The ErbB family of receptor tyrosine kinases comprises four members: epidermal growth factor receptor (EGFR)/ErbB1, HER2/ErbB2, ErbB3 and ErbB4, and plays roles in signal transduction at the plasma membrane upon ligand stimulation. Stimulation with neuregulin-1 (NRG-1) cleaves ErbB4 and releases the ErbB4 intracellular domain (4ICD) that translocates into the nucleus to control gene expression. However, little is known about the regulation of 4ICD nuclear signaling through tyrosine phosphorylation. We show here that 4ICD nuclear signaling is antagonized by EGF-induced c-Src activation through EGFR. Generation of 4ICD by NRG-1 leads to increased levels of trimethylated histone H3 on lysine 9 (H3K9me3) in a manner dependent on the nuclear accumulation of 4ICD and its tyrosine kinase activity. Once EGF activates c-Src downstream of EGFR concomitantly with NRG-1-induced ErbB4 activation, c-Src associates with phospho-Tyr950 and phospho-Tyr1056 on 4ICD, thereby decreasing nuclear accumulation of 4ICD and inhibiting an increase of H3K9me3 levels. Moreover, 4ICD-induced transcriptional repression of the human telomerase reverse transcriptase (hTERT) is inhibited by EGF–EGFR–Src signaling. Thus, our findings reveal c-Src-mediated inhibitory regulation of ErbB4 nuclear signaling upon EGFR activation.

Protein-tyrosine kinases are found in multicellular organisms and have important roles in a variety of the intracellular signal transduction pathways, which are critical in the control of cellular processes, such as cell proliferation, differentiation, gene expression, cell adhesion, and metabolic changes (Hubbard and Till, 2000). Receptor-type tyrosine kinases, a subclass of transmembrane-spanning receptor, transmit signals across the plasma membrane from extracellular milieus to the inside of cells (Ullrich and Schlessinger, 1990). On the other hand, the function of non-receptor-type tyrosine kinases depends upon their intracellular localization (Neet and Hunter, 1996). Although tyrosine phosphorylation is mainly associated with cytoplasmic events, some types of tyrosine kinases and phosphatases localize within the nucleus and nuclear tyrosine phosphorylation may play a role in nuclear events (Cans et al., 2000; Moorhead et al., 2007). We have shown that Lyn, a member of non-receptor-type Src-family tyrosine kinases (SFKs), is imported into and rapidly exported from the nucleus (Ikeda et al., 2008), and nuclear tyrosine phosphorylation by SFKs, Csk and c-Abl has a role in changes of chromatin structure and histone modification (Yamaguchi et al., 2001; Nakayama and Yamaguchi, 2005; Takahashi et al., 2009; Aoyama et al., 2011).

The ErbB receptor tyrosine kinases comprise the four members, epidermal growth factor receptor (EGFR)/ErbB1, HER2/ErbB2, ErbB3 and ErbB4. ErbB-family members are known to play critical roles in regulating cell proliferation, differentiation, apoptosis and migration (Yarden and Sliwkowski, 2001). Each of the receptors is a type I transmembrane protein and is composed of: (1) an extracellular domain that provides a ligand-binding site; (2) a single transmembrane domain; and (3) an intracellular domain that encodes a tyrosine kinase catalytic domain and a C-terminal region. The cytoplasmic domain contains multiple tyrosine residues (Schulze et al., 2005), and the receptor function is regulated by tyrosine phosphorylation. For instance, EGFR signaling is positively regulated by SFKs through tyrosine phosphorylation (Dittmann et al., 2008; Iida et al., 2013).

ErbB4 is known to have a unique signaling feature among the ErbB-family members. Stimulation with NRG-1, a ligand of ErbB4, not only activates downstream signaling molecules close to or on the cytoplasmic face of the plasma membrane but also cleaves the ErbB4 extracellular domain and the intracellular domain by TNF-α-converting enzyme (TACE) and γ-secretase, respectively (Rio et al., 2000; Ni et al., 2001). Cleavage by TACE removes the extracellular domain and leaves the membrane-associated 80-kDa ErbB4 fragment (m80 ErbB4). The m80 ErbB4 fragment is further cleaved by γ-secretase to release the soluble 80-kDa ErbB4 fragment, termed the ErbB4 intracellular domain (4ICD), into the cytoplasm. Then, 4ICD acts as a non-receptor-type tyrosine kinase and can shuttle between the cytoplasm and the nucleus. It is evident that 4ICD can signal directly to the nucleus to regulate transcription (Ni et al., 2001; Carpenter, 2003; Williams et al., 2004; Sardi et al., 2006). Despite the existence of multiple tyrosine phosphorylation sites on 4ICD (Kaushansky et al., 2008), the regulation of 4ICD nuclear signaling through tyrosine phosphorylation is poorly understood.

In this study, we investigate ErbB4-mediated nuclear signaling upon stimulation with NRG-1. Generation of 4ICD increases the levels of H3K9me3, which is antagonized by EGF-activated EGFR, another member of the ErbB family. We further identify activated c-Src downstream of EGFR as a negative regulator of nuclear ErbB4 signaling. Our findings demonstrate that tyrosine phosphorylation of 4ICD by EGFR-activated c-Src plays an inhibitory role in nuclear ErbB4 signaling.

ErbB4-induced increase of H3K9me3 following NRG-1 stimulation

To examine the role of ErbB4 signaling in the nucleus, T47D breast cancer cells were stimulated with NRG-1. We observed that endogenous ErbB4 was activated at 5 min and subsequent 4ICD release was peaked at 30 min after NRG-1 stimulation (Fig. 1A), consistent with previous observations (Ni et al., 2001). NRG-1 stimulation was found to decrease high-intensity areas (red and yellow) and increase low-intensity areas (green and blue) of DNA staining, leading to an increase in nucleus size (Fig. 1B). Upon NRG-1 stimulation, treatment with AG1478, an inhibitor of EGFR and ErbB4 (Egeblad et al., 2001), blocked NRG-1-induced 4ICD release and an increase in nuclear size (supplementary material Fig. S1A,B). Since NRG-1 stimulation did not induce BrdU incorporation (supplementary material Fig. S1C), the increase in nuclear size may be not due to DNA synthesis but rather chromatin structural changes.

Fig. 1.

Opposing effects of NRG-1 and EGF stimulations on H3K9me3 levels. (A) T47D cells were treated with NRG-1 for the indicated times. Cell lysates were analyzed by western blotting using anti-pTyr, anti-ErbB4 and anti-Lamin A/C (loading control) antibodies. Size markers in kilodaltons (kDa) are shown on the left. (B) T47D cells were left untreated (none) or treated with NRG-1 for 30 min and stained with propidium iodide (PI) for DNA. Two-dimensional isometric intensity profiles are shown in the lower panels. Spectrum of PI fluorescence intensity from 0 to 255 represents: background (black), low (blue), medium (green), high (yellow) and very high (red). Scale bars: 10 µm. (C) Quantification of histone modifications. T47D cells were left untreated (none) or treated with NRG-1 or EGF for 30 min, then stained with anti-H3K9me3, anti-H3K4me3 and anti-H4K16ac antibodies (left panels). Scale bars: 10 µm. Fluorescence intensities in individual cells are plotted on the right. Numbers in parentheses indicate mean values, and bars represent means ± s.d. from a representative experiment (n = 40∼63 cells). Asterisks indicate a significant difference (***P<0.001; NS, not significant) calculated by Student's t-test.

Fig. 1.

Opposing effects of NRG-1 and EGF stimulations on H3K9me3 levels. (A) T47D cells were treated with NRG-1 for the indicated times. Cell lysates were analyzed by western blotting using anti-pTyr, anti-ErbB4 and anti-Lamin A/C (loading control) antibodies. Size markers in kilodaltons (kDa) are shown on the left. (B) T47D cells were left untreated (none) or treated with NRG-1 for 30 min and stained with propidium iodide (PI) for DNA. Two-dimensional isometric intensity profiles are shown in the lower panels. Spectrum of PI fluorescence intensity from 0 to 255 represents: background (black), low (blue), medium (green), high (yellow) and very high (red). Scale bars: 10 µm. (C) Quantification of histone modifications. T47D cells were left untreated (none) or treated with NRG-1 or EGF for 30 min, then stained with anti-H3K9me3, anti-H3K4me3 and anti-H4K16ac antibodies (left panels). Scale bars: 10 µm. Fluorescence intensities in individual cells are plotted on the right. Numbers in parentheses indicate mean values, and bars represent means ± s.d. from a representative experiment (n = 40∼63 cells). Asterisks indicate a significant difference (***P<0.001; NS, not significant) calculated by Student's t-test.

We next examined histone modifications, because the regulation of chromatin structure is known to involve changes of histone modifications, such as methylation and acetylation (Jenuwein and Allis, 2001; Aoyama et al., 2011). Intriguingly, unlike trimethylated histone H3 on lysine 4 (H3K4me3) and acetylated histone H4 on lysine 16 (H4K16ac), the levels of H3K9me3 in T47D cells were increased by NRG-1 stimulation of ErbB4 but decreased by EGF stimulation of EGFR (Fig. 1C; supplementary material Fig. S1D). NRG-1 stimulation was also found to increase H3K9me3 levels in ErbB4-transfected COS-1 cells (supplementary material Fig. S2A). These results suggest that ErbB4 and EGFR, both of which are members of the ErbB-family, have opposing effects on H3K9me3.

Role of nuclear accumulation and the kinase activity of 4ICD in NRG-1-induced increase of H3K9me3

4ICD has one functional nuclear localization signal (NLS) and three putative nuclear export signals, and can shuttle between the cytoplasm and the nucleus (Ni et al., 2001; Williams et al., 2004). We asked whether an ErbB4-mediated increase of H3K9me3 involved the release of 4ICD and its nuclear translocation. When treatment with DAPT, a γ-secretase inhibitor, prevented γ-secretase processing of ErbB4 and accumulated the membrane-associated 80-kDa ErbB4 fragment (m80 ErbB4; Fig. 2A) (Määttä et al., 2006; Sardi et al., 2006; Paatero et al., 2012), we examined the effect of 4ICD generation on H3K9me3 levels. Fig. 2B shows that DAPT treatment blocked the NRG-1-induced increase of H3K9me3 levels, suggesting that 4ICD release has a critical role in NRG-1-induced increase of H3K9me3 levels. Next, to examine whether nuclear translocation of 4ICD increased H3K9me3 levels, we constructed NLS-4ICD by linking an additional NLS to the N-terminus of 4ICD (Fig. 2C). When 4ICD and NLS-4ICD were expressed in COS-1 cells, NLS-4ICD was mainly localized in the nucleus and the levels of H3K9me3 were much higher in cells expressing NLS-4ICD than those expressing 4ICD (Fig. 2D). Furthermore, to examine the effect of the kinase activity on H3K9me3, we compared NLS-4ICD with the kinase-dead mutant NLS-4ICD(KD) and an irrelevant NLS-tagged Src tyrosine kinase (NLS-Src; Fig. 2C). Although similar levels of nuclear localization were observed among NLS-4ICD, NLS-4ICD(KD), and NLS-Src (supplementary material Fig. S2B; Fig. 2F), the tyrosine-phosphorylated band pattern induced by NLS-4ICD was somewhat different from that induced by NLS-Src (Fig. 2E). Notably, H3K9me3 levels were much higher in cells expressing NLS-4ICD than those expressing NLS-4ICD(KD) or NLS-Src (Fig. 2F,G). In contrast to H3K9me3, H3K4me3 levels in cells expressing NLS-4ICD were comparable to those in cells expressing NLS-4ICD(KD) or NLS-Src (Fig. 2G). These results suggest that nuclear translocation of 4ICD is critical for an increase of H3K9me3 levels in a manner dependent on 4ICD kinase activity.

Fig. 2.

Role of nuclear 4ICD in NRG-1-induced increase of H3K9me3. (A) T47D cells were left untreated (none) or pretreated with 1 or 25 µM DAPT for 30 min, and stimulated with NGR-1 in the presence or absence of 1 or 25 µM DAPT for 30 min. Cell lysates were analyzed by western blotting, using anti-ErbB4 and anti-Lamin A/C antibodies. (B) T47D cells pretreated with AG1478 or 1 µM DAPT for 30 min were stimulated with NRG-1 for 30 min as described in A, and then stained with anti-H3K9me3 antibody. Fluorescence intensities of H3K9me3 immunostaining were measured as described in Fig. 1C. Scale bars: 10 µm. (C) Schematic representations of the constructs used in this study. The extracellular domain (ECD), transmembrane domain (TM), intracellular domain (ICD), kinase domain, FLAG epitope (FLAG), hemagglutinin-epitope (HA), nuclear localization signal (NLS), and Src homology (SH) domain are shown. KD, kinase-dead (K752R). (D) COS-1 cells transfected with vector alone, 4ICD, or NLS-4ICD were cultured for 48 hours, and stained with anti-ErbB4 (expressed proteins) and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm. (E–G) COS-1 cells transfected with the indicated construct were cultured for 48 hours. Cell lysates were analyzed by western blotting (E) using anti-pTyr, anti-ErbB4, anti-HA and anti-actin antibodies. An arrowhead indicates a nonspecific band. Cells were immunostained with anti-H3K9me3 and anti-HA (expressed proteins) antibodies and TOPRO-3 (F), and fluorescence intensities of H3K9me3 and H3K4me3 were measured as described in Fig. 1C (G). Scale bars: 10 µm. Asterisks indicate significant differences (**P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

Fig. 2.

Role of nuclear 4ICD in NRG-1-induced increase of H3K9me3. (A) T47D cells were left untreated (none) or pretreated with 1 or 25 µM DAPT for 30 min, and stimulated with NGR-1 in the presence or absence of 1 or 25 µM DAPT for 30 min. Cell lysates were analyzed by western blotting, using anti-ErbB4 and anti-Lamin A/C antibodies. (B) T47D cells pretreated with AG1478 or 1 µM DAPT for 30 min were stimulated with NRG-1 for 30 min as described in A, and then stained with anti-H3K9me3 antibody. Fluorescence intensities of H3K9me3 immunostaining were measured as described in Fig. 1C. Scale bars: 10 µm. (C) Schematic representations of the constructs used in this study. The extracellular domain (ECD), transmembrane domain (TM), intracellular domain (ICD), kinase domain, FLAG epitope (FLAG), hemagglutinin-epitope (HA), nuclear localization signal (NLS), and Src homology (SH) domain are shown. KD, kinase-dead (K752R). (D) COS-1 cells transfected with vector alone, 4ICD, or NLS-4ICD were cultured for 48 hours, and stained with anti-ErbB4 (expressed proteins) and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm. (E–G) COS-1 cells transfected with the indicated construct were cultured for 48 hours. Cell lysates were analyzed by western blotting (E) using anti-pTyr, anti-ErbB4, anti-HA and anti-actin antibodies. An arrowhead indicates a nonspecific band. Cells were immunostained with anti-H3K9me3 and anti-HA (expressed proteins) antibodies and TOPRO-3 (F), and fluorescence intensities of H3K9me3 and H3K4me3 were measured as described in Fig. 1C (G). Scale bars: 10 µm. Asterisks indicate significant differences (**P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

Inhibition of NRG-1-induced increase of H3K9me3 by EGFR-activated Src

ErbB4 activation decreases cell proliferation and induces differentiation, whereas the other ErbB-family members are involved in cell proliferation and survival (Yarden and Sliwkowski, 2001; Sardi et al., 2006). Given that NRG-1 and EGF bind to ErbB4 and EGFR, respectively (Linggi and Carpenter, 2006), expression levels of NRG-1 and EGF are well controlled in developing mammary glands (Schroeder and Lee, 1998). We thus hypothesized that H3K9me3 levels may be controlled by signal balance between ErbB4 and EGFR. To test this hypothesis, cells were co-stimulated with NRG-1 plus EGF. As a result, NRG-1-induced increase of H3K9me3 levels were decreased by concomitant stimulation with EGF (Fig. 3A). In addition, inhibition of cell proliferation by NRG-1 was substantially rescued by EGF stimulation (Fig. 3B). However, the effect of EGF on NRG-1-repressed cell proliferation was diminished after 4 days of stimulation (Fig. 3B), because EGF stimulation decreased the level of EGFR expression in a 4-day stimulation period (supplementary material Fig. S3) (Dikic and Giordano, 2003). Furthermore, concomitant stimulation with EGF and NRG-1 was incapable of inhibiting NRG-1-induced ErbB4 activation and 4ICD release (Fig. 3C). Taken together, these results suggest that EGFR activation blocks intracellular ErbB4 signaling toward an increase of H3K9me3 and growth inhibition.

Fig. 3.

Inhibition of NRG-1-induced increase of H3K9me3 by EGF-induced Src activation. (A) T47D cells were left untreated (none) or treated with NRG-1 or NRG-1 plus EGF for 30 min, and fluorescence intensities of H3K9me3 immunostaining were measured as described in Fig. 1C. Scale bars: 10 µm. (B) T47D cells were cultured in Iscove's modified Dulbecco's medium supplemented with 5% fetal bovine serum and the number of cells was counted (initial cell number). Cells were then left untreated (none) or treated with NRG-1, NRG-1 plus EGF, or EGF for the indicated periods in low serum conditions (0.05% bovine serum) with a medium change on the second day, and the number of cells was counted (top). The percentage increase in the number of cells was then determined (bottom). Values are means ± s.d., n = 3 (2 days) or n = 6 (4 days) independent experiments. (C) T47D cells were treated as described in A, and analyzed by western blotting using anti-ErbB4, anti-pTyr, anti-pERK1/2, anti-ERK2 and anti-Lamin A/C antibodies. (D) T47D cells were left untreated (none) or treated with NRG-1 or NRG-1 plus EGF in the presence of DMSO or PP2 for 30 min, and fluorescence intensities of H3K9me3 were measured as described in Fig. 1C. Scale bars: 10 µm. (E) (Top) SYF or SYF/c-Src cells transfected with ErbB4 were left untreated (none) or treated with NRG-1 for 30 min and stained with anti-ErbB4 and anti-H3K9me3 antibodies. (Middle) Fluorescence intensities of ErbB4 staining were measured (see supplementary material Fig. S5B), and then H3K9me3 levels were quantified in cells exhibiting similar levels of ErbB4 expression, as described in Fig. 1C. (Bottom) The levels of nuclear localization of ErbB4 were assessed by measuring a ratio of fluorescence intensity of anti-ErbB4 staining in the nucleus to that in the corresponding whole cell (nucleus/whole cell). The plot represents the level of nuclear localization of ErbB4 in each cell, and the bars show the means ± s.d., n = 4 independent experiments. N, nucleus; whole, whole cell. Scale bars: 10 µm. (F) COS-1 cells transfected with 4ICD plus vector or 4ICD plus c-Src-HA were cultured in the presence or absence of SU6656 for 24 hours, and stained with anti-ErbB4 and anti-HA (see supplementary material Fig. S5C) antibodies. The areas of the nuclei are marked by dotted lines. N, nucleus; Cyto, cytoplasm. The percentage of cells exhibiting N≥Cyto localization of 4ICD was quantified (right). Values are means ± s.d., n = 3 independent experiments. Scale bars: 10 µm. Asterisks indicate significant differences (**P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

Fig. 3.

Inhibition of NRG-1-induced increase of H3K9me3 by EGF-induced Src activation. (A) T47D cells were left untreated (none) or treated with NRG-1 or NRG-1 plus EGF for 30 min, and fluorescence intensities of H3K9me3 immunostaining were measured as described in Fig. 1C. Scale bars: 10 µm. (B) T47D cells were cultured in Iscove's modified Dulbecco's medium supplemented with 5% fetal bovine serum and the number of cells was counted (initial cell number). Cells were then left untreated (none) or treated with NRG-1, NRG-1 plus EGF, or EGF for the indicated periods in low serum conditions (0.05% bovine serum) with a medium change on the second day, and the number of cells was counted (top). The percentage increase in the number of cells was then determined (bottom). Values are means ± s.d., n = 3 (2 days) or n = 6 (4 days) independent experiments. (C) T47D cells were treated as described in A, and analyzed by western blotting using anti-ErbB4, anti-pTyr, anti-pERK1/2, anti-ERK2 and anti-Lamin A/C antibodies. (D) T47D cells were left untreated (none) or treated with NRG-1 or NRG-1 plus EGF in the presence of DMSO or PP2 for 30 min, and fluorescence intensities of H3K9me3 were measured as described in Fig. 1C. Scale bars: 10 µm. (E) (Top) SYF or SYF/c-Src cells transfected with ErbB4 were left untreated (none) or treated with NRG-1 for 30 min and stained with anti-ErbB4 and anti-H3K9me3 antibodies. (Middle) Fluorescence intensities of ErbB4 staining were measured (see supplementary material Fig. S5B), and then H3K9me3 levels were quantified in cells exhibiting similar levels of ErbB4 expression, as described in Fig. 1C. (Bottom) The levels of nuclear localization of ErbB4 were assessed by measuring a ratio of fluorescence intensity of anti-ErbB4 staining in the nucleus to that in the corresponding whole cell (nucleus/whole cell). The plot represents the level of nuclear localization of ErbB4 in each cell, and the bars show the means ± s.d., n = 4 independent experiments. N, nucleus; whole, whole cell. Scale bars: 10 µm. (F) COS-1 cells transfected with 4ICD plus vector or 4ICD plus c-Src-HA were cultured in the presence or absence of SU6656 for 24 hours, and stained with anti-ErbB4 and anti-HA (see supplementary material Fig. S5C) antibodies. The areas of the nuclei are marked by dotted lines. N, nucleus; Cyto, cytoplasm. The percentage of cells exhibiting N≥Cyto localization of 4ICD was quantified (right). Values are means ± s.d., n = 3 independent experiments. Scale bars: 10 µm. Asterisks indicate significant differences (**P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

It is known that the binding of EGF to EGFR activates SFKs, ERK1/2, and PI3K (supplementary material Fig. S1D; Yarden and Sliwkowski, 2001). To ask whether activation of SFKs, ERK1/2, or PI3K is required for EGFR-mediated inhibition of ErbB4 signaling, we treated cells with PP2 (SFK inhibitor), U0126 (MEK1/2 inhibitor), and wortmannin (PI3K inhibitor). In sharp contrast to U0126 and wortmannin, PP2 treatment increased H3K9me3 levels upon co-stimulation with NRG-1 plus EGF (supplementary material Fig. S4A; Fig. 3D). A similar effect of SU6656 (another SFK inhibitor; Blake et al., 2000) was seen upon co-stimulation with NRG-1 plus EGF (supplementary material Fig. S4B). To scrutinize the role of SFKs (e.g. c-Src, c-Yes, Fyn and Lyn) in EGFR-mediated inhibition of ErbB4 signaling, we used embryonic fibroblast SYF cells, which are genetically deficient in expression of c-Src, c-Yes and Fyn and are undetectable in expression of Lyn (Klinghoffer et al., 1999), and the add-back version of SYF cells expressing active c-Src (SYF/c-Src cells) (supplementary material Fig. S5A) (Takahashi et al., 2009). ErbB4 activation by NRG-1 increased H3K9me3 levels in ErbB4-transfected SYF cells but could not increase those in ErbB4-transfected SYF/c-Src cells (Fig. 3E, top and middle). Note that high levels of H3K9me3 were observed in SYF cells exhibiting increased levels of nuclear ErbB4 (supplementary material Fig. S5C) and that in SYF/c-Src cells, NRG-1 stimulation did not increase the levels of nuclear ErbB4 and H3K9me3 (Fig. 3E, bottom; supplementary material Fig. S5C). To further examine whether the kinase activity of c-Src affected nuclear translocation of 4ICD, COS-1 cells were co-transfected with 4ICD and c-Src. Although c-Src was mainly localized to the cytoplasm (supplementary material Fig. S5D) (Kasahara et al., 2007a), c-Src expression inhibited nuclear localization of 4ICD and SU6656 treatment recovered the level of 4ICD in the nucleus (Fig. 3F), suggesting the inhibitory role of c-Src kinase activity in nuclear translocation of 4ICD.

Role of Tyr950 and Tyr1056 on 4ICD in the blockade of 4ICD nuclear signaling by c-Src

Since phosphorylation of EGFR by activated SFKs changes subcellular localization and functions of EGFR (Stover et al., 1995; Dittmann et al., 2008; Iida et al., 2012), we examined whether 4ICD was phosphorylated by c-Src. When COS-1 cells were co-transfected with 4ICD and c-Src, expression of c-Src increased the levels of tyrosine phosphorylation of 4ICD and SU6656 treatment obviously inhibited c-Src-induced 4ICD phosphorylation (Fig. 4A, bottom). Intriguingly, 4ICD was co-immunoprecipitated with c-Src in a manner dependent on c-Src kinase activity (Fig. 4A, bottom). We next examined the role of Tyr950 (highly conserved among all ErbB-family members) and Tyr1056 (unique to ErbB4; Carpenter, 2003) on 4ICD in association of 4ICD with c-Src and nuclear translocation of 4ICD. Mutation at Tyr950 and Tyr1056 to Phe (Y950F/Y1056F) greatly decreased association of 4ICD with c-Src (Fig. 4B,C), and this Y950F/Y1056F mutant failed to induce c-Src-mediated inhibition of the nuclear translocation of 4ICD (Fig. 4D). However, it was difficult to evaluate the contribution of Y950F and Y1056F to the c-Src-mediated 4ICD phosphorylation sites, because ErbB4 has at least 19 potential sites of tyrosine phosphorylation (Kaushansky et al., 2008). Nonetheless, high accumulation of Y950F/Y1056F in the nucleus was found to induce high levels of H3K9me3 (Fig. 4E). These results suggest that phosphorylation of Tyr950 and Tyr1056 on 4ICD by c-Src mediates the association of 4ICD with c-Src, thereby inhibiting nuclear translocation of 4ICD. In other words, unphosphorylated 4ICD at Tyr950 and Tyr1056 translocates into the nucleus and increases H3K9me3 levels.

Fig. 4.

Association of c-Src with pTyr950 and pTyr1056 on 4ICD, inhibits the nuclear accumulation of 4ICD and H3K9me3 modification. (A) COS-1 cells transfected with 4ICD plus vector or 4ICD plus c-Src–HA were cultured in the presence or absence of SU6656 for 24 hours. (Top) Western blot of equal amounts of Triton X-100 lysates was probed with anti-pTyr, anti-ErbB4, anti-Src and anti-actin (loading control) antibodies. (Bottom) 4ICD was immunoprecipitated with anti-ErbB4 antibody, and then analyzed by western blotting using anti-ErbB4, anti-pTyr and anti-Src antibodies. The level of 4ICD phosphorylation was quantified by measuring the signal intensity using anti-pTyr antibody and normalized to the corresponding signal intensity detected by anti-ErbB4 antibody. The amount of co-immunoprecipitated c-Src in the presence of SU6656 is expressed relative to that in the absence of SU6656. An arrowhead indicates a nonspecific band. (B) Schematic representations of 4ICD and its phenylalanine mutants at Tyr950 and Tyr1056. (C,D) COS-1 cells transfected with 4ICD plus vector or each 4ICD construct plus c-Src were cultured for 24 hours, and then cell lysates and 4ICD immunoprecipitates were analyzed as described in A (C). The amounts of co-immunoprecipitated c-Src with 4ICD or its mutants are expressed relative to those of the 4ICD immunoprecipitate. (D) Cells were stained with anti-ErbB4 antibody, and N≥Cyto localization of 4ICD was quantified, as described in Fig. 3F. Values are mean percentages ± s.d., n = 5 independent experiments. Scale bars: 10 µm. (E) COS-1 cells transfected with vector alone, 4ICD, 4ICD-Y1056F, or 4ICD-Y950F/Y1056F were cultured for 48 hours, and stained with anti-ErbB4 (expressed proteins) and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm. Asterisks indicate significant differences (*P<0.05; **P<0.01; ***P<0.001) calculated by Student's t-test.

Fig. 4.

Association of c-Src with pTyr950 and pTyr1056 on 4ICD, inhibits the nuclear accumulation of 4ICD and H3K9me3 modification. (A) COS-1 cells transfected with 4ICD plus vector or 4ICD plus c-Src–HA were cultured in the presence or absence of SU6656 for 24 hours. (Top) Western blot of equal amounts of Triton X-100 lysates was probed with anti-pTyr, anti-ErbB4, anti-Src and anti-actin (loading control) antibodies. (Bottom) 4ICD was immunoprecipitated with anti-ErbB4 antibody, and then analyzed by western blotting using anti-ErbB4, anti-pTyr and anti-Src antibodies. The level of 4ICD phosphorylation was quantified by measuring the signal intensity using anti-pTyr antibody and normalized to the corresponding signal intensity detected by anti-ErbB4 antibody. The amount of co-immunoprecipitated c-Src in the presence of SU6656 is expressed relative to that in the absence of SU6656. An arrowhead indicates a nonspecific band. (B) Schematic representations of 4ICD and its phenylalanine mutants at Tyr950 and Tyr1056. (C,D) COS-1 cells transfected with 4ICD plus vector or each 4ICD construct plus c-Src were cultured for 24 hours, and then cell lysates and 4ICD immunoprecipitates were analyzed as described in A (C). The amounts of co-immunoprecipitated c-Src with 4ICD or its mutants are expressed relative to those of the 4ICD immunoprecipitate. (D) Cells were stained with anti-ErbB4 antibody, and N≥Cyto localization of 4ICD was quantified, as described in Fig. 3F. Values are mean percentages ± s.d., n = 5 independent experiments. Scale bars: 10 µm. (E) COS-1 cells transfected with vector alone, 4ICD, 4ICD-Y1056F, or 4ICD-Y950F/Y1056F were cultured for 48 hours, and stained with anti-ErbB4 (expressed proteins) and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm. Asterisks indicate significant differences (*P<0.05; **P<0.01; ***P<0.001) calculated by Student's t-test.

NRG-1-repressed hTERT gene expression antagonized by EGF stimulation

The hTERT is involved in cell proliferation and differentiation (Poole et al., 2001), and silencing of the hTERT gene involves repressive histone modifications, i.e. H3K9me3 and H3K27me3, in its promoter region (Zinn et al., 2007; Chen et al., 2012). To examine how the signaling pathways via ErbB4 and EGFR affected gene expression of hTERT, we analyzed the expression levels of the hTERT mRNA in T47D cells stimulated with NRG-1, EGF, or NRG-1 plus EGF or left untreated for 4 days. Semi-quantitative RT-PCR analyses showed that the levels of hTERT gene expression were dramatically decreased in cells stimulated with NRG-1 but co-stimulation with NRG-1 plus EGF failed to increase the levels of hTERT gene expression (Fig. 5A). The results could be explained by a decrease in expression of EGFR on day 4 after EGF stimulation (supplementary material Fig. S3). We thus examined the effects of ErbB4 and EGFR on hTERT gene expression after 30 min of stimulation, because stimulation with EGF for 30 min did not decrease the levels of EGFR (supplementary material Fig. S1D) and inhibited NRG-1-induced increase of H3K9me3 levels (Fig. 3A,D; supplementary material Fig. S4B). It is worthy of note that NRG-1 stimulation for 30 min actually decreased hTERT gene expression, and that co-stimulation with NRG-1 plus EGF for 30 min could restore the levels of hTERT gene expression to those seen in unstimulated cells (Fig. 5B). In the presence of SU6656, co-stimulation with NRG-1 plus EGF for 30 min again decreased the levels of hTERT gene expression. Previous studies reported that hTERT gene expression is decreased by breast cancer 1 (BRCA1) (Li et al., 2002) and prolonged stimulation with NRG-1 increases the expression levels of BRCA1 through the ErbB4-JNK pathway (Muraoka-Cook et al., 2006). Given that NRG-1 alone and NRG-1 plus EGF consistently increased BRCA1 gene expression on day 4 after stimulation (supplementary material Fig. S6), the increased levels of BRCA1 may be involved in repression of hTERT gene expression after 4-day stimulation (Fig. 5A). However, NRG-1 stimulation for 30 min did not increase the expression levels of BRCA1 (Fig. 5B), suggesting that an increase in expression of BRCA1 is dispensable for NRG-1-induced repression of hTERT gene expression that is inhibited by EGF-activated Src.

Fig. 5.

Repression of hTERT gene expression by NRG-1, which is blocked by EGF-induced Src activation. The levels of hTERT and BRCA1 expression were assessed with semi-quantitative RT-PCR, and the amounts of hTERT and BRCA1 products were quantified by measuring band intensities and normalizing to the levels of GAPDH. (A) T47D cells were left untreated (none) or treated for 4 days with NRG-1, EGF or NRG-1 plus EGF, as described in Fig. 3B. Transcriptional expression levels of hTERT are expressed relative to those in nontreated cells (None). Values are means ± s.d., n = 4 independent experiments. The position of size markers in base pair (bp) are shown on the right in the top panel. (B) T47D cells were treated for 30 min with the indicated stimulation in the presence or absence of SU6656 as described in supplementary material Fig. S5B. The levels of hTERT and BRCA1 were quantified as described in A. Values are means ± s.d., n = 5 independent experiments. (C) T47D cells were treated with NRG-1 in the presence or absence of 1 µM DAPT for 30 min as described in Fig. 2A, and then expression levels of hTERT were quantified as described in A. Values are means ± s.d., n = 5 independent experiments. (D–G) Parental HeLa S3 cells (None) or HeLa S3 cells expressing NLS-4ICD (NLS-4ICD) were cultured for 8 hours, and then stained with anti-ErbB4 and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm (D). (E,F) Cell lysates were analyzed by western blotting using anti-ErbB4, anti-actin, anti-histone H3, anti-H3K9me3 (E), and anti-H3K27me3 (F) antibodies. The level of H3K9 or H3K27 tri-methylation was quantified by measuring the signal intensity using anti-H3K9me3 or anti-H3K27me3 antibodies, and normalizing to the corresponding signal intensity detected by anti-histone H3 antibody. (G) Expression levels of hTERT and BRCA1 were expressed as described in A. Values are means ± s.d., n = 3 independent experiments. Asterisks indicate significant differences (*P<0.05; **P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

Fig. 5.

Repression of hTERT gene expression by NRG-1, which is blocked by EGF-induced Src activation. The levels of hTERT and BRCA1 expression were assessed with semi-quantitative RT-PCR, and the amounts of hTERT and BRCA1 products were quantified by measuring band intensities and normalizing to the levels of GAPDH. (A) T47D cells were left untreated (none) or treated for 4 days with NRG-1, EGF or NRG-1 plus EGF, as described in Fig. 3B. Transcriptional expression levels of hTERT are expressed relative to those in nontreated cells (None). Values are means ± s.d., n = 4 independent experiments. The position of size markers in base pair (bp) are shown on the right in the top panel. (B) T47D cells were treated for 30 min with the indicated stimulation in the presence or absence of SU6656 as described in supplementary material Fig. S5B. The levels of hTERT and BRCA1 were quantified as described in A. Values are means ± s.d., n = 5 independent experiments. (C) T47D cells were treated with NRG-1 in the presence or absence of 1 µM DAPT for 30 min as described in Fig. 2A, and then expression levels of hTERT were quantified as described in A. Values are means ± s.d., n = 5 independent experiments. (D–G) Parental HeLa S3 cells (None) or HeLa S3 cells expressing NLS-4ICD (NLS-4ICD) were cultured for 8 hours, and then stained with anti-ErbB4 and anti-H3K9me3 antibodies. H3K9me3 levels were quantified as described in Fig. 1C. Scale bars: 10 µm (D). (E,F) Cell lysates were analyzed by western blotting using anti-ErbB4, anti-actin, anti-histone H3, anti-H3K9me3 (E), and anti-H3K27me3 (F) antibodies. The level of H3K9 or H3K27 tri-methylation was quantified by measuring the signal intensity using anti-H3K9me3 or anti-H3K27me3 antibodies, and normalizing to the corresponding signal intensity detected by anti-histone H3 antibody. (G) Expression levels of hTERT and BRCA1 were expressed as described in A. Values are means ± s.d., n = 3 independent experiments. Asterisks indicate significant differences (*P<0.05; **P<0.01; ***P<0.001; NS, not significant) calculated by Student's t-test.

To further substantiate transcriptional repression of hTERT by nuclear 4ICD, the levels of hTERT gene expression were examined in the presence or absence of DAPT. Treatment with DAPT blocked NRG-1-induced repression of hTERT gene expression (Fig. 5C). We then analyzed the levels of hTERT gene expression in NLS-4ICD-expressing HeLa S3 cells where not only increased levels of H3K9me3 but also decreased levels of H3K27me3 were detected by immunostaining and western blotting (Fig. 5D–F; supplementary material Fig. S7). Fig. 5G showed that the levels of hTERT gene expression were decreased upon NLS-4ICD expression. Taken together, these results suggest that repression of hTERT gene expression through nuclear ErbB4 signaling is negatively regulated by EGFR-activated Src.

In the present study, we show that ErbB4 activation by NRG-1 induces an increase of H3K9me3 levels and transcriptional repression of hTERT. NRG-1 stimulation activates ErbB4 and releases 4ICD from the cytoplasm to the nucleus. 4ICD-mediated nuclear tyrosine phosphorylation is involved in increased H3K9me3 levels, repression of hTERT gene expression, and inhibition of cell proliferation. We further show that co-activation of ErbB4 and EGFR by NRG-1 and EGF, respectively, blocks 4ICD nuclear signaling. It is of note that EGFR activation does not inhibit ErbB4 activation and 4ICD release. Rather, c-Src activated by EGFR inhibits nuclear translocation of 4ICD in a manner dependent on c-Src kinase activity. We can illustrate a model of the mechanism for the antagonizing effect of EGF to NRG-1-induced signaling toward histone modification and gene expression as well (Fig. 6A,B). The two tyrosine residues, Tyr950 and Tyr1056, on 4ICD are critical for association of 4ICD with c-Src, leading to inhibition of nuclear translocation of 4ICD and subsequent blockade of 4ICD nuclear signaling. Regulation of ErbB4-mediated nuclear signaling by c-Src-mediated tyrosine phosphorylation plays an important role in changes of histone modification and gene expression. It is therefore intriguing to note that the two ErbB-family members ErbB4 and EGFR have opposing effects on nuclear signaling.

Fig. 6.

Mechanism of inhibition of NRG-1-mediated ErbB4 signaling by EGF-induced Src activation. (A) NRG-1 activates ErbB4 and releases 4ICD from ErbB4. After the release, 4ICD translocates into the nucleus thereby increasing H3K9me3 and repressing hTERT gene expression. (B) Co-stimulation of ErbB4 and EGFR with NRG-1 and EGF, respectively. EGF-activated Src associates with pY950 and pY1056 on 4ICD, thereby inhibiting nuclear accumulation of 4ICD. Thus, EGFR activation antagonizes the signaling of NRG-1-activated ErbB4 toward an increase of H3K9me3 and repression of hTERT transcription.

Fig. 6.

Mechanism of inhibition of NRG-1-mediated ErbB4 signaling by EGF-induced Src activation. (A) NRG-1 activates ErbB4 and releases 4ICD from ErbB4. After the release, 4ICD translocates into the nucleus thereby increasing H3K9me3 and repressing hTERT gene expression. (B) Co-stimulation of ErbB4 and EGFR with NRG-1 and EGF, respectively. EGF-activated Src associates with pY950 and pY1056 on 4ICD, thereby inhibiting nuclear accumulation of 4ICD. Thus, EGFR activation antagonizes the signaling of NRG-1-activated ErbB4 toward an increase of H3K9me3 and repression of hTERT transcription.

It is well known that non-receptor-type tyrosine kinases act as co-transducers of transmembrane signals emanating from a variety of receptors, including receptor-type tyrosine kinases (Neet and Hunter, 1996). c-Src is a signal transducer of EGFR, and EGFR-activated c-Src enhances EGFR signaling through phosphorylation of tyrosine residues in the cytoplasmic region of EGFR (Biscardi et al., 1999; Dittmann et al., 2008). In addition to c-Src, the other SFK members c-Yes and Lyn are involved in nuclear translocation of EGFR and the binding of EGFR to the promoter region of target genes, such as B-Myb and iNOS (Iida et al., 2012), suggesting that SFKs are positive regulators of EGFR signaling. In sharp contrast, our results indicate that activated c-Src antagonizes nuclear ErbB4 signaling. NRG-1-induced increase of H3K9me3 is decreased by concomitant stimulation with EGF, and treatment with PP2 or SU6656 increases H3K9me3 levels upon co-stimulation with NRG-1 plus EGF (Fig. 3D; supplementary material Fig. S4). Furthermore, NRG-1 stimulation increases H3K9me3 levels in SYF cells transfected with ErbB4, whereas it cannot increase H3K9me3 levels in c-Src-expressing SYF cells (SYF/c-Src) transfected with ErbB4 (Fig. 3E). Importantly, hTERT gene expression is repressed by NRG-1 stimulation through 4ICD release, which is blocked by EGF stimulation through Src kinase activity (Fig. 5B,C). These results indicate that c-Src inhibits the nuclear ErbB4 signaling pathway for an increase of H3K9me3 and repression of hTERT gene expression.

Although Src kinase activity does not affect 4ICD cleavage (Fig. 3C), c-Src inhibits nuclear accumulation of 4ICD in a manner dependent on its kinase activity (Fig. 3E,F; supplementary material Fig. S5C). Mutational analyses reveal that Tyr950 and Tyr1056 on 4ICD are involved in association of 4ICD with c-Src and inhibition of the nuclear translocation of 4ICD (Fig. 4A–D). Furthermore, expression of NLS-4ICD greatly increases H3K9me3 levels and represses hTERT gene expression (Fig. 2C–G; Fig. 5D,E,G), suggesting that nuclear accumulation of 4ICD has a critical role in NRG-1-induced increase of H3K9me3 levels and gene repression of hTERT.

Why does c-Src have opposing effects on ErbB4 and EGFR signaling? Although c-Src inhibits the nuclear translocation of 4ICD through phosphorylation of Tyr950 and Tyr1056 on 4ICD (Fig. 3F; Fig. 4D), nuclear accumulation of EGFR is increased by c-Src (Dittmann et al., 2008). Phosphorylation of Y1101 on EGFR by the SFK members c-Yes and Lyn promotes nuclear translocation of EGFR (Iida et al., 2012). The ErbB-family members show high homology in the kinase domain (59–81% identity), whereas their divergent C-terminal regions are not conserved (11–25% identity) (Schulze et al., 2005). Given that Y1056 on ErbB4 and Y1101 on EGFR are located in their C-terminal regions, Y1056 unique to ErbB4 is not conserved among ErbB members but Y950 present in the ErbB4 kinase domain is conserved among all of them. We assume that the nuclear-cytoplasmic distribution of ErbB-family members is largely controlled by phosphorylation of the particular tyrosine residues on their cytoplasmic C-terminal domains. In addition, we have shown that the trafficking pathway of c-Src is different from those of c-Yes and Lyn, and differential trafficking of SFKs is specified by the state of palmitoylation in the Src homology 4 domain (Kasahara et al., 2004; Sato et al., 2009; Obata et al., 2010). These results raise the intriguing possibility that the role of each ErbB member could be modulated by individual SFK member-specific tyrosine phosphorylation of their cytoplasmic C-terminal domains.

Despite the involvement of nuclear 4ICD in gene expression (Fig. 5D) (Williams et al., 2004; Sardi et al., 2006; Zhu et al., 2006), it is still unclear how tyrosine phosphorylation by nuclear ErbB4 is involved in gene expression through histone modifications. Recent studies show that H3K9me3 is controlled by histone methyltransferases and histone demethylases (Kouzarides, 2007). Although 4ICD is known to interact with Krab-associated protein 1 (KAP1), which can associate with the H3K9-methyltransferase SETDB1, the interaction of 4ICD with KAP1 is independent of its kinase activity and KAP1 phosphorylation by 4ICD has not been detected thus far (Schultz et al., 2002; Gilmore-Hebert et al., 2010). A small increase of H3K9me3 levels in cells expressing NLS-4ICD(KD) may be explained by the interaction of 4ICD with KAP1 (Fig. 2F,G). In addition to an increase of H3K9me3, a decrease of H3K27me3 was observed upon NLS-4ICD expression (Fig. 5F; supplementary material Fig. S7). Recent studies showed that demethylation of H3K27me3 by JMJD3 is promoted through its interaction with estrogen receptor α (Agger et al., 2007; Svotelis et al., 2011). 4ICD is also known to interact with STAT5 in lactational gene expression (Williams et al., 2004; Zhu et al., 2006). It is therefore likely that a decrease of H3K27me3 by NLS-4ICD (Fig. 5F), which might involve JMJD3 and STAT5, leads to cell differentiation, whereas an increase of H3K27me3 by the histone methyltransferase Ezh2 that is recruited by STAT5 was reported to repress Igk recombination in proliferative pre-B cells (Mandal et al., 2011).

The expression levels of the ErbB members and their ligands are dynamically changed in mammary gland development. During pregnancy where differentiation and development of mammary glands actively take place (Robinson et al., 1995), transcriptional levels of ErbB4 and NRG-1 are increased and in return those of EGF are repressed (Schroeder and Lee, 1998). On lactation after pregnancy, transcriptional levels of NRG-1 are contrariwise repressed and those of EGF are increased (Schroeder and Lee, 1998). Furthermore, impaired differentiation and lactational failure were observed in ErbB4-deficient mice (Long et al., 2003). Given that overexpression of c-Src inhibits mammary epithelial cell differentiation (Jehn et al., 1992), we assume that the interplay of the NRG-1-ErbB4 signaling with the EGF-EGFR-c-Src signaling is important for the regulation of cell proliferation and differentiation. In addition, cell proliferation involves hTERT activity but low-level expression of hTERT relates to cell differentiation (Smith et al., 2003). High-level expression of hTERT, involved in cell transformation and carcinogenesis (Elenbaas et al., 2001), has been shown to correlate with poor differentiation and poor prognosis for a number of cancers, including neuroblastoma, acute myelogenous leukemia, and breast cancer (Poole et al., 2001). Taken together with our findings that nuclear ErbB4 signaling is involved in repression of hTERT gene expression (Fig. 5), these results suggest that ErbB4-mediated nuclear signaling regulates histone modifications and gene expression for differentiation.

Furthermore, our recent studies showed that nuclear tyrosine phosphorylation is in fact involved in chromatin structural changes and histone modifications (Yamaguchi et al., 2001; Nakayama and Yamaguchi, 2005; Takahashi et al., 2009; Aoyama et al., 2011). It was also reported that histone H3 is tyrosine-phosphorylated by Janus kinase 2, a non-receptor-type tyrosine kinase (Dawson et al., 2009). These results provide evidence that nuclear tyrosine phosphorylation by nuclear-localized tyrosine kinases, including 4ICD, has important roles in histone modifications and gene expression for proliferation and differentiation.

In conclusion, we show for the first time that nuclear ErbB4 signaling is involved in an increase of H3K9me3 and transcriptional repression of hTERT, both of which are antagonized by EGFR-activated c-Src, and the signal balance between ErbB4 and EGFR may be important for cell proliferation and differentiation. It would be interesting to determine how c-Src inhibits nuclear translocation of 4ICD and to identify substrates downstream of 4ICD. Further studies will contribute to a better understanding of the regulation of nuclear ErbB4 signaling through tyrosine phosphorylation.

Plasmids

cDNA encoding human ErbB4 CYT-1 subcloned into the pcDNA3.1-Zeo (+) (Ogiso et al., 2002) was gifted from S. Yokoyama (RIKEN, Yokohama). To construct the ErbB4 intracellular domain (4ICD, residues 676–1308), the Kozak sequence was created by PCR using ErbB4 as a template with 5′-GTGGTCGACGCCACCATGGTTTATGTTAGAAGGAAGAGCATCAAAAAG-3′ (sense), and 5′-GCGACTAGTCGGAGCTGACA-3′ (antisense). The SalI−XbaI fragment of the PCR product was subcloned into the XhoI-XbaI site of pcDNA4/TO (Invitrogen). FLAG-HA-NLS (Takahashi et al., 2009; Aoyama et al., 2011)-fused 4ICD (NLS-4ICD) was generated as follows. 4ICD was amplified by PCR using ErbB4 as a template with 5′-GCTGGTCGACTTAGAAGGAAGAGCATCAAAAAGAAAAGAGCCTTG-3′ (sense), and 5′-GCGACTAGTCGGAGCTGACA-3′ (antisense), and the SalI–XbaI fragment of the PCR product was introduced into the XhoI–XbaI site of pcDNA4/TO-NLS-Lyn-HA (Takahashi et al., 2009), resulting in the insertion sequence VLDPAQWRPRL between the NLS and 4ICD. NLS-4ICD was amplified by PCR using 5′-GAAGTCGACATGTCTTCTGATGATGAAGCTACTGC-3′ (sense) and 5′-GGAGCTGACACGGAAGATCAACGGCGCCGGCGCAAC-3′ (antisense). The SalI–NotI fragment of the PCR product was introduced into the XhoI–NotI site of pOZ-FHN (Nakatani and Ogryzko, 2003; provided by A. Iwama, Chiba University). The sequence LDGGYP was inserted between the FLAG and HA epitopes, and the sequence GGLA was inserted between the HA epitope and the NLS. The resulting FHN-NLS-4ICD was subcloned into pcDNA4/TO. The Lys→Arg mutation at position 751 in the ATP-binding site [kinase-dead (KD), K751R] was generated by site-directed mutagenesis using FHN-NLS-4ICD as a template with 5′-GGGACCAGTTGTCTCATTAAGAATTCTAATAGCCACAGGAATCTTCAC-3′ (sense) and 5′-GTGAAGATTCCTGTGGCTATTAGAATTCTTAATGAGACAACTGGTCCC-3′ (antisense). The Tyr→Phe mutation at position 1056 (4ICD-Y1056F) was created by PCR using 4ICD as a template with 5′-GACACAGCCCTCCGCCGGCCTTCACCCCCATGTCAGGAAACCAGTTTG-3′ (sense) and 5′-CTGACATGGGGGTGAAGGCCGGCGGAGGGCTGTGTCCAATTTCACTCC-3′ (antisense). The Tyr→Phe mutation at positions 950/1056 (4ICD-Y950F/Y1056F) was created by PCR using 4ICD-Y1056F as a template with 5′-TCTGCACTATTGACGTCTTCATGGTCATGGTCAAATGTTGGATGATTG-3′ (sense) and 5′-TTTGACCATGACCATGAAGACGTCAATAGTGCAGATGGGAGGCTGAGG-3′ (antisense). The NLS was added to the N-terminus of Src-HA was subcloned into pcDNA4/TO (Takahashi et al., 2009). To construct pcDNA4/TOneoR/NLS-4ICD, the XbaI–XbaI fragment of pcDNA4/TO/NLS-4ICD was subcloned into pcDNA4/TOneoR (Nakayama et al., 2009). pcDNA4/TOneoR/c-Src-HA (c-Src) was constructed as follows. The HindIII–XhoI fragment of pcDNA3/c-Src-HA (provided by S. A. Laporte, McGill University) was subcloned into pcDNA4/TOneoR. As a mutation was found at position Leu175, the NotI–KpnI fragment was replaced with the NotI–KpnI fragment of human wild-type c-Src (Bjorge et al., 1995) (provided by D. J. Fujita, University of Calgary).

Antibodies

The following antibodies were used: phosphotyrosine (pTyr, 4G10; Upstate Biotechnology Inc.; provided by T. Tamura, National Institute of Infection Diseases, Tokyo, and T. Yoshimoto, Tokyo Medical University) (Tamura et al., 2000), HA (12CA5), ErbB4 (C18; Santa Cruz Biotechnology, and HFR1; Abcam), EGFR (provided by M. N. Fukuda, Sanford-Burnham Medical Research Institute), phosphor-p44/42 MAP kinase Thr202/Tyr204 (pERK1/2, E10; New England Biolabs), ERK2 (C-14; Santa Cruz Biotechnology), Src[Y416] (phospho-Src family; BioSource), Src (GD11; Millipore), H3K4me3 (ab8580; Abcam), H3K9me3 (ab8898; Abcam), H4K16ac (Sigma-Aldrich), H3K27me3 (ab6002; Abcam), lamin A/C (N-18; Santa Cruz Biotechnology), and actin (clone C4; CHEMICON International). Horseradish peroxidase (HRP)-conjugated F(ab′)2 fragment of anti-mouse IgG, anti-rabbit IgG or anti-goat IgG antibodies were purchased from Amersham Biosciences. TRITC-anti-rabbit IgG or anti-mouse IgG (Fc specific), Alexa Fluor 488 anti-mouse IgG or anti-rabbit IgG, and Alexa Fluor 647 anti-rabbit IgG secondary antibodies were from BioSource International, Sigma-Aldrich and Invitrogen.

Cells and transfection

COS-1 (Japanese Collection of Research Bioresources, Osaka), T47D (provided by M. Tagawa, Chiba Cancer Center Research Institute), and SYF cells (Klinghoffer et al., 1999) were cultured in Iscove's modified Dulbecco's medium containing 5% fetal bovine serum (FBS). SYF cells stably expressing c-Src-wt (SYF/c-Src) were prepared (Takahashi et al., 2009). Cells were transiently transfected with plasmid DNA using polyethylenimine (Fukumoto et al., 2010). For growth factor stimulation, T47D cells were starved under low serum conditions [0.05% bovine serum (BS)] for 24 hours, or SYF and SYF/c-Src cells transfected with vector or ErbB4 were cultured for 8 hours and then starved for 16 hours. Starved cells were left untreated or treated with 20 ng/ml NRG-1 (Neuregulin β-1; R&D Systems), 20 ng/ml EGF, or 20 ng/ml NRG-1 plus 20 ng/ml EGF for 30 min under low serum conditions (0.05% BS). A HeLa S3 cell clone expressing NLS-4ICD was generated in an inducible manner (HeLa S3/TR/NLS-4ICD), because we could not establish a cell line stably expressing NLS-4ICD. HeLa S3/TR (clone A3f5) cells, which stably express the tetracycline repressor (TR) (Aoyama et al., 2011), were transfected with pcDNA4/TOneo/NLS-4ICD, and cell clones inducibly expressing NLS-4ICD were selected in 500 µg/ml G418. Expression of NLS-4ICD was induced for 8 hours by 2 µg/ml doxycycline, a tetracycline derivative.

Inhibitors

The following inhibitors were used: 20 µM AG1478, 10 µM PP2, 10 µM SU6656, 20 µM U0126, 1 or 25 µM DAPT, and 100 nM wortmannin.

Immunofluorescence

Immunofluorescence staining was performed as described (Yamaguchi and Fukuda, 1995; Tada et al., 1999; Nakayama and Yamaguchi, 2005; Kasahara et al., 2007b; Ikeda et al., 2008; Takahashi et al., 2009; Nakayama et al., 2012). In brief, cells were washed in warmed phosphate-buffered saline (PBS) and fixed in 100% methanol for 5 min at −20°C (Ni et al., 2001; Aoyama et al., 2011). Fixed cells were permeabilized and blocked in PBS containing 0.1% saponin and 3% BSA for 20 min at room temperature, and then incubated with a primary and a secondary antibody for 1 hour each at room temperature. For DNA staining, cells were subsequently treated with 200 µg/ml RNase A for 30 min and 20 µg/ml propidium iodide (PI) or 20 nM TOPRO-3 for 30 min at room temperature. After washing with PBS containing 0.1% saponin, cells were mounted with ProLong antifade reagent (Molecular Probes). The resulting red emission of TOPRO-3-stained nuclei is pseudo-colored as blue. Confocal and Nomarski differential-interference-contrast (DIC) images were obtained at room temperature using a Fluoview FV500 confocal laser scanning microscope with a 40× 1.00 NA or a 60× 1.00 NA water-immersion objective (Olympus, Tokyo) and an LSM510 laser scanning microscope with 40× 0.75 NA or 63× 1.40 NA oil immersion objective (Carl Zeiss). One planar (xy) section slice (0.8- or 2.0-µm thickness) was shown in most experiments. Composite figures were prepared using Photoshop 11.0 and Illustrator 14.0 software (Adobe). For quantification of histone modifications, confocal images were obtained using a Fluoview FV500 confocal laser scanning microscope, and then fluorescence intensities of immunostaining were measured using ImageJ software (National Institutes of Health). Since two or three independent experiments gave similar results, a representative experiment was shown.

Measurement of DNA synthesis using bromodeoxyuridine incorporation

DNA synthesis was measured by incorporation of bromodeoxyuridine (BrdU; Roche) into the genomic DNA during the S phase (DNA replication) of the cell cycle. T47D cells were starved under low serum conditions (0.05% BS) for 24 hours, and then treated with NRG-1 or EGF in the presence of BrdU for 24 hours. Cells were fixed with ethanol and 50 mM glycine, pH 2.0, for 45 min at room temperature and denatured in 4 N HCl for 15 min. Subsequent immunodetection of BrdU was accomplished, according to the manufacturer's instructions.

Western blotting and immunoprecipitation

Cell lysates were prepared in SDS-PAGE sample buffer or Triton X-100 lysis buffer (20 mM HEPES, pH 7.8, 5% glycerol, 1% Triton X-100, 5 mM EDTA, 50 mM NaF, 20 mM β-glycerophosphate, 50 µg/ml aprotinin, 100 µM leupeptin, 25 µM pepstatin, 10 mM Na3VO4, and 1 mM PMSF), and subjected to SDS-PAGE and electrotransferred onto polyvinylidene difluoride membrane (Millipore). Immunodetection was performed by enhanced chemiluminescence (Amersham Biosciences) as described (Yamaguchi et al., 2001; Matsuda et al., 2006; Kasahara et al., 2007a; Kuga et al., 2007; Ikeda et al., 2008; Sato et al., 2009; Nakayama et al., 2012). Sequential reprobing of membranes with a variety of antibodies was performed after the complete removal of primary antibodies from membranes in stripping buffer or inactivation of HRP by 0.1% NaN3, according to the manufacturer's instructions. Results were analyzed using a ChemiDoc XRSPlus image analyzer (Bio-Rad), and quantitated with Quantity One software (Bio-Rad). Immunoprecipitation was performed using antibody-precoated protein-G beads, as described (Mera et al., 1999; Yamaguchi et al., 2001; Ikeda et al., 2008; Obata et al., 2010). Composite figures were prepared using Photoshop 11.0 and Illustrator 14.0 software (Adobe).

Semi-quantitative RT-PCR

Total RNAs were isolated from T47D cells with the ISOGEN reagent (Nippon Gene, Tokyo), and cDNAs were synthesized from 1 µg of each RNA preparation using the PrimeScript RT reagent Kit (TakaraBio, Shiga) as described (Kikuchi et al., 2010; Aoyama et al., 2011). To avoid saturation of PCR products, conditions of PCR were optimized before semi-quantitative RT-PCR was carried out. The primers used for PCR are as follows: human telomerase reverse transcriptase (hTERT), 5′-CGGAAGAGTGTCTGGAGCAA-3′ (sense) and 5′-GGATGAAGCGGAGTCTGGA-3′ (antisense) (Nakamura et al., 1997); breast cancer 1 (BRCA1), 5′-TTGCGGGAGGAAAATGGGTAGTTA-3′ (sense) and 5′-TGTGCCAAGGGTGAATGATGAAAG-3′ (antisense) (Ju et al., 2007); glyceraldehyde 3-phosphate dehydrogenase (GAPDH), 5′-ACCACAGTCCATGCCATCAC-3′ (sense) and 5′-TCCACCACCCTGTTGCTGTA-3′ (antisense) (Kikuchi et al., 2010; Aoyama et al., 2011). The sizes of PCR products are 146, 292 and 452 bp, respectively. Amplification was carried out using an MJ mini thermal cycler (Bio-Rad) with Ex Taq DNA polymerase (TakaraBio, Shiga) under the following conditions: For hTERT, initial heating at 95°C for 1 min, followed by 35 cycles of denaturation at 95°C for 30 sec, annealing at 56°C for 30 sec and extension at 72°C for 30 sec. For BRCA1 and GAPDH, initial heating at 94°C for 2 min, followed by 27 or 25 cycles of denaturation at 94°C for 30 sec, annealing at 53°C or 56°C for 30 sec and extension at 72°C for 1 min. The products of RT-PCR were electrophoresed on a 2.0 or 3.0% agarose gel containing ethidium bromide. The density of each amplified fragment was quantified with ChemiDoc XRSPlus and Quantity One software.

We thank S. Yokoyama, D. J. Fujita, S. A. Laporte, T. Tamura, T. Yoshimoto, M. Tagawa, A. Iwama and M. N. Fukuda for their invaluable plasmids, antibodies and cell lines.

Funding

This work was supported in part by grants-in-aid for Scientific Research, Global Center for Education and Research in Immune Regulation and Treatment program and Special Funds for Education and Research (Development of SPECT probes for Pharmaceutical Innovation) both from the Japanese Ministry of Education, Culture, Sports, Science and Technology; and the Nanohana Competition 2011 Award of Chiba University and the Futaba Electronic Memorial Foundation to Y.F.; and G-COE Research Assistantship to K.I., H.H., K.-M.A. and S.K.

Agger
K.
,
Cloos
P. A.
,
Christensen
J.
,
Pasini
D.
,
Rose
S.
,
Rappsilber
J.
,
Issaeva
I.
,
Canaani
E.
,
Salcini
A. E.
,
Helin
K.
(
2007
).
UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development.
Nature
449
,
731
734
.
Aoyama
K.
,
Fukumoto
Y.
,
Ishibashi
K.
,
Kubota
S.
,
Morinaga
T.
,
Horiike
Y.
,
Yuki
R.
,
Takahashi
A.
,
Nakayama
Y.
,
Yamaguchi
N.
(
2011
).
Nuclear c-Abl-mediated tyrosine phosphorylation induces chromatin structural changes through histone modifications that include H4K16 hypoacetylation.
Exp. Cell Res.
317
,
2874
2903
.
Biscardi
J. S.
,
Maa
M. C.
,
Tice
D. A.
,
Cox
M. E.
,
Leu
T. H.
,
Parsons
S. J.
(
1999
).
c-Src-mediated phosphorylation of the epidermal growth factor receptor on Tyr845 and Tyr1101 is associated with modulation of receptor function.
J. Biol. Chem.
274
,
8335
8343
.
Bjorge
J. D.
,
Bellagamba
C.
,
Cheng
H. C.
,
Tanaka
A.
,
Wang
J. H.
,
Fujita
D. J.
(
1995
).
Characterization of two activated mutants of human pp60c-src that escape c-Src kinase regulation by distinct mechanisms.
J. Biol. Chem.
270
,
24222
24228
.
Blake
R. A.
,
Broome
M. A.
,
Liu
X.
,
Wu
J.
,
Gishizky
M.
,
Sun
L.
,
Courtneidge
S. A.
(
2000
).
SU6656, a selective src family kinase inhibitor, used to probe growth factor signaling.
Mol. Cell. Biol.
20
,
9018
9027
.
Cans
C.
,
Mangano
R.
,
Barilá
D.
,
Neubauer
G.
,
Superti–Furga
G.
(
2000
).
Nuclear tyrosine phosphorylation: the beginning of a map.
Biochem. Pharmacol.
60
,
1203
1215
.
Carpenter
G.
(
2003
).
ErbB-4: mechanism of action and biology.
Exp. Cell Res.
284
,
66
77
.
Chen
I. P.
,
Henning
S.
,
Faust
A.
,
Boukamp
P.
,
Volkmer
B.
,
Greinert
R.
(
2012
).
UVA-induced epigenetic regulation of P16(INK4a) in human epidermal keratinocytes and skin tumor derived cells.
Photochem. Photobiol. Sci.
11
,
180
190
.
Dawson
M. A.
,
Bannister
A. J.
,
Göttgens
B.
,
Foster
S. D.
,
Bartke
T.
,
Green
A. R.
,
Kouzarides
T.
(
2009
).
JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin.
Nature
461
,
819
822
.
Dikic
I.
,
Giordano
S.
(
2003
).
Negative receptor signalling.
Curr. Opin. Cell Biol.
15
,
128
135
.
Dittmann
K.
,
Mayer
C.
,
Kehlbach
R.
,
Rodemann
H. P.
(
2008
).
Radiation-induced caveolin-1 associated EGFR internalization is linked with nuclear EGFR transport and activation of DNA-PK.
Mol. Cancer
7
,
69
.
Egeblad
M.
,
Mortensen
O. H.
,
van Kempen
L. C.
,
Jäättelä
M.
(
2001
).
BIBX1382BS, but not AG1478 or PD153035, inhibits the ErbB kinases at different concentrations in intact cells.
Biochem. Biophys. Res. Commun.
281
,
25
31
.
Elenbaas
B.
,
Spirio
L.
,
Koerner
F.
,
Fleming
M. D.
,
Zimonjic
D. B.
,
Donaher
J. L.
,
Popescu
N. C.
,
Hahn
W. C.
,
Weinberg
R. A.
(
2001
).
Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells.
Genes Dev.
15
,
50
65
.
Fukumoto
Y.
,
Obata
Y.
,
Ishibashi
K.
,
Tamura
N.
,
Kikuchi
I.
,
Aoyama
K.
,
Hattori
Y.
,
Tsuda
K.
,
Nakayama
Y.
,
Yamaguchi
N.
(
2010
).
Cost-effective gene transfection by DNA compaction at pH 4.0 using acidified, long shelf-life polyethylenimine.
Cytotechnology
62
,
73
82
.
Gilmore–Hebert
M.
,
Ramabhadran
R.
,
Stern
D. F.
(
2010
).
Interactions of ErbB4 and Kap1 connect the growth factor and DNA damage response pathways.
Mol. Cancer Res.
8
,
1388
1398
.
Hubbard
S. R.
,
Till
J. H.
(
2000
).
Protein tyrosine kinase structure and function.
Annu. Rev. Biochem.
69
,
373
398
.
Iida
M.
,
Brand
T. M.
,
Campbell
D. A.
,
Li
C.
,
Wheeler
D. L.
(
2013
).
Yes and Lyn play a role in nuclear translocation of the epidermal growth factor receptor.
Oncogene
32
,
759
767
.
Ikeda
K.
,
Nakayama
Y.
,
Togashi
Y.
,
Obata
Y.
,
Kuga
T.
,
Kasahara
K.
,
Fukumoto
Y.
,
Yamaguchi
N.
(
2008
).
Nuclear localization of Lyn tyrosine kinase mediated by inhibition of its kinase activity.
Exp. Cell Res.
314
,
3392
3404
.
Jehn
B.
,
Costello
E.
,
Marti
A.
,
Keon
N.
,
Deane
R.
,
Li
F.
,
Friis
R. R.
,
Burri
P. H.
,
Martin
F.
,
Jaggi
R.
(
1992
).
Overexpression of Mos, Ras, Src, and Fos inhibits mouse mammary epithelial cell differentiation.
Mol. Cell. Biol.
12
,
3890
3902
.
Jenuwein
T.
,
Allis
C. D.
(
2001
).
Translating the histone code.
Science
293
,
1074
1080
.
Ju
D. S.
,
Kim
M. J.
,
Bae
J. H.
,
Song
H. S.
,
Chung
B. S.
,
Lee
M. K.
,
Kang
C. D.
,
Lee
H. S.
,
Kim
D. W.
,
Kim
S. H.
(
2007
).
Camptothecin acts synergistically with imatinib and overcomes imatinib resistance through Bcr-Abl independence in human K562 cells.
Cancer Lett.
252
,
75
85
.
Kasahara
K.
,
Nakayama
Y.
,
Ikeda
K.
,
Fukushima
Y.
,
Matsuda
D.
,
Horimoto
S.
,
Yamaguchi
N.
(
2004
).
Trafficking of Lyn through the Golgi caveolin involves the charged residues on alphaE and alphaI helices in the kinase domain.
J. Cell Biol.
165
,
641
652
.
Kasahara
K.
,
Nakayama
Y.
,
Kihara
A.
,
Matsuda
D.
,
Ikeda
K.
,
Kuga
T.
,
Fukumoto
Y.
,
Igarashi
Y.
,
Yamaguchi
N.
(
2007a
).
Rapid trafficking of c-Src, a non-palmitoylated Src-family kinase, between the plasma membrane and late endosomes/lysosomes.
Exp. Cell Res.
313
,
2651
2666
.
Kasahara
K.
,
Nakayama
Y.
,
Sato
I.
,
Ikeda
K.
,
Hoshino
M.
,
Endo
T.
,
Yamaguchi
N.
(
2007b
).
Role of Src-family kinases in formation and trafficking of macropinosomes.
J. Cell. Physiol.
211
,
220
232
.
Kaushansky
A.
,
Gordus
A.
,
Budnik
B. A.
,
Lane
W. S.
,
Rush
J.
,
MacBeath
G.
(
2008
).
System-wide investigation of ErbB4 reveals 19 sites of Tyr phosphorylation that are unusually selective in their recruitment properties.
Chem. Biol.
15
,
808
817
.
Kikuchi
I.
,
Nakayama
Y.
,
Morinaga
T.
,
Fukumoto
Y.
,
Yamaguchi
N.
(
2010
).
A decrease in cyclin B1 levels leads to polyploidization in DNA damage-induced senescence.
Cell Biol. Int.
34
,
645
653
.
Klinghoffer
R. A.
,
Sachsenmaier
C.
,
Cooper
J. A.
,
Soriano
P.
(
1999
).
Src family kinases are required for integrin but not PDGFR signal transduction.
EMBO J.
18
,
2459
2471
.
Kouzarides
T.
(
2007
).
Chromatin modifications and their function.
Cell
128
,
693
705
.
Kuga
T.
,
Nakayama
Y.
,
Hoshino
M.
,
Higashiyama
Y.
,
Obata
Y.
,
Matsuda
D.
,
Kasahara
K.
,
Fukumoto
Y.
,
Yamaguchi
N.
(
2007
).
Differential mitotic activation of endogenous c-Src, c-Yes, and Lyn in HeLa cells.
Arch. Biochem. Biophys.
466
,
116
124
.
Li
H.
,
Lee
T. H.
,
Avraham
H.
(
2002
).
A novel tricomplex of BRCA1, Nmi, and c-Myc inhibits c-Myc-induced human telomerase reverse transcriptase gene (hTERT) promoter activity in breast cancer.
J. Biol. Chem.
277
,
20965
20973
.
Linggi
B.
,
Carpenter
G.
(
2006
).
ErbB receptors: new insights on mechanisms and biology.
Trends Cell Biol.
16
,
649
656
.
Long
W.
,
Wagner
K. U.
,
Lloyd
K. C.
,
Binart
N.
,
Shillingford
J. M.
,
Hennighausen
L.
,
Jones
F. E.
(
2003
).
Impaired differentiation and lactational failure of Erbb4-deficient mammary glands identify ERBB4 as an obligate mediator of STAT5.
Development
130
,
5257
5268
.
Määttä
J. A.
,
Sundvall
M.
,
Junttila
T. T.
,
Peri
L.
,
Laine
V. J.
,
Isola
J.
,
Egeblad
M.
,
Elenius
K.
(
2006
).
Proteolytic cleavage and phosphorylation of a tumor-associated ErbB4 isoform promote ligand-independent survival and cancer cell growth.
Mol. Biol. Cell
17
,
67
79
.
Mandal
M.
,
Powers
S. E.
,
Maienschein–Cline
M.
,
Bartom
E. T.
,
Hamel
K. M.
,
Kee
B. L.
,
Dinner
A. R.
,
Clark
M. R.
(
2011
).
Epigenetic repression of the Igk locus by STAT5-mediated recruitment of the histone methyltransferase Ezh2.
Nat. Immunol.
12
,
1212
1220
.
Matsuda
D.
,
Nakayama
Y.
,
Horimoto
S.
,
Kuga
T.
,
Ikeda
K.
,
Kasahara
K.
,
Yamaguchi
N.
(
2006
).
Involvement of Golgi-associated Lyn tyrosine kinase in the translocation of annexin II to the endoplasmic reticulum under oxidative stress.
Exp. Cell Res.
312
,
1205
1217
.
Mera
A.
,
Suga
M.
,
Ando
M.
,
Suda
T.
,
Yamaguchi
N.
(
1999
).
Induction of cell shape changes through activation of the interleukin-3 common beta chain receptor by the RON receptor-type tyrosine kinase.
J. Biol. Chem.
274
,
15766
15774
.
Moorhead
G. B.
,
Trinkle–Mulcahy
L.
,
Ulke–Lemée
A.
(
2007
).
Emerging roles of nuclear protein phosphatases.
Nat. Rev. Mol. Cell Biol.
8
,
234
244
.
Muraoka–Cook
R. S.
,
Caskey
L. S.
,
Sandahl
M. A.
,
Hunter
D. M.
,
Husted
C.
,
Strunk
K. E.
,
Sartor
C. I.
,
Rearick
W. A.
 Jr
,
McCall
W.
,
Sgagias
M. K.
et al.  (
2006
).
Heregulin-dependent delay in mitotic progression requires HER4 and BRCA1.
Mol. Cell. Biol.
26
,
6412
6424
.
Nakamura
T. M.
,
Morin
G. B.
,
Chapman
K. B.
,
Weinrich
S. L.
,
Andrews
W. H.
,
Lingner
J.
,
Harley
C. B.
,
Cech
T. R.
(
1997
).
Telomerase catalytic subunit homologs from fission yeast and human.
Science
277
,
955
959
.
Nakatani
Y.
,
Ogryzko
V.
(
2003
).
Immunoaffinity purification of mammalian protein complexes.
Methods Enzymol.
370
,
430
444
.
Nakayama
Y.
,
Yamaguchi
N.
(
2005
).
Multi-lobulation of the nucleus in prolonged S phase by nuclear expression of Chk tyrosine kinase.
Exp. Cell Res.
304
,
570
581
.
Nakayama
Y.
,
Igarashi
A.
,
Kikuchi
I.
,
Obata
Y.
,
Fukumoto
Y.
,
Yamaguchi
N.
(
2009
).
Bleomycin-induced over-replication involves sustained inhibition of mitotic entry through the ATM/ATR pathway.
Exp. Cell Res.
315
,
2515
2528
.
Nakayama
Y.
,
Matsui
Y.
,
Takeda
Y.
,
Okamoto
M.
,
Abe
K.
,
Fukumoto
Y.
,
Yamaguchi
N.
(
2012
).
c-Src but not Fyn promotes proper spindle orientation in early prometaphase.
J. Biol. Chem.
287
,
24905
24915
.
Neet
K.
,
Hunter
T.
(
1996
).
Vertebrate non-receptor protein-tyrosine kinase families.
Genes Cells
1
,
147
169
.
Ni
C. Y.
,
Murphy
M. P.
,
Golde
T. E.
,
Carpenter
G.
(
2001
).
γ-Secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase.
Science
294
,
2179
2181
.
Obata
Y.
,
Fukumoto
Y.
,
Nakayama
Y.
,
Kuga
T.
,
Dohmae
N.
,
Yamaguchi
N.
(
2010
).
The Lyn kinase C-lobe mediates Golgi export of Lyn through conformation-dependent ACSL3 association.
J. Cell Sci.
123
,
2649
2662
.
Ogiso
H.
,
Ishitani
R.
,
Nureki
O.
,
Fukai
S.
,
Yamanaka
M.
,
Kim
J. H.
,
Saito
K.
,
Sakamoto
A.
,
Inoue
M.
,
Shirouzu
M.
et al.  (
2002
).
Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains.
Cell
110
,
775
787
.
Paatero
I.
,
Jokilammi
A.
,
Heikkinen
P. T.
,
Iljin
K.
,
Kallioniemi
O. P.
,
Jones
F. E.
,
Jaakkola
P. M.
,
Elenius
K.
(
2012
).
Interaction with ErbB4 promotes hypoxia-inducible factor-1α signaling.
J. Biol. Chem.
287
,
9659
9671
.
Poole
J. C.
,
Andrews
L. G.
,
Tollefsbol
T. O.
(
2001
).
Activity, function, and gene regulation of the catalytic subunit of telomerase (hTERT).
Gene
269
,
1
12
.
Rio
C.
,
Buxbaum
J. D.
,
Peschon
J. J.
,
Corfas
G.
(
2000
).
Tumor necrosis factor-alpha-converting enzyme is required for cleavage of erbB4/HER4.
J. Biol. Chem.
275
,
10379
10387
.
Robinson
G. W.
,
McKnight
R. A.
,
Smith
G. H.
,
Hennighausen
L.
(
1995
).
Mammary epithelial cells undergo secretory differentiation in cycling virgins but require pregnancy for the establishment of terminal differentiation.
Development
121
,
2079
2090
.
Sardi
S. P.
,
Murtie
J.
,
Koirala
S.
,
Patten
B. A.
,
Corfas
G.
(
2006
).
Presenilin-dependent ErbB4 nuclear signaling regulates the timing of astrogenesis in the developing brain.
Cell
127
,
185
197
.
Sato
I.
,
Obata
Y.
,
Kasahara
K.
,
Nakayama
Y.
,
Fukumoto
Y.
,
Yamasaki
T.
,
Yokoyama
K. K.
,
Saito
T.
,
Yamaguchi
N.
(
2009
).
Differential trafficking of Src, Lyn, Yes and Fyn is specified by the state of palmitoylation in the SH4 domain.
J. Cell Sci.
122
,
965
975
.
Schroeder
J. A.
,
Lee
D. C.
(
1998
).
Dynamic expression and activation of ERBB receptors in the developing mouse mammary gland.
Cell Growth Differ.
9
,
451
464
.
Schultz
D. C.
,
Ayyanathan
K.
,
Negorev
D.
,
Maul
G. G.
,
Rauscher
F. J.
 3rd
(
2002
).
SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins.
Genes Dev.
16
,
919
932
.
Schulze
W. X.
,
Deng
L.
,
Mann
M.
(
2005
).
Phosphotyrosine interactome of the ErbB-receptor kinase family.
Mol. Syst. Biol.
1
,
2005.0008
.
Smith
L. L.
,
Coller
H. A.
,
Roberts
J. M.
(
2003
).
Telomerase modulates expression of growth-controlling genes and enhances cell proliferation.
Nat. Cell Biol.
5
,
474
479
.
Stover
D. R.
,
Becker
M.
,
Liebetanz
J.
,
Lydon
N. B.
(
1995
).
Src phosphorylation of the epidermal growth factor receptor at novel sites mediates receptor interaction with Src and P85 α.
J. Biol. Chem.
270
,
15591
15597
.
Svotelis
A.
,
Bianco
S.
,
Madore
J.
,
Huppé
G.
,
Nordell–Markovits
A.
,
Mes–Masson
A. M.
,
Gévry
N.
(
2011
).
H3K27 demethylation by JMJD3 at a poised enhancer of anti-apoptotic gene BCL2 determines ERα ligand dependency.
EMBO J.
30
,
3947
3961
.
Tada
J.
,
Omine
M.
,
Suda
T.
,
Yamaguchi
N.
(
1999
).
A common signaling pathway via Syk and Lyn tyrosine kinases generated from capping of the sialomucins CD34 and CD43 in immature hematopoietic cells.
Blood
93
,
3723
3735
.
Takahashi
A.
,
Obata
Y.
,
Fukumoto
Y.
,
Nakayama
Y.
,
Kasahara
K.
,
Kuga
T.
,
Higashiyama
Y.
,
Saito
T.
,
Yokoyama
K. K.
,
Yamaguchi
N.
(
2009
).
Nuclear localization of Src-family tyrosine kinases is required for growth factor-induced euchromatinization.
Exp. Cell Res.
315
,
1117
1141
.
Tamura
T.
,
Kunimatsu
T.
,
Yee
S. T.
,
Igarashi
O.
,
Utsuyama
M.
,
Tanaka
S.
,
Miyazaki
S.
,
Hirokawa
K.
,
Nariuchi
H.
(
2000
).
Molecular mechanism of the impairment in activation signal transduction in CD4(+) T cells from old mice.
Int. Immunol.
12
,
1205
1215
.
Ullrich
A.
,
Schlessinger
J.
(
1990
).
Signal transduction by receptors with tyrosine kinase activity.
Cell
61
,
203
212
.
Williams
C. C.
,
Allison
J. G.
,
Vidal
G. A.
,
Burow
M. E.
,
Beckman
B. S.
,
Marrero
L.
,
Jones
F. E.
(
2004
).
The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone.
J. Cell Biol.
167
,
469
478
.
Yamaguchi
N.
,
Fukuda
M. N.
(
1995
).
Golgi retention mechanism of beta-1,4-galactosyltransferase. Membrane-spanning domain-dependent homodimerization and association with alpha- and beta-tubulins.
J. Biol. Chem.
270
,
12170
12176
.
Yamaguchi
N.
,
Nakayama
Y.
,
Urakami
T.
,
Suzuki
S.
,
Nakamura
T.
,
Suda
T.
,
Oku
N.
(
2001
).
Overexpression of the Csk homologous kinase (Chk tyrosine kinase) induces multinucleation: a possible role for chromosome-associated Chk in chromosome dynamics.
J. Cell Sci.
114
,
1631
1641
.
Yarden
Y.
,
Sliwkowski
M. X.
(
2001
).
Untangling the ErbB signalling network.
Nat. Rev. Mol. Cell Biol.
2
,
127
137
.
Zhu
Y.
,
Sullivan
L. L.
,
Nair
S. S.
,
Williams
C. C.
,
Pandey
A. K.
,
Marrero
L.
,
Vadlamudi
R. K.
,
Jones
F. E.
(
2006
).
Coregulation of estrogen receptor by ERBB4/HER4 establishes a growth-promoting autocrine signal in breast tumor cells.
Cancer Res.
66
,
7991
7998
.
Zinn
R. L.
,
Pruitt
K.
,
Eguchi
S.
,
Baylin
S. B.
,
Herman
J. G.
(
2007
).
hTERT is expressed in cancer cell lines despite promoter DNA methylation by preservation of unmethylated DNA and active chromatin around the transcription start site.
Cancer Res.
67
,
194
201
.

Supplementary information