Nuclear PKM2 regulates [bgr]-catenin transactivation upon EGFR activation
Weiwei Yang,Yan Xia,Haitao Ji,Yanhua Zheng,Ji Liang,Zhimin Lu,Wenhua Huang,Xiang Gao,Kenneth Aldape
DOI: https://doi.org/10.1038/nature10598
IF: 64.8
2011-01-01
Nature
Abstract:Because both EGFR activation and PKM2 expression are instrumental in tumorigenesis5, 6, 7, we examined whether EGFR activation regulates PKM2 functions in a subcellular-compartment-dependent manner. Immunofluorescence analysis showed that EGF treatment resulted in the nuclear accumulation of PKM2 in U87/EGFR human glioblastoma (GBM) cells (Fig. 1a). In addition, expression of the constitutively active EGFRvIII mutant in U87 cells resulted in a higher amount of nuclear PKM2 than was observed in EGF-untreated U87/EGFR cells (Supplementary Fig. 2a). The finding that EGF induces nuclear translocation of PKM2 was further supported by cell fractionation analysis of DU145 prostate cancer cells, MDA-MB-231 breast cancer cells and U87/EGFR cells (Supplementary Fig. 2b). In addition, PKM1 failed to translocate into the nucleus upon EGF stimulation (Supplementary Fig. 2c), indicating that EGF specifically regulates the subcellular distribution of PKM2 in multiple types of cancer cells. To examine whether PKM2 directly regulates gene transcription and cell proliferation, we expressed PKM2 short hairpin RNA (shRNA) in U87/EGFR cells (Supplementary Fig. 3a). PKM2 depletion largely reduced both basal and EGF-induced tumour cell proliferation (Fig. 1b) and blocked EGF-enhanced expression of cyclin D1 and c-Myc (Fig. 1c), which are known to be important regulators of cell proliferation and downstream genes of β-catenin transactivation8. To examine whether these PKM2-dependent effects were mediated by β-catenin, we performed TCF/LEF-1 luciferase reporter analyses, showing that PKM2 depletion significantly inhibited EGF-induced β-catenin transactivation (Fig. 1d). In addition, chromatin immunoprecipitation (ChIP) analyses showed that EGFR activation resulted in increased binding of β-catenin to the promoter region of CCND1 (coding for cyclin D1) (Fig. 1e) and c-Myc (data not shown), which was blocked by PKM2 depletion. In addition, co-immunoprecipitation analyses showed that PKM2 depletion inhibited the EGF-induced interaction between β-catenin and Myc-tagged TCF4 (Fig. 1f). However, PKM2 depletion failed to inhibit WNT3A- or WNT1- (data not shown) induced β-catenin transactivation (Supplementary Fig. 3b) and cyclin D1 expression (Supplementary Fig. 3c). In addition, WNT3A did not induce PKM2 nuclear translocation (Supplementary Fig. 3d). These results indicate that EGF induces β-catenin transactivation via a mechanism distinct from that induced by Wnt/Wingless4 and that PKM2 expression has a pivotal role in EGF-, but not Wnt-induced β-catenin transactivation. To examine the mechanism underlying PKM2-regulated β-catenin transactivation, we performed co-immunoprecipitation analyses, showing that EGF stimulation resulted in an interaction between endogenous PKM2 and β-catenin in the nuclear, but not cytosolic, fraction of U87/EGFR cells (Fig. 1g). However, an in vitro glutathione S-transferase (GST) pull-down assay showed that purified GST–β-catenin failed to bind to purified His–PKM2 (Supplementary Fig. 4). These results suggest that the interaction of these two proteins might require post-translational modifications of the proteins. PKM2 binds to tyrosine-phosphorylated peptides, and expression of the phosphotyrosine-binding form is required for cancer cell growth9. To examine whether β-catenin is tyrosine phosphorylated, we performed immunoblotting analyses with a phospho-Tyr antibody, showing that EGF stimulation induced Tyr phosphorylation of immunoprecipitated β-catenin in the nucleus, but not in the cytosol or at the plasma membrane (Fig. 1h). Treatment of the immunoprecipitated β-catenin with calf intestinal alkaline phosphatase (CIP) resulted in β-catenin dephosphorylation and abrogation of the PKM2–β-catenin interaction (Fig. 1i). Thus, EGF-induced Tyr phosphorylation of β-catenin is required for the PKM2–β-catenin interaction. The PKM2 K433E mutant, which fails to bind to tyrosine-phosphorylated peptides9, had similar glycolytic enzyme activity to its wild-type counterpart (Supplementary Fig. 5)9. Co-immunoprecipitation analyses showed that EGF treatment induced the binding of β-catenin to Flag-tagged wild-type PKM2, but not to the PKM2 K433E mutant (Fig. 1j). In contrast, a kinase-dead Flag–PKM2 K367M mutant10, 11, acting like its wild-type counterpart, binds to β-catenin (Fig. 1k). These results indicate that the K433 binding residue of PKM2, but not its catalytic activity, is critical for the PKM2–β-catenin interaction. Abl and c-Src have been reported to phosphorylate β-catenin12, 13. Pre-treatment with SU6656 (an Src inhibitor) or an Abl inhibitor completely abrogated EGF-induced activation of c-Src or Abl, as shown by the reduced levels of c-Src (Y418) or Abl (Y412) phosphorylation (Supplementary Fig. 6a). However, inhibition of c-Src, but not Abl, blocked EGF-induced Tyr phosphorylation of β-catenin (Fig. 2a). In addition, deficiency of c-Src (Fig. 2b), but not of Abl (Supplementary Fig. 6b), abrogated EGF-induced β-catenin Tyr phosphorylation and the PKM2–β-catenin interaction. These results indicate that c-Src, a downstream effector of EGFR, phosphorylates β-catenin, which is required for the PKM2–β-catenin interaction. To examine the subcellular compartment in which c-Src phosphorylates β-catenin, we conducted fractionation analyses. We found that EGF stimulation resulted in the nuclear translocation of c-Src (Supplementary Fig. 6c). In addition, co-immunoprecipitation analyses showed that EGF treatment induced an enhanced interaction between β-catenin and c-Src in nuclear fractions (Fig. 2c). These results, combined with the evidence that β-catenin is phosphorylated in the nucleus (Fig. 1h), strongly suggest that c-Src translocates into the nucleus and subsequently interacts with and phosphorylates β-catenin. The Y86 residue of β-catenin has been shown to be phosphorylated by c-Src as well as BCR–ABL12, 13, and analysis of the amino acid sequence identified an additional potential Src phosphorylation site at Y333. Immunoblotting analysis showed that EGF stimulation resulted in tyrosine phosphorylation of Flag-tagged wild-type β-catenin and β-catenin Y86F, but not β-catenin Y333F, which was further validated by immunoblotting with a phospho-β-catenin Y333 antibody (Fig. 2d). In addition, cell fractionation analysis demonstrated that EGF induced β-catenin Y333 phosphorylation primarily in the nucleus (Supplementary Fig. 6d). Furthermore, an in vitro protein kinase assay showed that active c-Src was able to phosphorylate wild-type β-catenin (Fig. 2e), but the β-catenin Y333F mutation largely reduced total Tyr-phosphorylation levels and completely abrogated the Y333 phosphorylation. These results reveal that c-Src binds and phosphorylates β-catenin at Y333 in vitro and in vivo. To examine whether phosphorylation of β-catenin Y333 regulates its binding to PKM2, we performed a GST pull-down assay by mixing purified GST–β-catenin and His–PKM2 with or without purified active c-Src. Figure 2f shows that wild-type PKM2 did not bind to unphosphorylated wild-type β-catenin. However, the presence of c-Src enabled the binding of wild-type PKM2 to β-catenin, which was abrogated by mutation of β-catenin at Y333 or PKM2 at K433. These in vitro results were further validated by co-immunoprecipitation analyses, showing that in contrast to Flag-tagged wild-type β-catenin or β-catenin Y86F, Flag–β-catenin Y333F failed to bind to endogenous PKM2 (Fig. 2g). These results indicate that c-Src-mediated β-catenin Y333 phosphorylation is required for the PKM2–β-catenin interaction. We next examined the importance of the PKM2–β-catenin interaction in β-catenin transactivation. Figure 3a shows that Flag–β-catenin Y333F failed to interact with Myc-tagged TCF4 upon EGF stimulation, in contrast to its wild-type counterpart. ChIP analyses demonstrated that Flag-tagged wild-type β-catenin bound to the CCND1 promoter region upon EGFR activation, which was abrogated by the Y333F mutation (Fig. 3b). In addition, reconstituted expression of the RNA interference (RNAi)-resistant (r)β-catenin Y333F mutant, but not of wild-type rβ-catenin, in endogenous β-catenin-depleted U87/EGFRvIII cells failed to induce cyclin D1 and c-Myc expression (Fig. 3c). Furthermore, Flag–PKM2 K433E and the inactive Flag–PKM2 K367M mutant translocated into the nucleus upon EGF stimulation (Supplementary Fig. 7), but reconstituted expression of these mutants (Supplementary Fig. 8) failed to induce cyclin D1 expression as did wild-type rPKM2 expression (Fig. 3d). Thus, both PKM2 catalytic activity and the PKM2–β-catenin interaction are required for cyclin D1 expression upon EGFR activation. To compare downstream targets of EGF and Wnt signalling, we examined the expression of other Wnt/β-catenin downstream genes: AXIN2, DKK1 and βTrCP (also known as BTRCP)8. Quantitative reverse transcription–polymerase chain reaction (RT–PCR) analysis showed that EGF treatment increased mRNA levels of DKK1, but not of AXIN2 or βTrCP, which was blocked by PKM2 depletion (Supplementary Fig. 9). These results indicate that EGF-induced and PKM2-dependent β-catenin transactivation induced transcription of a set of genes, which do not completely overlap with those induced by Wnt signalling. The finding that PKM2 was not required for WNT3A-induced β-catenin transactivation (Supplementary Fig. 3b) suggested that c-Src-dependent β-catenin Y333 phosphorylation is not involved in Wnt-induced signalling or cell adhesion. This assumption was supported by the results, which showed that β-catenin Y333F behaved similarly to wild-type β-catenin both in binding to APC, AXIN2 and E-cadherin and in Wnt-induced β-catenin transactivation and cellular functions (Supplementary Fig. 10a–g). In contrast, expression of β-catenin Y333F blocked EGF- but not WNT3A-induced cell migration (Supplementary Fig. 10h). To investigate the mechanisms underlying PKM2- and β-catenin-dependent cyclin D1 expression, we performed ChIP analyses. Figure 3e shows that EGF induced an enhanced binding of wild-type Flag–PKM2 and Flag–PKM2 K367M, but not of Flag–PKM2 K433E, to the CCND1 promoter region. In addition, binding of Flag–PKM2 to the promoter region was not detected in ChIP analyses after β-catenin had been immunodepleted from cell lysates (Fig. 3f). These results indicate that the PKM2–β-catenin interaction, but not PKM2 kinase activity, is required for both proteins to bind to the CCND1 promoter region. We next examined whether PKM2 binding to the CCND1 promoter region regulates histone H3 acetylation, which is important for gene transcription. ChIP analyses showed that EGF treatment resulted in a significant increase of histone H3 acetylation in the CCND1 promoter region, which was blocked by PKM2 depletion (Fig. 3g). Reconstituted expression of rPKM2 K433E and the inactive rPKM2 K367M mutant failed to restore EGF-induced histone H3 acetylation, compared with the wild-type protein. To understand further the mechanism underlying PKM2-regulated histone H3 acetylation, we performed ChIP analyses with antibodies against ubiquitously expressed histone deacetylase (HDAC)1, HDAC2 and HDAC3 (ref. 14). Figure 3h shows that HDAC3, but not HDAC1 or HDAC2 (Supplementary Fig. 11a), was pre-bound to the CCND1 promoter. EGF treatment resulted in the disassociation of HDAC3 from the CCND1 promoter, and this disassociation was blocked by PKM2 depletion. Furthermore, reconstituted expression of wild-type rPKM2, but not of rPKM2 K367M or rPKM2 K433E mutants, was able to restore EGF-induced disassociation of HDAC3 from the promoter (Fig. 3h). These results indicate that the kinase activity of PKM2 and its binding with β-catenin to the CCND1 promoter region are required for HDAC3 removal from the promoter. An additional co-immunoprecipitation analysis showed that wild-type PKM2, but not PKM2 K367M, interacted with HDAC3 upon EGF treatment (Supplementary Fig. 11b). These results suggest that PKM2 can interact with HDAC3 in an active conformation, which facilitates HDAC3 removal from the CCND1 promoter, acetylation of histone H3, and cyclin D1 expression. To support the findings that EGF-induced and c-Src-dependent PKM2–β-catenin interaction and subsequent cyclin D1 expression are not cell-line specific, we treated GSC11 and GSC23 human primary GBM cells with EGF. Supplementary Fig. 12a shows that EGF treatment results in nuclear translocation of PKM2 and cyclin D1 expression in these cells. In addition, EGF-induced phosphorylation of β-catenin Y333 (Supplementary Fig. 12b) and its association with PKM2 (Supplementary Fig. 12c) were blocked by pre-treatment with SU6656. We next examined the significance of the PKM2–β-catenin interaction in tumour cell proliferation. Figure 4a shows that U87/EGFRvIII cells proliferated much faster than did parental U87 cells. Expression of β-catenin shRNA (Fig. 3c) largely inhibited U87/EGFRvIII cell proliferation, which was rescued by reconstituted expression of wild-type rβ-catenin, but not of the rβ-catenin Y333F mutant (Fig. 4a). In addition, EGFRvIII-promoted cell proliferation was similarly inhibited by PKM2 depletion, which was rescued by the reconstituted expression of wild-type rPKM2, but not of the rPKM2 K433E or rPKM2 K367M mutant (Supplementary Fig. 13a and Fig. 4b). β-Catenin-regulated cyclin D1 expression is critical for G1–S phase transition and cell cycle progression15. Depletion of β-catenin or PKM2 resulted in an accumulation of U87/EGFRvIII cells in G0/G1 phase, which was rescued by reconstituted expression of wild-type rβ-catenin or wild-type rPKM2, but not of the rβ-catenin Y333F or rPKM2 K433E mutant (Supplementary Fig. 13b). Thus, the PKM2–β-catenin interaction is essential for cell cycle progression. To determine the role of PKM2-dependent β-catenin transactivation in brain tumour development, we intracranially injected U87 or U87/EGFRvIII cells into athymic nude mice. U87 cells did not form a detectable tumour 2 weeks after injection (Fig. 4c, bottom left). In contrast, U87/EGFRvIII cells elicited rapid tumorigenesis (Fig. 4c, top left). Notably, depletion of β-catenin (Fig. 4c, top) or PKM2 (Fig. 4c, bottom) abrogated EGFRvIII-driven tumour growth, which was rescued by expression of wild-type rβ-catenin or wild-type rPKM2, but not of the rβ-catenin Y333F or rPKM2 K433E mutant. Similar results were obtained using GSC11 cells (Supplementary Fig. 14a). In addition, adding doxycycline to the drinking water 14 days after intracranial injection of GSC11 cells that expressed a tetracycline-inducible PKM2 shRNA partially depleted PKM2 expression in tumour tissues and inhibited tumour growth (Supplementary Fig. 14b). Thus, the PKM2–β-catenin interaction and PKM2 expression is instrumental in tumour growth. To support further the role of c-Src in EGFR-induced β-catenin transactivation in vivo, we injected SU6656 intratumorally, which significantly blocked tumour growth (Supplementary Fig. 15a, b), reduced the phosphorylation levels of c-Src Y418 and β-catenin Y333, and inhibited cyclin D1 expression in tumour tissue (Supplementary Fig. 15c). The requirement of β-catenin transactivation in tumorigenesis was also examined by expression of WNT1 in U87/EGFRvIII-PKM2 shRNA cells. WNT1 expression resulted in the induction of cyclin D1 (Supplementary Fig. 16a) and largely rescued PKM2 depletion-blocked tumorigenesis (Supplementary Fig. 16b, c). In addition, this effect was further enhanced using WNT1-expressing U87/EGFRvIII cells with reconstituted expression of rPKM2 K433E, which retains its catalytic activity for glycolysis. These results suggest that, whereas the metabolic function of PKM2 has a critical role in aerobic glycolysis7 and tumorigenesis, brain tumour development promoted by EGFR requires PKM2-modulated β-catenin transactivation. Analysis of publicly available microarray data sets (Affymetrix, U133) from The Cancer Genome Atlas (TCGA) and other sources16, 17, 18, 19 revealed a correlation of c-Myc and CCND1 expression with EGFR expression in GBM samples (Supplementary Fig. 17). In addition, phosphorylation levels of β-catenin Y333 correlated with phosphorylation levels of activated c-Src in seven human primary GBM cell lines (Supplementary Fig. 18). We next performed immunohistochemical (IHC) analyses to examine c-Src activity, β-catenin Y333 phosphorylation, and PKM2 nuclear localization in serial sections of 55 human primary GBM specimens by using antibodies with validated specificities (Supplementary Fig. 19). Figure 4d shows that the levels of c-Src Y418 phosphorylation, β-catenin Y333 phosphorylation, and nuclear PKM2 expression were correlated with each other. In addition, β-catenin Y333 phosphorylation accumulated in the nuclei of a large percentage of tumour cells (Fig. 4d, left). Quantification of the staining upon a scale of 0 to 8.0 showed that these correlations were significant (Supplementary Fig. 20). The survival durations of 84 patients, all of whom received standard adjuvant radiotherapy after surgery, followed by treatment with an alkylating agent (temozolomide in the majority of cases), with low (0–5 staining) versus high (5.1–8 staining) β-catenin Y333 phosphorylation and nuclear PKM2 expression were compared. Patients whose tumours had low β-catenin Y333 phosphorylation or nuclear PKM2 expression had a median survival of 185.2 and 130.0 weeks, respectively. The median survival of patients decreased to 69.4 and 82.5 weeks, respectively, when their tumours showed high levels of β-catenin Y333 phosphorylation or nuclear PKM2 expression. In a Cox multivariate model, the IHC scores of β-catenin phosphorylation (Fig. 4e) and nuclear PKM2 expression (Fig. 4f) were independent predictors of glioblastoma patient survival, after adjusting for the age of the patient, a relevant clinical covariate. These results support the role of PKM2 association with c-Src-phosphorylated β-catenin Y333 in the clinical behaviour of human GBM and reveal a relationship between β-catenin Y333 phosphorylation/nuclear PKM2 localization and clinical aggressiveness of the tumour. These findings were further supported by IHC analyses, showing significantly lower levels of β-catenin Y333 phosphorylation in low-grade diffuse astrocytoma (30 cases) (World Health Organization (WHO) grade II; median survival time >5 years) than were present in GBM specimens (WHO grade IV)20 (Supplementary Fig. 21). We previously showed that GSK-3β-independent transactivation of β-catenin by growth factor receptor occurs by mechanisms distinct from Wnt-dependent canonical signalling4, 21, 22, 23. In this study, we describe an important and previously unknown mechanism underlying EGFR activation-induced β-catenin transactivation through interaction with PKM2, which has a critical role in transcription of CCND1 and c-Myc (Supplementary Fig. 1). The understanding that phosphorylation of β-catenin Y333 and its interaction with PKM2 are required for tumour cell proliferation and tumour development, and that the levels of β-catenin Y333 phosphorylation and nuclear PKM2 correlate with grades of glioma malignancy and prognosis, may provide a molecular basis for improved diagnosis and treatment of tumours with activated EGFR and upregulated PKM2. Our findings, in combination with previous reports7, 24, delineate two essential mechanisms underlying tumour development by regulation of metabolic and non-metabolic functions of PKM2: (1) PKM2 enhances aerobic glycolysis7, 24; and (2) PKM2 promotes tumour cell proliferation by binding to and transactivating Y333-phosphorylated β-catenin. Thus, PKM2 has dual roles that are essential for tumorigenesis: regulating cancer cell metabolism and gene transcription required for cell proliferation. The coordinated control of metabolism and proliferation by PKM2 is essential for tumorigenesis. U87, U87/EGFR and U251 GBM cells; DU145 prostate cancer cells; MDA-MB-231 breast cancer cells; A431 epidermoid carcinoma cells and NIH3T3, 293T, c-SRC+/+, c-SRC−/−, ABL+/+ and ABL−/− cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% bovine calf serum (HyClone). Human primary GBM cells were maintained in DMEM/F-12 50/50 supplemented with B27, EGF (10 ng ml−1), bFGF (10 ng ml−1). Rat pheochromocytoma PC12 cells were maintained in DMEM supplemented with 10% horse serum and 5% fetal bovine serum. Cell cultures were made quiescent by growing them to confluence, and the medium was replaced with fresh medium containing 0.5% serum for 1 day. EGF with 100 ng ml−1 final concentration was used for cell stimulation. Rabbit polyclonal antibodies recognizing phospho-β-catenin Y333, PKM1, PKM2, c-Src, Abl, phospho-Abl Y412 and c-Myc were obtained from Signallway Biotechnology. A mouse antibody recognizing phospho-tyrosine was obtained from BD Biosciences. A monoclonal antibody against phospho-c-Src Y418 was purchased from Millipore. Polyclonal antibodies for cyclin D1 and PCNA and monoclonal antibodies for β-catenin and Myc tag were purchased from Santa Cruz Biotechnology. A polyclonal antibody of acetylated histone H3 and a monoclonal antibody for HDAC3 were obtained from Upstate Biotechnology. EGF and mouse monoclonal antibodies for Flag, GST, His and tubulin were purchased from Sigma. Hygromycin, puromycin, G418, SU6656, Abl inhibitor, cycloheximide, DNase-free RNase A and propidium iodide were purchased from EMD Biosciences. Active c-Src was obtained from Signallchem. Hoechst 33342 and Alexa Fluor 488 goat anti-rabbit antibody was from Molecular Probes. HyFect transfection reagents were from Denville Scientific. GelCode Blue Stain Reagent was obtained from Pierce. Cells were plated at a density of 4 × 105 per 60-mm dish 18 h before transfection. Transfection was performed as previously described14. Extraction of proteins with a modified buffer from cultured cells was followed by immunoprecipitation and immunoblotting with corresponding antibodies, as described previously25. 2 × 104 cells were plated and counted 7 days after seeding in DMEM with 0.5% bovine calf serum. Data represent the means ± s.d. of three independent experiments. PCR-amplified human PKM1 was cloned into pcDNA3.1/hygro (+) vector between BamHI and XhoI. PCR-amplified human PKM2 was cloned into pcDNA3.1/hygro (+) vector between BamHI and NotI. β-Catenin was subcloned into pGEX-4T-1 vector between BamHI and NotI. pcDNA 3.1/hygro (+)-PKM2, -K433E, -K367M, pcDNA 3.1/hygro (+)-β-catenin Y86F, and -β-catenin Y333F were made using the QuikChange site-directed mutagenesis kit (Stratagene). pCDNA 3.1-rPKM2 contains mutations of C1192T and C1194G. The pGIPZ control was generated with a control oligonucleotide GCTTCTAACACCGGAGGTCTT. pGIPZ PKM2 shRNA and pTRIPZ PKM2 shRNA were generated with TTATTTGAGGAACTCCGCCGC oligonucleotide targeting exon 10 of the PKM2 transcript. pGIPZ β-catenin shRNA was generated with CATGCACAAGAATGGATCACAA. 1 × 106 treated cells were fixed in cold 70% ethanol for 3 h, spun down, and incubated for 1 h at 37 °C in PBS with DNase-free RNase A (100 μg ml−1) and propidium iodide (50 μg ml−1). Cells were then analysed by fluorescence-activated cell sorting (FACS). The wild-type and mutants of GST–β-catenin were expressed in bacteria and purified as described previously14. The kinase reactions were performed as described previously22. The transcriptional activation of β-catenin in 293T cells was measured as previously described22. The relative levels of luciferase activity were normalized to the levels of untreated cells and to the levels of luciferase activity of the Renilla control plasmid. ChIP was performed using an Upstate Biotechnology kit. Chromatin prepared from cells (in a 10-cm dish) was used to determine total DNA input and for overnight incubation with the specific antibodies or with normal rabbit or mouse immunoglobulin G. The human CCND1 promoter-specific primers used in PCR were 5′-GGGGCGATTTGCATTTCTAT-3′ (forward) and 5′-CGGTCGTTGAGGAGGTTGG-3′ (reverse). Immunofluorescence analysis was performed as described previously22. Nuclei, cytosol and cell membranes were isolated using the Nuclear Extract Kit from Active Motif North America and the ProteoExtract Subcellular Proteome Extraction Kit from Calbiochem. Mouse tumour tissues were fixed and prepared for staining. The specimens were stained with Mayer’s haematoxylin and subsequently with eosin (Biogenex Laboratories). Afterwards, the slides were mounted using Universal Mount (Research Genetics). The tissue sections from paraffin-embedded human GBM specimens were stained with antibodies against phospho-c-Src Y418, phospho-β-catenin Y333, PKM2, or nonspecific IgG as a negative control. We quantitatively scored the tissue sections according to the percentage of positive cells and staining intensity, as previously defined21. We assigned the following proportion scores: 0 if 0% of the tumour cells showed positive staining, 1 if 0% to 1% of cells were stained, 2 if 1% to 10% stained, 3 if 11% to 30% stained, 4 if 31% to 70% stained, and 5 if 71% to 100% stained. We rated the intensity of staining on a scale of 0 to 3: 0, negative; 1, weak; 2, moderate; and 3, strong. We then combined the proportion and intensity scores to obtain a total score (range, 0–8), as described previously21. Scores were compared with overall survival, defined as the time from date of diagnosis to death or last known date of follow-up. All patients received standard adjuvant radiotherapy after surgery, followed by treatment with an alkylating agent (temozolomide in the majority of cases). The use of human brain tumour specimens and the database was approved by the institutional review board at MD Anderson Cancer Center. We intracranially injected 5 × 105 GBM cells (in 5 μl of DMEM per mouse), with or without regulation of β-catenin or PKM2 expression, into 4-week-old female athymic nude mice. The intracranial injections were performed as described in a previous publication26. Seven mice per group in each experiment were included. Animals injected with U87/EGFRvIII or GSC 11 cells were killed 2 weeks or 30 days after glioma cell injection, respectively. The brain of each mouse was harvested, fixed in 4% formaldehyde, and embedded in paraffin. Tumour formation and phenotype were determined by histological analysis of H&E-stained sections. For doxycycline induction studies, 7 mice were killed 14 days after GBM cell injection to examine tumour growth, whereas the remaining 14 mice in two groups were fed with or without water containing 800 μg ml−1 doxycycline. The water containing doxycycline was changed every 3 days. The mice in these two groups were killed at day 30. For the SU6656 treatment studies, 7 mice were killed 5 days after U87/EGFRvIII cell injection to examine tumour growth, whereas the remaining 14 mice in two groups were treated with either DMSO or SU6656. SU6656 (0.015 mg kg−1 in 5 μl of DMSO) was intracranially injected into the tumour every 3 days, and the mice in both groups were killed at day 14 after GBM cell injection. Mouse NGF (Upstate Biotechnology) was added at 100 ng ml−1 to PC12 cells in culture, and the medium was changed every 3 days. Two hundred cells from 10 randomly chosen microscopic fields for each condition were examined. A cell was considered to respond to NGF if it extended neurites at least two cell bodies long after the incubation duration. The activity of bacterially purified wild-type PKM2 (0.1 μg) and PKM2 K433E (0.1 μg) towards PEP was measured by a pyruvate kinase assay (BioVision), according to the manufacturer’s instruction. Data represent the mean ± s.d. of three independent experiments. Matrigel-transwell assay was performed as described previously22. We transfected 293T cells in 150-mm plates with 12 μg pFU-CGW WNT1, 4 μg pMDL, 4 μg pRSV-Rev, and 4 μg pCMV-VSVG, a plasmid encoding the G-protein of the vesicular stomatitis virus (VSV-G) envelope. The medium was changed the next day. The medium containing lentivirus was harvested at 48 h and 72 h after transfection. Virus particles were concentrated and purified by ultra-high-speed centrifugation (25,000g for 2 h at 4 °C). Cells were infected with lentivirus (1 × 106) in the presence of 6 μg ml−1 polybrene (Sigma). Total RNA was extracted using a RNA High-purity Total RNA Rapid Extraction Kit (Signallway Biotechnology). cDNA was prepared using oligonucleotide (dT), random primers, and a Thermo Reverse Transcription kit (Signallway Biotechnology). Quantitative real-time PCR analysis was performed using 2× SIBR real-time PCR Premixture (Signallway Biotechnology) under the following conditions: 5 min at 95 °C followed by 40 cycles at 95 °C for 30 s, 55 °C for 40 s, and 72 °C for 1 min using an ABI Prism 7700 sequence detection system. Data were normalized to expression of a control gene (b-actin) for each experiment. The following primer pairs were used for quantitative real-time PCR: DKK1, 5′-CATTGACAACTACCAGCCGTAC-3′ (forward) and 5′-GGGCAGCACATAGCGTGA-3′ (reverse); AXIN2, 5′-GGGAGCCTAAAGGTCGTG-3′ (forward) and 5′-GGGTTCTCGGGAAATGAG-3′ (reverse); βTRCP, 5′-CCCCAACTGACATTACCC-3′ (forward) and 5′-TCGAATACAACGCACCAA-3′ (reverse); β-actin, 5′-ATGGATGACGATATCGCTGCGC-3′ (forward) and 5′-GCAGCACAGGGTGCTCCTCA-3′ (reverse). Download references We thank T. Hunter (The Salk Institute for Biological Studies) for Abl and Src knockout cells, H. Clevers (Netherlands Institute for Developmental Biology) for the pTOP-FLASH and the pFOP-FLASH, and Y. Li (Baylor College of Medicine) for a WNT1 lenti-vector. This work was supported by National Cancer Institute grants 5R01CA109035 (Z.L.), 5 P50 CA127001-03 and CA16672 (Cancer Center Support Grant); a research grant (RP110252; Z.L.) from the Cancer Prevention and Research Institute of Texas (CPRIT), an American Cancer Society Research Scholar Award RSG-09-277-01-CSM (Z.L.), and a Sister Institution Network Fund from The University of Texas MD Anderson Cancer Center (Z.L.). Subscribe to comments
What problem does this paper attempt to address?
-
Nuclear PKM2 Regulates Β-Catenin Transactivation Upon EGFR Activation.
Weiwei Yang,Yan Xia,Haitao Ji,Yanhua Zheng,Ji Liang,Wenhua Huang,Xiang Gao,Kenneth Aldape,Zhimin Lu
DOI: https://doi.org/10.1038/nature10598
IF: 64.8
2011-01-01
Nature
Abstract:The embryonic pyruvate kinase M2 (PKM2) isoform is highly expressed in human cancer. In contrast to the established role of PKM2 in aerobic glycolysis or the Warburg effect 1 , 2 , 3 , its non-metabolic functions remain elusive. Here we demonstrate, in human cancer cells, that epidermal growth factor receptor (EGFR) activation induces translocation of PKM2, but not PKM1, into the nucleus, where K433 of PKM2 binds to c-Src-phosphorylated Y333 of β-catenin. This interaction is required for both proteins to be recruited to the CCND1 promoter, leading to HDAC3 removal from the promoter, histone H3 acetylation and cyclin D1 expression. PKM2-dependent β-catenin transactivation is instrumental in EGFR-promoted tumour cell proliferation and brain tumour development. In addition, positive correlations have been identified between c-Src activity, β-catenin Y333 phosphorylation and PKM2 nuclear accumulation in human glioblastoma specimens. Furthermore, levels of β-catenin phosphorylation and nuclear PKM2 have been correlated with grades of glioma malignancy and prognosis. These findings reveal that EGF induces β-catenin transactivation via a mechanism distinct from that induced by Wnt/Wingless 4 and highlight the essential non-metabolic functions of PKM2 in EGFR-promoted β-catenin transactivation, cell proliferation and tumorigenesis.
-
β-Catenin Controls the Electrophysiologic Properties of Skeletal Muscle Cells by Regulating the α2 Isoform of Na + /K + -ATPase.
Congying Zhao,Yonglin Yu,Yi Zhang,Jue Shen,Lihua Jiang,Guoxia Sheng,Weiqin Zhang,Lu Xu,Kewen Jiang,Shanshan Mao,Peifang Jiang,Feng Gao
DOI: https://doi.org/10.3389/fnins.2019.00831
IF: 4.3
2019-01-01
Frontiers in Neuroscience
Abstract:beta-Catenin is a key component of the canonical Wnt signaling pathway. It has been shown to have an important role in formation of the neuromuscular junction. Our previous studies showed that in the absence of beta-catenin, the resting membrane potential (RMP) is depolarized in muscle cells and expression of the alpha 2 subunit of sodium/potassium adenosine triphosphatase (alpha 2NKA) is reduced. To understand the underlying mechanisms, we investigated the electrophysiologic properties of a primary cell line derived from mouse myoblasts (C2C12 cells) that were transfected with small-interfering RNAs and over-expressed plasmids targeting beta-catenin. We found that the RMP was depolarized in beta-catenin knocked-down C2C12 cells and was unchanged in beta-catenin over-expressed muscle cells. An action potential (AP) was not released by knockdown or over-expression of beta-catenin. alpha 2NKA expression was reduced by beta-catenin knockdown, and increased by beta-catenin over-expression. We showed that beta-catenin could interact physically with alpha 2NKA (but not with alpha 1NKA) in muscle cells. NKA activity and alpha 2NKA content in the cell membranes of skeletal muscle cells were modulated positively by beta-catenin. These results suggested that beta-catenin (at least in part) regulates the RMP and AP in muscle cells, and does so by regulating alpha 2NKA.
-
Mechanisms of Cytoplasmic β-Catenin Accumulation and Its Involvement in Tumorigenic Activities Mediated by Oncogenic Splicing Variant of the Receptor Originated from Nantes Tyrosine Kinase
Xiang-Ming Xu,Yong-Qing Zhou,Ming-Hai Wang
DOI: https://doi.org/10.1074/jbc.M414699200
IF: 5.485
2005-01-01
Journal of Biological Chemistry
Abstract:The beta-catenin pathway plays a critical role in the pathogenesis of certain types of cancers. To gain insight into mechanisms by which altered receptor tyrosine kinases regulate cytoplasmic beta-catenin accumulation, the effect of an oncogenic receptor originated from Nantes (RON) variant on beta-catenin accumulation and the role of beta-catenin in RON-mediated tumorigenic activities were studied. In NIH3T3 cells harboring oncogenic variant RON Delta 160, increased beta-catenin accumulation with tyrosine phosphorylation and nuclear translocation was observed. Overexpression of RON Delta 160 also resulted in increased expression of beta-catenin target genes c-myc and cyclin D1. By analyzing cellular proteins that regulate beta-catenin stabilities, it was found that RON Delta 160 activates the protein disheveled (DVL) and inactivates glycogen synthase kinase-3 beta by Ser-9 residue phosphorylation. These effects were channeled by RON Delta 160-activated PI 3-kinase-AKT pathways that are sensitive to specific inhibitors, such as wortmannin, but not to other chemical inhibitors. Silencing RON Delta 160 expression by specific small interfering RNA blocked not only beta-catenin expression but also c-myc and cyclin D1 expression, suggesting that RON expression is required for the activation of the beta-catenin signaling pathway. Moreover, it was found that knockdown of the beta-catenin gene expression by small interfering RNA techniques reduces significantly the RON Delta 160-mediated NIH3T3 cell proliferation, focus-forming activities and anchorage-independent growth. Thus, the oncogenic RON variant regulates beta-catenin stabilities through activation of DVL and inactivation of glycogen synthase kinase-3 beta. The activated beta-catenin cascade is one of the pathways involved in tumorigenic activities mediated by the oncogenic RON variant.
-
Opposing Roles of Conventional and Novel PKC Isoforms in Hippo-YAP Pathway Regulation
Gong, Rui,Hong, Audrey W,Plouffe, Steven W,Zhao, Bin,Liu, Guangbo,Yu, Fa-Xing,Xu, Yanhui,Guan, Kun-Liang
DOI: https://doi.org/10.1038/cr.2015.88
IF: 44.1
2015-01-01
Cell Research
Abstract:LETTER TO THE EDITOR Cell Research (2015) 25:985-988. © 2015 IBCB, SIBS, CAS All rights reserved 1001-0602/15 $ 32.00 www.nature.com/cr npg Opposing roles of conventional and novel PKC isoforms in Hippo-YAP pathway regulation Cell Research (2015) 25:985-988. doi:10.1038/cr.2015.88; published online 24 July 2015 Dear Editor, The Hippo-YAP pathway is an evolutionally conserved signaling module that controls tissue growth during de- velopment and its dysregulation causes cancer [1]. Core components of the Hippo pathway include a kinase cas- cade comprising MST1/2 and LATS1/2 kinases, in which MST1/2 phosphorylates and activates LATS1/2 [2]. The major downstream effectors of the Hippo pathway are the transcriptional co-activators YAP and TAZ, which are phosphorylated and inhibited by LATS1/2 [3, 4]. Un- phosphorylated YAP/TAZ localizes in the nucleus and promotes target gene expression through binding to the TEAD family transcription factors [5, 6]. Protein kinase C (PKC) controls a broad range of biological processes and can be classified into three sub-groups based on sequence homology and activation mechanisms: the Ca 2+ - and dia- cylglycerol (DAG)-dependent conventional PKC (cPKC α, βI, βII, and γ), the DAG-dependent novel PKC (nPKC δ, θ, e, and η), and the Ca 2+ - and DAG-independent atypical PKC (aPKC ι and ζ) [7]. Recent studies have established that extracellular diffusible signals act through G-protein coupled receptors (GPCRs) to regulate the Hippo-YAP pathway [8, 9]. PKC represents one of the major effectors downstream of GPCRs (especially Gq/11-coupled recep- tors). This led us to investigate whether PKC regulates the Hippo-YAP pathway. We used the DAG analog TPA to activate PKC in HE- K293A cells and observed that TPA induced a rapid and robust YAP dephosphorylation as determined by western blotting using the phospho-specific (serine 127) YAP an- tibody and by differential electrophoretic mobility shift on phos-tag-containing gels. Similarly, TPA also induced TAZ dephosphorylation as indicated by a faster electro- phoretic migration (Figure 1A). TPA-induced YAP de- phosphorylation was also observed in HeLa and U251MG cells (Supplementary information, Figure S1A and S1B). Phosphorylated YAP localizes in the cytoplasm, whereas dephosphorylated YAP translocates to the nucleus to pro- mote target gene expression. Consistent with this model, TPA induced YAP nuclear localization in HEK293A cells (Figure 1B). Moreover, GO6983, an inhibitor of both cPKC and nPKC, effectively blocked TPA-induced YAP/ TAZ dephosphorylation (Figure 1A). These observations suggest a role of PKC in YAP/TAZ activation. Next, we examined whether activation of PKC by physiological stimuli would activate YAP/TAZ. We found that addition of acetylcholine to U251MG cells, which express the Gq/11-coupled muscarinic acetylcholine re- ceptor M3, resulted in significant YAP dephosphorylation (Figure 1C). Inhibition of either M3 receptor by 4-DAMP or PKC by GO6983 suppressed acetylcholine-induced YAP dephosphorylation (Figure 1C), suggesting that YAP activation by acetylcholine is mediated by the M3 recep- tor and PKC. Moreover, Gq/11 knockdown in U251MG cells suppressed YAP dephosphorylation by acetylcho- line (Supplementary information, Figure S1C). Collec- tively, these data indicate that PKC acts downstream of Gq/11-coupled receptors to activate YAP. In the efforts to examine the effect of TPA on YAP/ TAZ phosphorylation in different cell types, we were surprised that TPA induced a dramatic YAP/TAZ phos- phorylation in Swiss3T3 cells (Figure 1D), a response completely opposite to what was observed in HEK293A and several other cell lines (Figure 1A and Supplementa- ry information, Figure S1A and S1B). Notably, TPA-in- duced YAP/TAZ phosphorylation could be blocked by GO6983 (Figure 1D), indicating that PKC activation was also responsible for the increase of YAP/TAZ phosphory- lation in Swiss3T3 cells. Similar effects were observed in MEF cells and the lung cancer A549 cells (Supplementary information, Figure S1D and S1E). Consistent with the increased YAP phosphorylation, TPA treatment promoted a YAP cytoplasmic localization in MEF cells (Figure 1E). Although seemingly paradoxical, the above results are very interesting and show that PKC can either positively or negatively regulate YAP activity in a cell-type-depen- dent manner. The opposing effects of TPA on YAP phosphorylation observed in different cell types prompted us to speculate that different PKC isoforms may exert opposite effects on YAP regulation. To test this, individual cPKC or nPKC isoform was co-transfected with YAP into HEK293A cells and TPA-induced YAP phosphorylation/dephosphoryla-
-
Egf-Induced Erk Activation Promotes Ck2-Mediated Disassociation of Alpha-Catenin from Beta-Catenin and Transactivation of Beta-Catenin
Haitao Ji,Ji Wang,Heinz Nika,David Hawke,Susan Keezer,Qingyuan Ge,Bingliang Fang,Xuexun Fang,Dexing Fang,David W. Litchfield,Kenneth Aldape,Zhimin Lu
DOI: https://doi.org/10.1016/j.molcel.2009.09.034
2009-01-01
Molecules and Cells
Abstract:Increased transcriptional activity of beta-catenin resulting from Wnt/Wingless-dependent or -independent signaling has been detected in many types of human cancer, but the underlying mechanism of Wnt-independent regulation remains unclear. We demonstrate here that EGFR activation results in disruption of the complex of beta-catenin and alpha-catenin, thereby abrogating the inhibitory effect of alpha-catenin on beta-catenin transactivation via CK2alpha-dependent phosphorylation of alpha-catenin at S641. ERK2, which is activated by EGFR signaling, directly binds to CK2alpha via the ERK2 docking groove and phosphorylates CK2alpha primarily at T360/S362, subsequently enhancing CK2alpha activity toward alpha-catenin phosphorylation. In addition, levels of alpha-catenin S641 phosphorylation correlate with levels of ERK1/2 activity in human glioblastoma specimens and with grades of glioma malignancy. This EGFR-ERK-CK2-mediated phosphorylation of alpha-catenin promotes beta-catenin transactivation and tumor cell invasion. These findings highlight the importance of the crosstalk between EGFR and Wnt pathways in tumor development.
-
PKM2 Dephosphorylation by Cdc25A Promotes the Warburg Effect and Tumorigenesis
Ji Liang,Ruixiu Cao,Yajuan Zhang,Yan Xia,Yanhua Zheng,Xinjian Li,Liwei Wang,Weiwei Yang,Zhimin Lu
DOI: https://doi.org/10.1038/ncomms12431
IF: 16.6
2016-01-01
Nature Communications
Abstract:Many types of human tumour cells overexpress the dual-specificity phosphatase Cdc25A. Cdc25A dephosphorylates cyclin-dependent kinase and regulates the cell cycle, but other substrates of Cdc25A and their relevant cellular functions have yet to be identified. We demonstrate here that EGFR activation results in c-Src-mediated Cdc25A phosphorylation at Y59, which interacts with nuclear pyruvate kinase M2 (PKM2). Cdc25A dephosphorylates PKM2 at S37, and promotes PKM2-dependent β-catenin transactivation and c-Myc-upregulated expression of the glycolytic genes GLUT1, PKM2 and LDHA, and of CDC25A; thus, Cdc25A upregulates itself in a positive feedback loop. Cdc25A-mediated PKM2 dephosphorylation promotes the Warburg effect, cell proliferation and brain tumorigenesis. In addition, we identify positive correlations among Cdc25A Y59 phosphorylation, Cdc25A and PKM2 in human glioblastoma specimens. Furthermore, levels of Cdc25A Y59 phosphorylation correlate with grades of glioma malignancy and prognosis. These findings reveal an instrumental function of Cdc25A in controlling cell metabolism, which is essential for EGFR-promoted tumorigenesis.
-
Regulation of Met‐β‐catenin Interaction in Hepatocytes by Tyrosine Phosphorylation
G Zeng,A Bell,A Micsenyi,SPS Monga
DOI: https://doi.org/10.1096/fasebj.20.5.a1090-b
2006-01-01
Abstract:β-Catenin is a key component of the canonical Wnt pathway where it is regulated by serine-threonine phosphorylation. β-Catenin also complexes with E-cadherin or HGF receptor Met, especially at hepatocyte membrane. HGF induces dissociation of Met-β-catenin complex only, secondary to tyrosine phosphorylation of Met & β-catenin followed by nuclear translocation & activation of β-catenin. To further characterize this association, we generated point mutations in various tyrosine residues (86, 142, 604, 654, and 670) in the N- and C-terminal of β-catenin. Based on our analysis we additionally generated 3 double mutants (654 & 670, 654 & 142, 670 & 142). These mutants & wildtype (WT) β-catenin were subcloned into P3XFLAG-CMV-10 vector & transiently transfected into the JM1 rat hepatoma cells. Any redistribution of FLAG/β-catenin was monitored in response to HGF treatment for 30 minutes. Y to E (positive controls) mutations showed spontaneous nuclear localization of FLAG/β-catenin in 86, 142, 654, and 670 only indicating the importance of these residues in β-catenin activation. Y to F mutations showed increased nuclear localization of FLAG/β-catenin in response to HGF only, indicating multiple tyrosine residues might be influenced by HGF/c-Met stimulation. While Y to F double mutants involving 654 & 142, 670 & 142 continued to display nuclear localization of FLAG/β-catenin in response to HGF, 654 & 670 double mutants failed to do so, demonstrating the importance of these tyrosine residues located at β-catenin C-terminal, in its HGF-mediated activation. Thus, Tyr-654 & 670 seems to be crucial in regulating Met-β-catenin interaction in response to HGF in the liver.
-
EGF promotes PKM2 O-GlcNAcylation by stimulating O-GlcNAc transferase phosphorylation at Y976 and their subsequent association
Yang Wang,Hengyao Shu,Jia Liu,Xin Jin,Lihua Wang,Yanzhao Qu,Mingjie Xia,Pinghui Peng,Yunpeng Feng,Min Wei
DOI: https://doi.org/10.1016/j.jbc.2022.102340
IF: 5.485
2022-09-01
Journal of Biological Chemistry
Abstract:Epidermal growth factor (EGF) is one of the most well-characterized growth factors and plays a crucial role in cell proliferation and differentiation. Its receptor EGFR has been extensively explored as a therapeutic target against multiple types of cancers, such as lung cancer and glioblastoma. Recent studies have established a connection between deregulated EGF signaling and metabolic reprogramming, especially rewiring in aerobic glycolysis, which is also known as the Warburg effect and recognized as a hallmark in cancer. Pyruvate kinase M2 (PKM2) is a rate-limiting enzyme controlling the final step of glycolysis and serves as a major regulator of the Warburg effect. We previously showed that PKM2 T405/S406 O-GlcNAcylation, a critical mark important for PKM2 detetramerization and activity, was markedly upregulated by EGF. However, the mechanism by which EGF regulates PKM2 O-GlcNAcylation still remains uncharacterized. Here, we demonstrated that EGF promoted O-GlcNAc transferase (OGT) binding to PKM2 by stimulating OGT Y976 phosphorylation. As a consequence, we found PKM2 O-GlcNAcylation and detetramerization were upregulated, leading to a significant decrease in PKM2 activity. Moreover, distinct from PKM2, we observed that the association of additional phosphotyrosine-binding proteins with OGT was also enhanced when Y976 was phosphorylated. These proteins included STAT1, STAT3, STAT5, PKCδ, and p85, which are reported to be O-GlcNAcylated. Together, we show EGF-dependent Y976 phosphorylation is critical for OGT-PKM2 interaction and propose that this posttranslational modification might be important for substrate selection by OGT.
biochemistry & molecular biology
-
EGFR-Induced and PKCε Monoubiquitylation-Dependent NF-κB Activation Upregulates PKM2 Expression and Promotes Tumorigenesis
Weiwei Yang,Yan Xia,Yu Cao,Yanhua Zheng,Wen Bu,Lin Zhang,M. James You,Mei Yee Koh,Gilbert Cote,Kenneth Aldape,Yi Li,Inder M. Verma,Paul J. Chiao,Zhimin Lu
DOI: https://doi.org/10.1016/j.molcel.2012.09.028
IF: 16
2018-01-01
Molecular Cell
Abstract:Many types of human tumor cells have overexpressed pyruvate kinase M2 (PKM2). However, the mechanism underlying this increased PKM2 expression remains to be defined. We demonstrate here that EGFR activation induces PLCγ1-dependent PKCε monoubiquitylation at Lys321 mediated by RINCK1 ubiquitin ligase. Monoubiquitylated PKCε interacts with a ubiquitin-binding domain in NEMO zinc finger and recruits the cytosolic IKK complex to the plasma membrane, where PKCε phosphorylates IKKβ at Ser177 and activates IKKβ. Activated RelA interacts with HIF1α, which is required for RelA to bind the PKM promoter. PKCε- and NF-κB-dependent PKM2 upregulation is required for EGFR-promoted glycolysis and tumorigenesis. In addition, PKM2 expression correlates with EGFR and IKKβ activity in human glioblastoma specimens and with grade of glioma malignancy. These findings highlight the distinct regulation of NF-κB by EGF, in contrast to TNF-α, and the importance of the metabolic cooperation between the EGFR and NF-κB pathways in PKM2 upregulation and tumorigenesis.
-
Translocalization of enhanced PKM2 protein into the nucleus induced by cancer upregulated gene 2 confers cancer stem cell-like phenotypes
Natpaphan Yawut,Sirichat Kaowinn,Il-Rae Cho,Phatcharaporn Budluang,Seonghye Kim,Suhkmann Kim,So Eun Youn,Sang Seok Koh,Young-Hwa Chung
DOI: https://doi.org/10.5483/BMBRep.2022.55.2.118
Abstract:Increased mRNA levels of cancer upregulated gene (CUG)2 have been detected in many different tumor tissues using Affymetrix microarray. Oncogenic capability of the CUG2 gene has been further reported. However, the mechanism by which CUG2 overexpression promotes cancer stem cell (CSC)-like phenotypes remains unknown. With recent studies showing that pyruvate kinase muscle 2 (PKM2) is overexpressed in clinical tissues from gastric, lung, and cervical cancer patients, we hypothesized that PKM2 might play an important role in CSC-like phenotypes caused by CUG2 overexpression. The present study revealed that PKM2 protein levels and translocation of PKM2 into the nucleus were enhanced in CUG2-overexpressing lung carcinoma A549 and immortalized bronchial BEAS-2B cells than in control cells. Expression levels of c-Myc, CyclinD1, and PKM2 were increased in CUG2-overexpressing cells than in control cells. Furthermore, EGFR and ERK inhibitors as well as suppression of Yap1 and NEK2 expression reduced PKM2 protein levels. Interestingly, knockdown of β-catenin expression failed to reduce PKM2 protein levels. Furthermore, reduction of PKM2 expression with its siRNA hindered CSC-like phenotypes such as faster wound healing, aggressive transwell migration, and increased size/number of sphere formation. The introduction of mutant S37A PKM2-green fluorescence protein (GFP) into cells without ability to move to the nucleus did not confer CSC-like phenotypes, whereas forced expression of wild-type PKM2 promoted such phenotypes. Overall, CUG2-induced increase in the expression of nuclear PKM2 contributes to CSC-like phenotypes by upregulating c-Myc and CyclinD1 as a co-activator. [BMB Reports 2022;55(2): 98-103].
-
PKM2 phosphorylates MLC2 and regulates cytokinesis of tumour cells
Yuhui Jiang,Yugang Wang,Ting Wang,David H. Hawke,Yanhua Zheng,Xinjian Li,Qin Zhou,Sadhan Majumder,Erfei Bi,David X. Liu,Suyun Huang,Zhimin Lu
DOI: https://doi.org/10.1038/ncomms6566
IF: 16.6
2014-01-01
Nature Communications
Abstract:Pyruvate kinase M2 (PKM2) is expressed at high levels during embryonic development and tumour progression and is important for cell growth. However, it is not known whether it directly controls cell division. Here, we found that Aurora B phosphorylates PKM2, but not PKM1, at T45; this phosphorylation is required for PKM2’s localization and interaction with myosin light chain 2 (MLC2) in the contractile ring region of mitotic cells during cytokinesis. PKM2 phosphorylates MLC2 at Y118, which primes the binding of ROCK2 to MLC2 and subsequent ROCK2-dependent MLC2 S15 phosphorylation. PKM2-regulated MLC2 phosphorylation, which is greatly enhanced by EGF stimulation or EGFRvIII, K-Ras G12V and B-Raf V600E mutant expression, plays a pivotal role in cytokinesis, cell proliferation and brain tumour development. These findings underscore the instrumental function of PKM2 in oncogenic EGFR-, K-Ras- and B-Raf-regulated cytokinesis and tumorigenesis.
-
Arf guanine nucleotide‐exchange factors BIG1 and BIG2 regulate β‐catenin signaling via direct interaction and cAMP/PKA pathway (604.1)
Kang Le,Chun‐Chun Li,Joel Moss,Martha Vaughan
DOI: https://doi.org/10.1096/fasebj.28.1_supplement.604.1
2014-04-01
The FASEB Journal
Abstract:BIG1 and BIG2, brefeldin A‐inhibited guanine nucleotide‐exchange factors (GEFs) that activate ADP‐ribosylation factors (Arfs) by accelerating the replacement of bound GDP with GTP, contain A kinase‐anchoring protein (AKAP) sequences that act as scaffolds for multimolecular complexes to facilitate and limit intracellular cAMP signaling in both time and space. β‐Catenin, with its critical roles in cell‐cell adhesion and Wnt signaling, was among HeLa cell proteins co‐immunoprecipitated by antibodies against BIG1 or BIG2. Direct interaction of BIG1 N‐terminal sequence with β‐catenin, found in yeast two‐hybrid studies, was confirmed using in vitro‐synthesized proteins. Depletion of BIG1 and/or BIG2 or over‐expression of a GEF‐inactive mutant, but not wild‐type, BIG1 or BIG2 resulted in β‐catenin accumulation in perinuclear Golgi structures. As association of protein kinase A (PKA) with β‐catenin, BIG1, and BIG2 in HeLa cells, and PKA‐catalyzed phosphorylation of β‐catenin S675 were diminished after depletion of BIG1 or BIG2, we infer that function of the AKAP sequences in BIG1 and/or BIG2 may be required for PKA‐catalyzed β‐catenin modification. In addition, transcriptional activity of β‐catenin was lower in HeLa cells after BIG1 and/or BIG2 depletion, further consistent with their roles in regulating β‐catenin action via S675 phosphorylation.
cell biology,biochemistry & molecular biology,biology
-
CGK062, a small chemical molecule, inhibits cancer upregulated gene 2‑induced oncogenesis through NEK2 and β‑catenin
Sirichat Kaowinn,Sangtaek Oh,Jeong Moon,Ah Young Yoo,Ho Young Kang,Mi Rim Lee,Ji Eun Kim,Dae Youn Hwang,So Eun Youn,Sang Seok Koh,Young-Hwa Chung
DOI: https://doi.org/10.3892/ijo.2019.4724
Abstract:The mechanisms through which cancer‑upregulated gene 2 (CUG2), a novel oncogene, affects Wnt/β‑catenin signaling, essential for tumorigenesis, are unclear. In this study, we aimed to elucidate some of these mechanisms in A549 lung cancer cells. Under the overexpression of CUG2, the protein levels and activity of β‑catenin were evaluated by western blot analysis and luciferase assay. To examine a biological consequence of β‑catenin under CUG2 overexpression, cell migration, invasion and sphere formation assay were performed. The upregulation of β‑catenin induced by CUG2 overexpression was also accessed by xenotransplantation in mice. We first found that CUG2 overexpression increased β‑catenin expression and activity. The suppression of β‑catenin decreased cancer stem cell (CSC)‑like phenotypes, indicating that β‑catenin is involved in CUG2‑mediated CSC‑like phenotypes. Notably, CUG2 overexpression increased the phosphorylation of β‑catenin at Ser33/Ser37, which is known to recruit E3 ligase for β‑catenin degradation. Moreover, CUG2 interacted with and enhanced the expression and kinase activity of never in mitosis gene A‑related kinase 2 (NEK2). Recombinant NEK2 phosphorylated β‑catenin at Ser33/Ser37, while NEK2 knockdown decreased the phosphorylation of β‑catenin, suggesting that NEK2 is involved in the phosphorylation of β‑catenin at Ser33/Ser37. Treatment with CGK062, a small chemical molecule, which promotes the phosphorylation of β‑catenin at Ser33/Ser37 through protein kinase C (PKC)α to induce its degradation, reduced β‑catenin levels and inhibited the CUG2‑induced features of malignant tumors, including increased cell migration, invasion and sphere formation. Furthermore, CGK062 treatment suppressed CUG2‑mediated tumor formation in nude mice. Taken together, the findings of this study suggest that CUG2 enhances the phosphorylation of β‑catenin at Ser33/Ser37 by activating NEK2, thus stabilizing β‑catenin. CGK062 may thus have potential for use as a therapeutic drug against CUG2‑overexpressing lung cancer cells.
-
Tyrosine Residues 654 and 670 in Β-Catenin Are Crucial in Regulation of Met–β-catenin Interactions
Gang Zeng,Udayan Apte,Amanda Micsenyi,Aaron Bell,Satdarshan P. S. Monga
DOI: https://doi.org/10.1016/j.yexcr.2006.08.003
IF: 4.145
2006-01-01
Experimental Cell Research
Abstract:β-catenin, a key component of the canonical Wnt pathway, is also regulated by tyrosine phosphorylation that regulates its association to E-cadherin. Previously, we reported its association with the hepatocyte growth factor (HGF) receptor Met at the membrane. HGF induced Met–β-catenin dissociation and nuclear translocation of β-catenin, which was tyrosine-phosphorylation-dependent. Here, we further investigate the Met–β-catenin interaction by selectively mutating several tyrosine residues, alone or in combination, in β-catenin. The mutants were subcloned into FLAG-CMV vector and stably transfected into rat hepatoma cells, which were treated with HGF. All single or double-mutant-transfected cells continued to show HGF-induced nuclear translocation of FLAG-β-catenin except the mutations affecting 654 and 670 simultaneously (Y654/670F), which coincided with the lack of formation of β-catenin–TCF complex and DNA synthesis, in response to the HGF treatment. In addition, the Y654/670F-transfected cells also showed no phosphorylation of β-catenin or dissociation from Met in response to HGF. Thus, intact 654 and 670 tyrosine residues in β-catenin are crucial in HGF-mediated β-catenin translocation, activation and mitogenesis.
-
PKM2 Regulates Chromosome Segregation and Mitosis Progression of Tumor Cells.
Yuhui Jiang,Xinjian Li,Weiwei Yang,David H Hawke,Yanhua Zheng,Yan Xia,Kenneth Aldape,Chongyang Wei,Fang Guo,Yan Chen,Zhimin Lu
DOI: https://doi.org/10.1016/j.molcel.2013.11.001
IF: 16
2013-01-01
Molecular Cell
Abstract:Tumor-specific pyruvate kinase M2 (PKM2) is instrumental in both aerobic glycolysis and gene transcription. PKM2 regulates G1-S phase transition by controlling cyclin D1 expression. However, it is not known whether PKM2 directly controls cell-cycle progression. We show here that PKM2, but not PKM1, binds to the spindle checkpoint protein Bub3 during mitosis and phosphorylates Bub3 at Y207. This phosphorylation is required for Bub3-Bub1 complex recruitment to kinetochores, where it interacts with Blinkin and is essential for correct kinetochore-microtubule attachment, mitotic/spindle-assembly checkpoint, accurate chromosome segregation, cell survival and proliferation, and active EGF receptor-induced brain tumorigenesis. In addition, the level of Bub3 Y207 phosphorylation correlated with histone H3-S10 phosphorylation in human glioblastoma specimens and with glioblastoma prognosis. These findings highlight the role of PKM2 as a protein kinase controlling the fidelity of chromosome segregation, cell-cycle progression, and tumorigenesis.
-
Glycogen synthase kinase-3 is a negative regulator of extracellular signal-regulated kinase
Q Wang,Y Zhou,X Wang,B M Evers
DOI: https://doi.org/10.1038/sj.onc.1209004
IF: 8.756
2005-11-07
Oncogene
Abstract:Glycogen-synthase kinase-3 (GSK-3) and extracellular signal-regulated kinase (ERK) are critical downstream signaling proteins for the PI3-kinase/Akt and Ras/Raf/MEK-1 pathway, respectively, and regulate diverse cellular processes including embryonic development, cell differentiation and apoptosis. Here, we show that inhibition of GSK-3 using GSK-3 inhibitors or RNA interference (RNAi) significantly induced the phosphorylation of ERK1/2 in human colon cancer cell lines HT29 and Caco-2. Pretreatment with the PKCδ-selective inhibitor rottlerin or transfection with PKCδ siRNA attenuated the phosphorylation of ERK1/2 induced by the GSK-3 inhibitor SB-216763 and, furthermore, treatment with SB-216763 or transfection with GSK-3α and GSK-3β siRNA increased PKCδ activity, thus identifying a role for PKCδ in the induction of ERK1/2 phosphorylation by GSK-3 inhibition. Treatment with SB-216763 increased expression of cyclooxygenase-2 (COX-2) and IL-8, which are downstream targets of ERK1/2 activation; this induction was abolished by MEK/ERK inhibition, suggesting GSK-3 inhibition induced COX-2 and IL-8 through ERK1/2 activation. The transcriptional induction of COX-2 and IL-8 by GSK-3 inhibition was further demonstrated by the increased COX-2 and IL-8 promoter activity after SB-216763 treatment or transfection with GSK-3α or GSK-3β siRNA. Importantly, our findings identify GSK-3, acting through PKCδ, as a negative regulator of ERK1/2, thus revealing a novel crosstalk mechanism between these critical signaling pathways.
oncology,genetics & heredity,biochemistry & molecular biology,cell biology
-
PKM2 Phosphorylates Histone H3 and Promotes Gene Transcription and Tumorigenesis
Weiwei Yang,Yan Xia,David Hawke,Xinjian Li,Ji Liang,Dongming Xing,Kenneth Aldape,Tony Hunter,W.K. Alfred Yung,Zhimin Lu
DOI: https://doi.org/10.1016/j.cell.2012.07.018
IF: 64.5
2012-08-01
Cell
Abstract:Tumor-specific pyruvate kinase M2 (PKM2) is essential for the Warburg effect. In addition to its well-established role in aerobic glycolysis, PKM2 directly regulates gene transcription. However, the mechanism underlying this nonmetabolic function of PKM2 remains elusive. We show here that PKM2 directly binds to histone H3 and phosphorylates histone H3 at T11 upon EGF receptor activation. This phosphorylation is required for the dissociation of HDAC3 from the CCND1 and MYC promoter regions and subsequent acetylation of histone H3 at K9. PKM2-dependent histone H3 modifications are instrumental in EGF-induced expression of cyclin D1 and c-Myc, tumor cell proliferation, cell-cycle progression, and brain tumorigenesis. In addition, levels of histone H3 T11 phosphorylation correlate with nuclear PKM2 expression levels, glioma malignancy grades, and prognosis. These findings highlight the role of PKM2 as a protein kinase in its nonmetabolic functions of histone modification, which is essential for its epigenetic regulation of gene expression and tumorigenesis.
cell biology,biochemistry & molecular biology
-
KAT2A succinyltransferase activity-mediated 14-3-3ζ upregulation promotes β-catenin stabilization-dependent glycolysis and proliferation of pancreatic carcinoma cells.
Yingying Tong,Dong Guo,Dong Yan,Chunmin Ma,Fei Shao,Yugang Wang,Shudi Luo,Liming Lin,Jingjing Tao,Yuhui Jiang,Zhimin Lu,Dongming Xing
DOI: https://doi.org/10.1016/j.canlet.2019.09.015
IF: 9.756
2019-01-01
Cancer Letters
Abstract:Frequently occurring histone lysine succinylation is a newly identified histone modification that can be regulated by KAT2A histone succinyltransferase, which is also a histone acetyltransferase. KAT2A histone succinyltransferase activity is important for tumorigenesis; however, the mechanism underlying this tumor-promoting effect remains elusive. Here we demonstrate that KAT2A is highly expressed in human pancreatic ductal adenocarcinoma (PDAC) specimens and positively correlated with advanced stages of PDAC and short patients' survival. In addition, KAT2A expression in PDAC specimens is correlated with 14-3-3ζ expression, and KAT2A regulates H3K79 succinylation in the promoter region of YWHAZ (encoding for 14-3-3ζ) to promote YWHAZ mRNA and 14-3-3ζ expression, thereby preventing β-catenin degradation. Expression of succinyltransferase activity-defective KAT2A Y645A reduces H3K79 succinylation and 14-3-3ζ expression, leading to decreased β-catenin stability and subsequently decreased expression of cyclin D1, c-Myc, GLUT1, and LDHA. KAT2A-mediated 14-3-3ζ and β-catenin expression promotes glycolysis, cell proliferation, and migration and invasion of PDAC cells with epithelial-to-mesenchymal transition. These findings reveal a novel and instrumental role of KAT2A-mediated histone succinylation in regulation of gene expression and β-catenin stability to promote tumor cell proliferation and invasion.
-
PKM2 controls the translation of TFE3 to maintain the integrity of the Golgi apparatus for the survival of HeLa and ME‐180 cervical cancer cells
Yuki Taguchi,Kengo Ito,Fumi Kano
DOI: https://doi.org/10.1111/febs.16740
2023-01-29
FEBS Journal
Abstract:The M2 isoform of pyruvate kinase (PKM2) is abundantly expressed in various cancer cells and associated with tumorigenesis, tumor proliferation and tumor progression. However, the role of PKM2 in these oncological processes is not fully understood. In the present study, we depleted PKM2 expression using RNA interference (RNAi), which induced apoptotic cell death and was accompanied by the downregulation of GM130, giantin, and p115 in HeLa and ME‐180 cervical cancer cells. The decreased expression of these proteins caused structural and functional disturbances in the Golgi apparatus, which manifested as the dispersion of the Golgi apparatus and delayed anterograde trafficking from the ER to the Golgi. The transcription factor, TFE3, which functions in the Golgi stress response, was responsible for the expression of GM130, giantin, and p115 that maintained the integrity of the organelle under normal growth conditions. In PKM2‐knockdown cells, the translation of TFE3 was markedly reduced. Knockdown of TFE3 by RNAi resulted in the downregulation of GM130, giantin, and p115, dispersion of the Golgi apparatus, and apoptotic cell death, similar to those observed following PKM2 knockdown. Conversely, the exogenous expression of TFE3 in PKM2 knockdown cells partially mitigated the aforementioned effects. We also demonstrated that PKM2 bound to the 5′ UTR on TFE3 mRNA and promoted translation. This study is the first to identify a new function for PKM2, which activates the basal Golgi stress response to maintain the integrity of the Golgi apparatus through the translation of TFE3 and promote cancer cell survival.
biochemistry & molecular biology
-
The c-Cbl Ubiquitin Ligase Regulates Nuclear β-Catenin and Angiogenesis by Its Tyrosine Phosphorylation Mediated through the Wnt Signaling Pathway*
Hui Feng,Moshe Shashar,J. Francis,R. Meyer,N. Rahimi,V. Chitalia,S. Shivanna,Itrat Harrold,E. Edelman,Qing Zhao,Chrystelle Kiang
DOI: https://doi.org/10.1074/jbc.M114.616623
IF: 5.485
2015-03-17
Journal of Biological Chemistry
Abstract:Background: β-Catenin is an important mediator of angiogenesis; however, its molecular regulation is poorly understood. Results: Wnt promotes phosphorylation of c-Cbl on Tyr-731, enhancing its dimerization and nuclear translocation to degrade nuclear β-catenin. Conclusion: Wnt-mediated c-Cbl phosphorylation regulates nuclear β-catenin. Significance: Targeting c-Cbl E3 ligase activity could destabilize β-catenin and inhibit pathological angiogenesis. Wnt signaling plays important roles in both the tumor-induced angiogenesis and tumorigenesis through the transcriptionally active nuclear β-catenin. Recently, c-Cbl was identified as a unique E3 ubiquitin ligase targeting the active nuclear β-catenin. However, little is known about the molecular mechanisms by which c-Cbl regulates ubiquitination and degradation of active β-catenin. Here, we demonstrate that Wnt activation promotes the phosphorylation of c-Cbl at tyrosine 731(Tyr-731), which increases c-Cbl dimerization and binding to β-catenin. Tyr-731 phosphorylation and dimerization mediate c-Cbl nuclear translocation and lead to the degradation of nuclearly active β-catenin in the Wnt-on phase. c-Cbl activation also inhibits expression of the pro-angiogenic Wnt targets, IL-8 and VEGF. Phospho-Tyr-731-inactive mutant c-Cbl (Y731F) enhances and phosphomimetic mutant c-Cbl (Y731E) suppresses angiogenesis in zebrafish. Taken together, we have identified a novel mechanism for the regulation of active nuclear β-catenin by c-Cbl and its critical role in angiogenesis. This mechanism can be further explored to modulate both the pathological angiogenesis and the tumorigenesis.
Biology,Medicine,Chemistry