving activators and downstream targets of TOPK remain elusive. On the other hand, the upstream effectors of the JNK signaling pathways have been suggested to include Rac-Cdc42, MAPK kinase kinase, and MKK4/MKK7 (33). Recently, another upstream signaling component, protein kinase C, has also been implicated in the regulation of JNKs (34, 35). Here, we reported that TOPK functions as an upstream regulator of UVB-induced JNK activation, and showed that TOPK mediates H-Ras–induced cell transformation. These data identify TOPK as a novel kinase for regulating JNK activity with a possible role in oncogenesis. Our findings also suggested that the cross-talk between TOPK and JIP1 might play a key role in regulating JNK activity.

Of interest is that TOPK specifically regulated the JNK signaling pathway leading to the phosphorylation of c-Jun at Ser63 and Ser73 in response to UVB or TNFα, but not TPA (5, 36, 37). The upstream kinases or effectors of JNK activation signaling cascades can be different based on the specific physiologic stimuli (2, 14, 28). Thus, these findings suggested that TOPK might function as a mediator of JNK1 signaling in a stimuli-dependent manner. Moreover, results of experiments with TOPK-overexpressing or knockdown cells supported the notion that TOPK might contribute to UVB-mediated JNK signaling cascades. These results implied that TOPK might be considered as a key mediator regulating downstream MAPK signaling depending on specific stimuli, especially UVB.

The JIP1 scaffolding complex is believed to consist of JNK, MKK7, and members of the MLK group of MA3PKs (16). Previous studies indicated that the JIP1 scaffolding complex enhanced JNK activation and JIP1 deficiency impaired stress-induced JNK activation in hippocampal neurons (16, 17). The results of the present study indicated that TOPK associated with JIP1 and thus seemed to enhance the ability of JIP1 scaffolding to augment JNK activity. These findings suggested that TOPK might, at least in part, contribute to JNK signaling through its interaction with JIP1. Thus, although TOPK directly interacted with, and phosphorylated JNK1, the TOPK association with the JIP1-dependent scaffolding process seemed to be required for the full activation of JNK1. Intriguingly, JIP1 scaffolding activity was enhanced when TOPK was coexpressed with MLK3 or MKK7. In addition, TOPK did not seem to compete with MLK3, MKK7, or JNK1 for JIP1 binding (data not shown), suggesting that TOPK might synergistically function with MLK3 or MKK7 as one component of the JIP1 scaffolding complex. Given the role of MLK3 or MKK7 in the activation of JNKs, these results provided important evidence for the interplay between TOPK and MLK3 or MKK7 in the JNK activation pathway. On the other hand, JNKs might be activated by alternative signaling pathways independently of JIP1 scaffolding (3840). Therefore, we cannot exclude the possibility that TOPK might be involved in JIP1 scaffold-independent signaling pathways leading to JNK activation.

Skin cancer is known to be one of the most common human cancers, and solar UV irradiation such as UVA and UVB has been suggested to function as an important carcinogen in the development of skin cancers, including nonmelanoma skin cancers, squamous cell carcinomas, and cutaneous malignant melanomas (41, 42). TOPK has been shown to be highly expressed in proliferating malignant cell lines including melanomas, and was suggested to be a molecular marker of breast cancer (21, 24). The H-ras oncogene has been proposed to strongly increase JNK activity and c-Jun phosphorylation (10, 29) and AP-1 components, particularly c-Jun, which play a key role in H-Ras–induced cell proliferation and transformation (43). These findings, showing that TOPK acts as a key effector in UVB-H-Ras-JNK signaling, might provide a mechanistic basis for a detailed role of TOPK in tumorigenesis. Notably, TOPK apparently mediated H-Ras-JNK signaling and the UVB-induced JNK activation pathway in mouse epidermal JB6 Cl41 cells or human RPMI 7951 melanoma cells, suggesting a role for TOPK in skin cancers, a finding that agrees with a previous report that TOPK might have an oncogenic effect in breast cancers (24).

Based on these observations, we propose that TOPK is a novel effector in oncogenic signaling and might be especially implicated in UVB-Ras-JNK–mediated tumorigenesis leading to skin cancers. Taken together, we conclude that the TOPK-JNK interaction is crucial for their H-Ras–mediated oncogenic potential and suggest that TOPK might be a potential molecular and therapeutic target for skin cancers.

Note: The University of Minnesota is an equal opportunity educator and employer.

Grant support: Hormel Foundation and NIH grants CA77646, CA81064, CA88961, CA27502, and CA111356.

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
Boulton TG, Nye SH, Robbins DJ, et al. ERKs: a family of protein-serine/threonine kinases that are activated and tyrosine phosphorylated in response to insulin and NGF.
Cell
1991
;
65
:
663
–75.
2
Ip YT, Davis RJ. Signal transduction by the c-Jun N-terminal kinase (JNK)-from inflammation to development.
Curr Opin Cell Biol
1998
;
10
:
205
–19.
3
Schaeffer HJ, Weber MJ. Mitogen-activated protein kinases: specific messages from ubiquitous messengers.
Mol Cell Biol
1999
;
19
:
2435
–44.
4
Su B, Karin M. Mitogen-activated protein kinase cascades and regulation of gene expression.
Curr Opin Immunol
1996
;
8
:
402
–11.
5
Davis RJ. The mitogen-activated protein kinase signal transduction pathway.
J Biol Chem
1993
;
268
:
14553
–6.
6
Hibi M, Lin A, Smeal T, Minden A, Karin M. Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain.
Genes Dev
1993
;
7
:
2135
–48.
7
Pulverer BJ, Kyriakis JM, Avruch J, Nikolakaki E, Woodgett JR. Phosphorylation of c-jun mediated by MAP kinases.
Nature
1991
;
353
:
670
–4.
8
Bradley JR, Pober JS. Tumor necrosis factor receptor-associated factors (TRAFs).
Oncogene
2001
;
20
:
6482
–91.
9
Chen N, Nomura M, She QB, et al. Suppression of skin tumorigenesis in c-Jun NH(2)-terminal kinase-2-deficient mice.
Cancer Res
2001
;
61
:
3908
–12.
10
Choi BY, Choi HS, Ko K, et al. The tumor suppressor p16(INK4a) prevents cell transformation through inhibition of c-Jun phosphorylation and AP-1 activity.
Nat Struct Mol Biol
2005
;
12
:
699
–707.
11
Huang C, Li J, Ma WY, Dong Z. JNK activation is required for JB6 cell transformation induced by tumor necrosis factor-α but not by 12-O-tetradecanoylphorbol-13-acetate.
J Biol Chem
1999
;
274
:
29672
–6.
12
Lu C, Zhu F, Cho YY, et al. Cell apoptosis: requirement of H2AX in DNA ladder formation, but not for the activation of caspase-3.
Mol Cell
2006
;
23
:
121
–32.
13
She QB, Ma WY, Zhong S, Dong Z. Activation of JNK1, RSK2, and MSK1 is involved in serine 112 phosphorylation of Bad by ultraviolet B radiation.
J Biol Chem
2002
;
277
:
24039
–48.
14
Karin M. Mitogen-activated protein kinase cascades as regulators of stress responses.
Ann N Y Acad Sci
1998
;
851
:
139
–46.
15
Dickens M, Rogers JS, Cavanagh J, et al. A cytoplasmic inhibitor of the JNK signal transduction pathway.
Science
1997
;
277
:
693
–6.
16
Whitmarsh AJ, Cavanagh J, Tournier C, Yasuda J, Davis RJ. A mammalian scaffold complex that selectively mediates MAP kinase activation.
Science
1998
;
281
:
1671
–4.
17
Whitmarsh AJ, Kuan CY, Kennedy NJ, et al. Requirement of the JIP1 scaffold protein for stress-induced JNK activation.
Genes Dev
2001
;
15
:
2421
–32.
18
Yasuda J, Whitmarsh AJ, Cavanagh J, Sharma M, Davis RJ. The JIP group of mitogen-activated protein kinase scaffold proteins.
Mol Cell Biol
1999
;
19
:
7245
–54.
19
Abe Y, Matsumoto S, Kito K, Ueda N. Cloning and expression of a novel MAPKK-like protein kinase, lymphokine-activated killer T-cell-originated protein kinase, specifically expressed in the testis and activated lymphoid cells.
J Biol Chem
2000
;
275
:
21525
–31.
20
Gaudet S, Branton D, Lue RA. Characterization of PDZ-binding kinase, a mitotic kinase.
Proc Natl Acad Sci U S A
2000
;
97
:
5167
–72.
21
Simons-Evelyn M, Bailey-Dell K, Toretsky JA, et al. PBK/TOPK is a novel mitotic kinase which is upregulated in Burkitt's lymphoma and other highly proliferative malignant cells.
Blood Cells Mol Dis
2001
;
27
:
825
–9.
22
Zhao S, Dai J, Zhao W, et al. PDZ-binding kinase participates in spermatogenesis.
Int J Biochem Cell Biol
2001
;
33
:
631
–6.
23
Matsumoto S, Abe Y, Fujibuchi T, et al. Characterization of a MAPKK-like protein kinase TOPK.
Biochem Biophys Res Commun
2004
;
325
:
997
–1004.
24
Park JH, Lin ML, Nishidate T, Nakamura Y, Katagiri T. PDZ-binding kinase/T-LAK cell-originated protein kinase, a putative cancer/testis antigen with an oncogenic activity in breast cancer.
Cancer Res
2006
;
66
:
9186
–95.
25
Cho YY, He Z, Zhang Y, et al. The p53 protein is a novel substrate of ribosomal S6 kinase 2 and a critical intermediary for ribosomal S6 kinase 2 and histone H3 interaction.
Cancer Res
2005
;
65
:
3596
–603.
26
Clark GJ, Cox AD, Graham SM, Der CJ. Biological assays for Ras transformation.
Methods Enzymol
1995
;
255
:
395
–412.
27
Morrison DK, Davis RJ. Regulation of MAP kinase signaling modules by scaffold proteins in mammals.
Annu Rev Cell Dev Biol
2003
;
19
:
91
–118.
28
Bode AM, Dong Z. Mitogen-activated protein kinase activation in UV-induced signal transduction.
Sci STKE
2003
;
2003
:
RE2
.
29
Derijard B, Hibi M, Wu IH, et al. JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain.
Cell
1994
;
76
:
1025
–37.
30
Smeal T, Binetruy B, Mercola DA, Birrer M, Karin M. Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73.
Nature
1991
;
354
:
494
–6.
31
Nandi A, Tidwell M, Karp J, Rapoport AP. Protein expression of PDZ-binding kinase is up-regulated in hematologic malignancies and strongly down-regulated during terminal differentiation of HL-60 leukemic cells.
Blood Cells Mol Dis
2004
;
32
:
240
–5.
32
Yuryev A, Wennogle LP. Novel raf kinase protein-protein interactions found by an exhaustive yeast two-hybrid analysis.
Genomics
2003
;
81
:
112
–25.
33
Kawakami Y, Hartman SE, Holland PM, Cooper JA, Kawakami T. Multiple signaling pathways for the activation of JNK in mast cells: involvement of Bruton's tyrosine kinase, protein kinase C, and JNK kinases, SEK1 and MKK7.
J Immunol
1998
;
161
:
1795
–802.
34
Lang W, Wang H, Ding L, Xiao L. Cooperation between PKC-α and PKC-ε in the regulation of JNK activation in human lung cancer cells.
Cell Signal
2004
;
16
:
457
–67.
35
Lopez-Bergami P, Habelhah H, Bhoumik A, Zhang W, Wang LH, Ronai Z. RACK1 mediates activation of JNK by protein kinase C [corrected].
Mol Cell
2005
;
19
:
309
–20.
36
Dong Z. Effects of food factors on signal transduction pathways.
Biofactors
2000
;
12
:
17
–28.
37
Varfolomeev EE, Ashkenazi A. Tumor necrosis factor: an apoptosis JuNKie?
Cell
2004
;
116
:
491
–7.
38
Xu S, Cobb MH. MEKK1 binds directly to the c-Jun N-terminal kinases/stress-activated protein kinases.
J Biol Chem
1997
;
272
:
32056
–60.
39
Xia Y, Wu Z, Su B, Murray B, Karin M. JNKK1 organizes a MAP kinase module through specific and sequential interactions with upstream and downstream components mediated by its amino-terminal extension.
Genes Dev
1998
;
12
:
3369
–81.
40
Su YC, Han J, Xu S, Cobb M, Skolnik EY. NIK is a new Ste20-related kinase that binds NCK and MEKK1 and activates the SAPK/JNK cascade via a conserved regulatory domain.
EMBO J
1997
;
16
:
1279
–90.
41
de Gruijl FR, van Kranen HJ, Mullenders LH. UV-induced DNA damage, repair, mutations and oncogenic pathways in skin cancer.
J Photochem Photobiol B
2001
;
63
:
19
–27.
42
Setlow RB. The wavelengths in sunlight effective in producing skin cancer: a theoretical analysis.
Proc Natl Acad Sci U S A
1974
;
71
:
3363
–6.
43
Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation.
Biochim Biophys Acta
1991
;
1072
:
129
–57.