Transforming growth factor β (TGFβ) signaling controls many cellular responses including proliferation, epithelial to mesenchymal transition and apoptosis, through the activation of canonical (Smad) as well as non-canonical (e.g. Par6) pathways. Previous studies from our lab have demonstrated that aPKC inhibition regulates TGFβ receptor trafficking and signaling. Here, we report that downstream TGFβ-dependent transcriptional responses in aPKC-silenced NSCLC cells were reduced compared with those of control cells, despite a temporal extension of Smad2 phosphorylation. We assessed SARA–Smad2–Smad4 association and observed that knockdown of aPKC increased SARA (also known as ZFYVE9) levels and SARA–Smad2 complex formation, increased cytoplasmic retention of Smad2 and reduced Smad2–Smad4 complex formation, which correlated with reduced Smad2 nuclear translocation. Interestingly, we also detected an increase in p38 MAPK phosphorylation and apoptosis in aPKC-silenced cells, which were found to be TRAF6-dependent. Taken together, our results suggest that aPKC isoforms regulate Smad and non-Smad TGFβ pathways and that aPKC inhibition sensitizes NSCLC cells to undergo TGFβ-dependent apoptosis.

Transforming growth factor β (TGFβ) signaling regulates many cellular processes including proliferation, apoptosis, and epithelial to mesenchymal transition (EMT), and, in addition to being a key pathway during embryonic development, aberrant TGFβ signaling is a hallmark of several pathological conditions, including cancer and fibrosis (Derynck and Akhurst, 2007; Derynck et al., 2001; Elliott and Blobe, 2005; Massagué, 1992; Massagué and Chen, 2000). The canonical TGFβ pathway involves the cell-surface binding of TGFβ ligand to the TGFβ type II receptor (TβRII), which then binds to and phosphorylates the TGFβ type I receptor (TβRI) (Massagué, 1998). Phosphorylation of TβRI leads to its activation and its ability to transduce intracellular signaling through the phosphorylation of substrate proteins such as the receptor-regulated Smads (R-Smads) Smad2 and Smad3 (Massagué, 1998; Massagué and Chen, 2000). Once phosphorylated, R-Smads accumulate in the nucleus, where they act as transcription factors to regulate subsequent TGFβ gene response (Attisano and Wrana, 2002; Massagué, 1998; Massagué and Chen, 2000; Shi and Massagué, 2003; Siegel and Massagué, 2003). Entry of R-Smads into the nucleus is facilitated by direct binding of these proteins to the nucleopore complex, binding to importins (for Smad3) or by the binding of the common Smad, Smad 4 (Massagué, 2003; Shi and Massagué, 2003)

Importantly, proteins that control the membrane trafficking and endocytosis of TGFβ receptors play a role in regulating the intensity and duration of TGFβ signals. For example, the efficient regulation of Smad signaling can be facilitated by adaptor proteins such as the Smad cytosolic cofactor SARA (Smad anchor for receptor activation, also known as ZFYVE9). SARA contains a Smad-binding domain, as well as a TGFβ receptor complex-interacting region, and it acts as a bridge, facilitating R-Smad presentation to the activated receptor complex (Tsukazaki et al., 1998; Wu et al., 2000). SARA contains a phosphatidylinositol 3-phosphate (PI3P)-binding FYVE domain, which links it to the early endosome, implicating receptor endocytosis and trafficking in efficient Smad signal transduction (Di Guglielmo et al., 2003; Tsukazaki et al., 1998). Once SARA-bound R-Smads are phosphorylated, they dissociate from the SARA–receptor complex, bind to Smad4 and accumulate in the nucleus to regulate transcription (Xu et al., 2000). Interestingly, although receptor endocytosis has been reported to be dispensable for the phosphorylation of R-Smads, it has been reported that endocytosis is required for the efficient dissociation of R-Smads from SARA, nuclear accumulation and subsequent transcriptional response (Runyan et al., 2005). The precise regulation of transcriptional activity of Smads in the nucleus is important for the proper execution of embryonic development by controlling tissue patterning and normal organ development, and it is also important for controlling cellular growth and apoptotic response in adult tissues; deviations are associated with various pathologies (Derynck et al., 2001; Heldin et al., 2009; Pardali and Moustakas, 2007; Shi and Massagué, 2003; Siegel and Massagué, 2003).

Although it is established that Smads are central regulators of gene response to TGFβ, multiple Smad-independent pathways are also initiated upon TGFβ receptor activation (Derynck and Zhang, 2003; Heldin et al., 2009; Massagué, 2003; Moustakas and Heldin, 2005; Pardali and Moustakas, 2007). In addition to Smads, TGFβ can also activate the mitogen-associated protein kinase family (MAPK). There are three principle classes of MAPK proteins – ERK, JNK, and p38 – each of which has a complex but apparent role in the development and progression of cancer (Wagner and Nebreda, 2009). The p38 MAPK pathway downstream of TGFβ has gained considerable interest as a pathway that regulates apoptosis. Briefly, TGFβ receptor activation leads to the recruitment and Lys63-linked autoubiquitylation and activation of TRAF6 (TNF-receptor-associated factor 6), an E3 ubiquitin ligase. This stimulates a cascade that culminates in the activation of p38 MAPK and, ultimately, apoptosis of various cell types (Edlund et al., 2003; Sorrentino et al., 2008; Yamashita et al., 2008). Interestingly, atypical protein kinase C (aPKC) isoforms interact with TRAF6 to mediate cytokine signaling (Sanz et al., 2000), but less is known about the involvement of aPKC in modulating the TGFβ–p38 MAPK pathway.

We have previously shown that aPKC isoforms can alter TGFβ signaling patterns in non-small cell lung cancer (NSCLC) cells by altering receptor trafficking, degrading specific receptor complexes and by enhancing Par6-dependent phosphorylation (Gunaratne et al., 2012; Gunaratne et al., 2013). However, we had not examined gene changes on a large scale. Furthermore, we have not examined whether aPKC isoforms alter TGFβ-induced MAPK pathways. The aPKC isoforms, which consist of PKCι and PKCζ, are a subset of the protein kinase C family that are Ca2+ and diacylglycerol (DAG)-independent (Griner and Kazanietz, 2007). Importantly, the aPKCs show altered expression and activities in various cancers (Huang and Muthuswamy, 2010), and PKCι has been described as an oncogene (Fields and Regala, 2007; Regala et al., 2005). Interestingly, aPKC isoforms are known to play a role in p38-MAPK-induced apoptosis, as inhibition or knockdown of aPKC sensitizes glioblastoma cells to chemotherapeutic agents through a p38-dependent mechanism (Baldwin et al., 2006).

In this report, we examine gene changes in aPKC-silenced cells by microarray analyses, and examine how knockdown of aPKC alters Smad and MAPK signaling pathways and stimulates TGFβ-dependent apoptosis of NSCLC cells.

Knockdown of aPKC isoforms alters TGFβ-induced gene expression

We reported previously that aPKC gene silencing using small interfering RNA (siRNA) temporally extended TGFβ-induced Smad2 phosphorylation (Gunaratne et al., 2012) and inhibited Par6-dependent EMT (Gunaratne et al., 2013). In order to assess the effects of aPKC silencing on TGFβ-dependent transcription, we silenced PKCι and PKCζ in combination (PKCι/ζ) using siRNA and conducted microarray analysis. Table 1 summarizes fold change differences between siControl- and siPKCι/ζ-treated cells following TGFβ induction as determined by microarray analysis. This list was selected from a set of genes commonly known to be regulated by TGFβ (Siegel and Massagué, 2003). The full microarray dataset can be accessed online at the NCBI Gene Expression Omnibus website (GEO; GSE26241). We observed that several classical TGFβ-regulated genes had similar expression patterns between control and aPKC-silenced cells, including BMP4, SNAI1 and SNAI2. However, there were several genes that displayed a reduced TGFβ-dependent response in aPKC-silenced cells compared to that of control cells, including IL1A, SMURF2, MMP2 and MMP9 (shown in bold). In order to assess gene changes in further detail, we investigated several transcripts using real-time PCR (qPCR).

Table 1.
aPKC knockdown alters TGFβ gene response as shown by microarray analysis
graphic
graphic

A549 cells transfected with control (siControl) or siRNA directed against aPKC isoforms (siPKCι/ζ) were serum starved and treated with 250 pM TGFβ for 1 h, washed and further incubated for 24 h in low-serum medium. Total RNA was then extracted and subjected to gene expression array analysis. Shown is a selected list of gene responses in control and aPKC-silenced cells after 24 h for several genes regulated by TGFβ (list adapted from Siegel and Massagué, 2003). Fold change comparisons are expressed relative to untreated siControl cells (siControl, −TGFβ) and represent the average of three separate experiments (n = 3). In bold are genes that exhibited a reduced response in aPKC-knockdown cells.

Knockdown of aPKC isoforms alters TGFβ-induced gene expression in qPCR analysis

We first assessed the expression of PKCι, PKCζ and PKCα to confirm that our siRNA approach was efficient and specific, and we observed that siRNA treatment was successful in reducing the gene expression of the atypical PKCs (aPKCι and ζ) but not the classical PKCα (Fig. 1A). We next assessed the transcription of known TGFβ-dependent genes in response to TGFβ in control and aPKC-knockdown cells at 4 and 24 h after stimulation with TGFβ. Several genes showed a significantly reduced induction by TGFβ in aPKC-silenced cells at 4 and 24 h after ligand-mediated stimulation. These included SMURF2 (Fig. 1B) in A549 cells (but not in H1299 cells; supplementary material Fig. S1A), SERPINE1 (also known as PAI-1; Fig. 1C; supplementary material Fig. S1B) and MMP9 (Fig. 1D; supplementary material Fig. S1C). We also observed cases in which the TGFβ-dependent response was either partial (SNAI1; Fig. 1E; supplementary material Fig. S1D) or was similar in both aPKC-knockdown and control cells (CDH1; Fig. 1F). Finally, KLF10 (also known as TIEG1) was unresponsive to TGFβ in both treatment groups (data not shown). Taken together, our microarray and qPCR data suggest that TGFβ-dependent transcription is muted in aPKC-knockdown cells, despite the fact that these cells exhibited extended Smad2 phosphorylation in response to TGFβ induction (Gunaratne et al., 2012; supplementary material Fig. S2).

Fig. 1.

aPKC silencing alters TGFβ-dependent gene induction. Real-time PCR analysis of TGFβ-induced mRNA levels in A549 control siRNA cells versus cells with knockdown of both PKCι and PKCζ (siPKCι/ζ). RNA extracts were isolated from cells treated for 1 h with TGFβ followed by 4-h or 24-h incubation in the absence of ligand. Two-way ANOVA analysis followed by post-hoc Bonferonni's tests were used to determine statistical significance of gene expression changes of PKCι, PKCζ and PKCα (A), SMURF2 (B), SERPINE1 (PAI-1; C), MMP-9 (D), SNAI1 (Snail; E) and CDH1 (E-cadherin; F). All data show the mean±s.e.m.; *P≤0.05; **P≤0.01.

Fig. 1.

aPKC silencing alters TGFβ-dependent gene induction. Real-time PCR analysis of TGFβ-induced mRNA levels in A549 control siRNA cells versus cells with knockdown of both PKCι and PKCζ (siPKCι/ζ). RNA extracts were isolated from cells treated for 1 h with TGFβ followed by 4-h or 24-h incubation in the absence of ligand. Two-way ANOVA analysis followed by post-hoc Bonferonni's tests were used to determine statistical significance of gene expression changes of PKCι, PKCζ and PKCα (A), SMURF2 (B), SERPINE1 (PAI-1; C), MMP-9 (D), SNAI1 (Snail; E) and CDH1 (E-cadherin; F). All data show the mean±s.e.m.; *P≤0.05; **P≤0.01.

Knockdown of aPKC increases cytosolic retention of Smad2 by SARA

Activated TGFβ receptors phosphorylate receptor-regulated Smads (Smad2 and Smad3) on a C-terminal SSXS motif, which facilitates their dissociation from SARA, association with Smad4 and accumulation in the nucleus to modulate transcription (Shi and Massagué, 2003). Therefore, the reduced TGFβ-dependent transcriptional response that we observed in aPKC-silenced cells might be due to a reduced nuclear translocation of Smad2. The subcellular localization of R-Smads can be controlled by a balance between binding factors that retain them in the cytoplasm versus transcription factors that retain them in the nucleus. One such cytoplasmic retention factor is SARA, a protein enriched on early endosomes (Itoh et al., 2002; Tsukazaki et al., 1998).

SARA preferentially binds to the non-C-terminally phosphorylated form of Smad2, and it is thought that the activated receptor complex formed at the plasma membrane is captured by SARA in the early endosome, which then presents the bound R-Smad to the receptor for phosphorylation (Massagué and Chen, 2000). Smad2 then dissociates from SARA and associates with Smad4 prior to nuclear translocation and the initiation of transcription (Massagué and Chen, 2000). Furthermore, although phosphorylated R-Smads can activate transcription alone, a full TGFβ response requires complex formation by Smad2 and Smad4 (Levy and Hill, 2005; Wrana, 2009). To test this, we immunoprecipitated Smad2 from control and aPKC-silenced cells treated with TGFβ to examine whether SARA would dissociate from Smad2 upon TGFβ addition. TGFβ addition reduced SARA association with Smad2 and resulted in a concomitant increase in binding to Smad4 (Fig. 2A). Interestingly, in aPKC-silenced cells, TGFβ addition reduced SARA–Smad2 dissociation as well as the binding of Smad2 to Smad4, suggesting a deficit in this exchange. This indicated that SARA might be retaining Smad2 in the cytoplasm to a greater degree in aPKC-silenced cells, and might reduce nuclear translocation of Smad2. Because we have previously reported alterations in the membrane trafficking of TGFβ receptors upon PKC inhibition (Gunaratne et al., 2012), we next examined whether SARA levels were altered in aPKC-silenced cells. Interestingly, aPKC-silenced cells showed significantly increased total protein levels of SARA compared with control cells, although no appreciable changes were observed with TGFβ addition (Fig. 2B). We next examined whether increased SARA expression might disrupt its localization to the early endosome, which, in turn, might disrupt normal Smad signaling in aPKC-silenced cells. Using immunofluorescence microscopy, we did not detect any appreciable alteration of SARA colocalization with EEA-1 (an early endosome marker), suggesting that SARA still accessed the early endosome in aPKC-silenced cells (Fig. 2C). We next analyzed whether the increased SARA levels in aPKC-depleted cells could be retaining Smad2 in the cytoplasm.

Fig. 2.

aPKC knockdown alters TGFβ-induced SARA–Smad2–Smad4 interactions. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were serum starved and treated with or without 250 pM TGFβ for 1 h prior to lysis. Cell lysates were then immunoprecipitated (IP) using anti (α)-Smad2 antibodies and subjected to SDS-PAGE and immunoblotting using anti-SARA, anti-Smad4 and anti-Smad2 antibodies. IgG heavy chain is indicated. Cell lysates were included to show relative endogenous protein expression. Densitometric analysis of Smad2-associated SARA or Smad4 levels from three independent replicate experiments are shown. Data show the mean±s.e.m (n = 3); *P≤0.05; **P≤0.01 (two-way ANOVA). (B) A549 cells transfected with control siRNA or siRNA targeting aPKC isoforms were lysed and immunoblotted using antibodies against SARA and actin as indicated on the right of the panels. Densitometric analysis of steady-state SARA levels from three independent experiments is shown graphically to the right of the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05 (two-way ANOVA). (C) aPKC knockdown does not inhibit the localization of SARA in the early endosome. A549 cells were transfected as described in A and processed for immunofluorescence microscopy to visualize EEA1 (green) and SARA (red). DAPI was used to visualize DNA (blue). The boxed areas are shown at higher magnification in the lower images. Representative images from at least three independent experiments are shown. Scale bars: 10 µm.

Fig. 2.

aPKC knockdown alters TGFβ-induced SARA–Smad2–Smad4 interactions. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were serum starved and treated with or without 250 pM TGFβ for 1 h prior to lysis. Cell lysates were then immunoprecipitated (IP) using anti (α)-Smad2 antibodies and subjected to SDS-PAGE and immunoblotting using anti-SARA, anti-Smad4 and anti-Smad2 antibodies. IgG heavy chain is indicated. Cell lysates were included to show relative endogenous protein expression. Densitometric analysis of Smad2-associated SARA or Smad4 levels from three independent replicate experiments are shown. Data show the mean±s.e.m (n = 3); *P≤0.05; **P≤0.01 (two-way ANOVA). (B) A549 cells transfected with control siRNA or siRNA targeting aPKC isoforms were lysed and immunoblotted using antibodies against SARA and actin as indicated on the right of the panels. Densitometric analysis of steady-state SARA levels from three independent experiments is shown graphically to the right of the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05 (two-way ANOVA). (C) aPKC knockdown does not inhibit the localization of SARA in the early endosome. A549 cells were transfected as described in A and processed for immunofluorescence microscopy to visualize EEA1 (green) and SARA (red). DAPI was used to visualize DNA (blue). The boxed areas are shown at higher magnification in the lower images. Representative images from at least three independent experiments are shown. Scale bars: 10 µm.

aPKC knockdown reduces TGFβ-induced Smad2 nuclear accumulation

To examine TGFβ-dependent nuclear translocation of Smad2 in control and aPKC-silenced cells, we carried out immunofluorescence microscopy analysis (Fig. 3A). As expected, in cells transfected with control siRNA, TGFβ induced an increase in Smad2 nuclear staining, suggesting nuclear accumulation of Smad2 (Fig. 3A). Consistent with the microarray and qPCR data, aPKC-silenced cells showed reduced nuclear accumulation of Smad2 in response to TGFβ (Fig. 3A). The reduction in Smad nuclear translocation in aPKC-silenced cells was also observed for another TGFβ receptor R-Smad, Smad3, and also occurred in another NSCLC cell line, H1299 NSCLC cells (supplementary material Fig. S3A,B). Because we observed an increase in SARA expression and association with Smad2 in aPKC-silenced cells (Fig. 2), we next assessed whether reducing SARA levels would reverse the cytoplasmic retention of Smad2 in aPKC-silenced cells. To investigate this possibility, we first silenced SARA using siRNA and observed no differences in Smad2 phosphorylation (supplementary material Fig. S2C) or nuclear accumulation in A549 cells (Fig. 3A). Interestingly, concomitant siRNA targeting of aPKC isoforms and SARA partially restored the nuclear accumulation of Smad2 that was absent in cells with knockdown of aPKC only (Fig. 3A).

Fig. 3.

aPKC silencing reduces TGFβ-induced Smad2 nuclear accumulation. (A) A549 cells were transfected with the indicated siRNA, serum starved and treated with 250 pM TGFβ for 1 h. The cells were processed for immunofluorescence microscopy with antibodies against Smad2. DAPI was used to visualize DNA. Representative images from at least three independent replicate experiments are shown. Scale bars: 10 µm. (B) A549 cells were transfected and treated with TGFβ as described in A. The cells were then subjected to subcellular fractionation to isolate cytoplasmic and nuclear fractions. The fractions were subjected to SDS-PAGE and immunoblotted using anti (α)-Smad2, anti-tubulin and anti-histone H3 antibodies to determine the subcellular distribution of Smad2. Histone H3 and tubulin antibodies were used as loading controls for the nuclear and cytoplasmic fractions, respectively. Average nuclear Smad2 levels from three independent replicate experiments were quantified and the data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05 (two-way ANOVA).

Fig. 3.

aPKC silencing reduces TGFβ-induced Smad2 nuclear accumulation. (A) A549 cells were transfected with the indicated siRNA, serum starved and treated with 250 pM TGFβ for 1 h. The cells were processed for immunofluorescence microscopy with antibodies against Smad2. DAPI was used to visualize DNA. Representative images from at least three independent replicate experiments are shown. Scale bars: 10 µm. (B) A549 cells were transfected and treated with TGFβ as described in A. The cells were then subjected to subcellular fractionation to isolate cytoplasmic and nuclear fractions. The fractions were subjected to SDS-PAGE and immunoblotted using anti (α)-Smad2, anti-tubulin and anti-histone H3 antibodies to determine the subcellular distribution of Smad2. Histone H3 and tubulin antibodies were used as loading controls for the nuclear and cytoplasmic fractions, respectively. Average nuclear Smad2 levels from three independent replicate experiments were quantified and the data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05 (two-way ANOVA).

To verify and quantify the observation of reduced nuclear Smad accumulation, we also conducted subcellular fractionation studies and immunoblotting of cellular cytosolic and nuclear fractions when cells were treated in the presence or absence of TGFβ (Fig. 3B). Consistent with our immunofluorescence microscopy analysis, TGFβ treatment stimulated an increase in nuclear Smad2 levels in cells transfected with control siRNA. In contrast, aPKC-knockdown cells contained significantly reduced nuclear Smad2 levels upon TGFβ stimulation (Fig. 3B). The amount of TGFβ-dependent nuclear accumulation of Smad2 in SARA-silenced cells was similar to that of control cells (Fig. 3B). However, consistent with the immunofluorescence analysis, the silencing of aPKC isoforms and SARA partially restored the accumulation of Smad2 in aPKC-silenced cells (Fig. 3B).

Taken together, these results suggest that an increase in SARA protein in the cytoplasm of aPKC-silenced cells might be inhibiting Smad2 from translocating to the nucleus after TGFβ stimulation, and possibly altering transcriptional activity. However, the results do not exclude the possibility that other signaling pathways might be altering Smad function upon aPKC knockdown. We therefore examined whether aPKC knockdown would affect MAPK pathways, as these pathways have been shown to crosstalk with the Smad pathway and have been reported to alter R-Smad nuclear targeting.

Knockdown of aPKC enhances phosphorylated p38 MAPK levels

Smads shuttle to and from the nucleus, and their subcellular localization is primarily controlled through phosphorylation events. MAPK pathways are known to crosstalk with Smads through the phosphorylation of the Smad linker region, which can alter Smad localization and function, including nuclear exclusion (Massagué, 2003; Kretzschmar et al., 1999). Given our observation of reduced nuclear accumulation of Smad2/3 in aPKC-silenced cells, we next assessed whether MAPK pathways were altered in aPKC-silenced cells. We analyzed the levels of the three activated MAPK pathways in response to TGFβ in control and aPKC-silenced cells (Fig. 4). Interestingly, aPKC knockdown increased basal and TGFβ-induced levels of phosphorylated p38 MAPK at both 1 h and 24 h time-points, whereas no appreciable differences were observed for phosphorylated ERK or phosphorylated JNK (Fig. 4A).

Fig. 4.

aPKC knockdown increases and temporally extends phosphorylated p38 MAPK levels in response to TGFβ. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were treated with or without 250 pM TGFβ for 1 or 24 h prior to lysis. Samples were then processed for SDS-PAGE and immunoblotted with anti (α)-phospho-specific antibodies directed against phosphorylated (P-) forms of ERK, p38 and JNK as indicated on the right of the panels. Shown are representative immunoblots from at least three independent replicate experiments. Immunoblotting for actin was performed as a loading control. (B) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 30 min, washed and further incubated for 1 or 4 h prior to lysis. Lysates were then processed for SDS-PAGE and immunoblotted with the anti-phospho-specific p38 and total p38 MAPK antibodies as indicated on the right of the panels. Phosphorylated p38 MAPK levels from three independent replicate experiments were quantified, and the data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05; **P≤0.01 (two-way ANOVA).

Fig. 4.

aPKC knockdown increases and temporally extends phosphorylated p38 MAPK levels in response to TGFβ. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were treated with or without 250 pM TGFβ for 1 or 24 h prior to lysis. Samples were then processed for SDS-PAGE and immunoblotted with anti (α)-phospho-specific antibodies directed against phosphorylated (P-) forms of ERK, p38 and JNK as indicated on the right of the panels. Shown are representative immunoblots from at least three independent replicate experiments. Immunoblotting for actin was performed as a loading control. (B) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 30 min, washed and further incubated for 1 or 4 h prior to lysis. Lysates were then processed for SDS-PAGE and immunoblotted with the anti-phospho-specific p38 and total p38 MAPK antibodies as indicated on the right of the panels. Phosphorylated p38 MAPK levels from three independent replicate experiments were quantified, and the data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); *P≤0.05; **P≤0.01 (two-way ANOVA).

We then assessed shorter timecourses of p38 MAPK activation in control and aPKC-knockdown cells and further observed that TGFβ-induced p38 MAPK phosphorylation was increased and extended in aPKC-silenced cells at 0.5, 1.5 and 4.5 h after TGFβ treatment (Fig. 4B). Because MAPK crosstalk has been reported to alter Smad nuclear–cytoplasmic shuttling dynamics (Burch et al., 2010; Kamato et al., 2013; Massagué, 2003), we reasoned that increased p38 MAPK activity might be reducing Smad2 nuclear import in aPKC-silenced cells. When we tested this hypothesis, we observed that p38 MAPK inhibition did not rescue Smad2 nuclear accumulation in aPKC-silenced cells (supplementary material Fig. S3C). This result suggested that p38 MAPK activity might not be responsible for the reduced Smad2 nuclear accumulation observed in aPKC-silenced cells, and that the Smad2 and p38 MAPK pathways might be independent of each other.

Knockdown of aPKC increases TGFβ-induced apoptotic response through p38 MAPK

TGFβ receptors can activate the p38 MAPK pathway to stimulate apoptosis (Derynck and Zhang, 2003; Yu et al., 2002). We therefore examined whether the increased p38 MAPK signaling observed in aPKC-silenced cells sensitized cells to TGFβ-induced apoptotic response. To measure apoptosis, control and aPKC-silenced cells were treated with or without TGFβ for 48 h, and the level of apoptosis was measured through the assessment of nuclear morphology after Hoescht staining (Fig. 5A). Cells treated with control siRNA showed a modest apoptotic response to TGFβ. This was in contrast to aPKC-silenced cells, which exhibited a significant increase in cell death when treated with TGFβ (Fig. 5A). Importantly, aPKC-silenced cells treated with a p38 MAPK inhibitor showed a reduced number of apoptotic nuclei, indicating that the apoptotic response observed was downstream of p38 MAPK signaling (Fig. 5A). We observed similar results using another NSCLC cell line, H1299 cells (supplementary material Fig. S4A,B). TGFβ-induced cleaved-Parp levels (c-Parp), a marker of apoptosis, were significantly higher in aPKC-silenced cells than in control cells, and c-Parp levels were reduced with the p38 inhibitor (Fig. 5B), although the p38 inhibitor had no effect on Smad2 phosphorylation levels (supplementary material Fig. S4C). These results further highlight the importance of p38 signaling in this apoptotic response.

Fig. 5.

aPKC knockdown enhances TGFβ-induced apoptotic response. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were serum deprived and treated with or without 250 pM TGFβ for 48 h in the presence or absence of a p38 MAPK inhibitor. Hoescht 33342 was used to stain the nuclei of cells prior to image acquisition and cell counting. Scale bars: 10 µm. Quantification of apoptotic nuclei (yellow arrowheads) from four independent experiments is expressed graphically below the representative images. Data show the mean±s.e.m. (n = 4); *P≤0.05 (two-way ANOVA). (B) A549 cells were treated as in A and then lysed. Cell lysates were processed for SDS-PAGE and immunoblotted with anti (α)-cleaved (c-)PARP and anti-actin antibodies. Densitometric analysis from four independent replicate experiments is shown graphically below the representative immunoblot. Data show the mean±s.e.m. (n = 4); *P≤0.05 (two-way ANOVA).

Fig. 5.

aPKC knockdown enhances TGFβ-induced apoptotic response. (A) A549 cells transfected with control siRNA (siControl) or siRNA directed against the aPKC isoforms (PKCι/ζ) were serum deprived and treated with or without 250 pM TGFβ for 48 h in the presence or absence of a p38 MAPK inhibitor. Hoescht 33342 was used to stain the nuclei of cells prior to image acquisition and cell counting. Scale bars: 10 µm. Quantification of apoptotic nuclei (yellow arrowheads) from four independent experiments is expressed graphically below the representative images. Data show the mean±s.e.m. (n = 4); *P≤0.05 (two-way ANOVA). (B) A549 cells were treated as in A and then lysed. Cell lysates were processed for SDS-PAGE and immunoblotted with anti (α)-cleaved (c-)PARP and anti-actin antibodies. Densitometric analysis from four independent replicate experiments is shown graphically below the representative immunoblot. Data show the mean±s.e.m. (n = 4); *P≤0.05 (two-way ANOVA).

aPKC knockdown stabilizes TGFβ-induced p38 MAPK signaling through TRAF6

TGFβ-stimulated apoptosis through p38 MAPK has been previously reported to occur through the recruitment and activation of the E3 ubiquitin ligase TRAF6 (Sorrentino et al., 2008; Yamashita et al., 2008). Briefly, upon TGFβ activation, TRAF6 is recruited to TβRI of the TGFβ receptor complex. This causes TRAF6 to become auto-ubiquitylated, which activates TAK1 (also known as MAP3K7; a MAP3K), which triggers the MAPK cascade to p38 activation (Sorrentino et al., 2008; Yamashita et al., 2008). We have shown previously that aPKC expression can alter the binding and degradation patterns of TβRI and its substrates (Gunaratne et al., 2012; Gunaratne et al., 2013). We suspected that the increased TGFβ–p38 MAPK signals that we observed in aPKC-silenced cells might be due to increased levels of TRAF6 and TGFβ receptor complexes when aPKC was depleted. We tested this by immunoprecipitating endogenous TRAF6 from control and aPKC-silenced cells, followed by immunoblotting for TβRI (Fig. 6A). Interestingly, TβRI associated to a greater degree with TRAF6 in the absence of aPKC expression (Fig. 6A). This finding suggested that TβRI–TRAF6 complexes were more stable in aPKC-knockdown cells. We reasoned that this increase in TβRI–TRAF6 complexes was responsible for the increased TGFβ-induced p38 MAPK signals in aPKC-silenced cells. To test this, we next examined whether siRNA-mediated TRAF6 knockdown could abrogate p38 MAPK signaling in aPKC-silenced cells. We used siRNA to knock down the aPKCs (siPKCι/ζ), TRAF6 alone (siTRAF6) or aPKC and TRAF6 together (siPKCι/ζ+TRAF6) (Fig. 6B). As we had observed before, knockdown of aPKC enhanced TGFβ-induced phosphorylated p38 MAPK levels (Fig. 6B, lanes 3 and 4). Interestingly, in cells where aPKC and TRAF6 were knocked down simultaneously, TGFβ-induced p38 MAPK phosphorylation was abrogated (Fig. 6B, lanes 7 and 8). Furthermore, the p38 signaling patterns were reflective of the level of apoptotic stimulation, as TRAF6 siRNA also reduced the TGFβ-induced apoptotic response seen in aPKC-silenced cells (Fig. 6C). These results suggested that the enhanced p38 MAPK signaling and apoptosis that we had observed in aPKC-silenced cells were TRAF6 dependent.

Fig. 6.

aPKC knockdown enhances TGFβ-induced p38 MAPK signaling and apoptotic response through TRAF6. (A) HEK 293T cells transfected with control siRNA (siControl) or siRNA targeting aPKC isoforms (PKCι/ζ) were co-transfected with cDNA encoding Flag-tagged TGFβ type 1 receptor (FlagTβRI) as indicated. Cells were then lysed, and endogenous TRAF6 was immunoprecipitated (IP) using anti-TRAF6 antibodies. The immunoprecipitates were processed for SDS-PAGE and immunoblotted with anti (α)-Flag and anti-TRAF6 antibodies to visualize immunoprecipitated Flag-tagged TβRI and TRAF6 (upper panel). Cell lysates were immunoblotted with anti-PKCι, anti-PKCζ, anti-Flag and anti-TRAF6 antibodies to visualize endogenous aPKC and TRAF6 levels as well as expressed Flag-tagged TβRI (lower panel). Representative immunoblots from at least from three independent replicate experiments are shown. (B) A549 cells transfected with control siRNA or with siRNA directed against aPKC (siPKCι/ζ), TRAF6 (siTRAF6) or both aPKC and TRAF6 (siPKCι/ζ+TRAF6) were serum starved and treated with 250 pM TGFβ for 1 h prior to lysis. Lysates were then processed for SDS-PAGE and immunoblotted with the anti-phospho-specific p38 and total p38 MAPK antibodies as indicated on the right of the panels. Immunoblotting using anti-TRAF6, anti-PKCι and anti-PKCζ antibodies was used to determine knockdown levels. Phosphorylated p38 MAPK levels from three independent replicate experiments were quantified by densitometric analysis and data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); **P≤0.01 (two-way ANOVA). (C) A549 cells transfected as in B were serum deprived and treated with or without 250 pM TGFβ for 48 h. Hoescht 33342 was then used to stain the nuclei of cells prior to image acquisition and cell counting. Scale bar: 10 µm. Quantification of apoptotic nuclei (yellow arrowheads) from three independent experiments is shown below the representative images. Data show the mean±s.e.m. (n = 3); **P≤0.01 (two-way ANOVA).

Fig. 6.

aPKC knockdown enhances TGFβ-induced p38 MAPK signaling and apoptotic response through TRAF6. (A) HEK 293T cells transfected with control siRNA (siControl) or siRNA targeting aPKC isoforms (PKCι/ζ) were co-transfected with cDNA encoding Flag-tagged TGFβ type 1 receptor (FlagTβRI) as indicated. Cells were then lysed, and endogenous TRAF6 was immunoprecipitated (IP) using anti-TRAF6 antibodies. The immunoprecipitates were processed for SDS-PAGE and immunoblotted with anti (α)-Flag and anti-TRAF6 antibodies to visualize immunoprecipitated Flag-tagged TβRI and TRAF6 (upper panel). Cell lysates were immunoblotted with anti-PKCι, anti-PKCζ, anti-Flag and anti-TRAF6 antibodies to visualize endogenous aPKC and TRAF6 levels as well as expressed Flag-tagged TβRI (lower panel). Representative immunoblots from at least from three independent replicate experiments are shown. (B) A549 cells transfected with control siRNA or with siRNA directed against aPKC (siPKCι/ζ), TRAF6 (siTRAF6) or both aPKC and TRAF6 (siPKCι/ζ+TRAF6) were serum starved and treated with 250 pM TGFβ for 1 h prior to lysis. Lysates were then processed for SDS-PAGE and immunoblotted with the anti-phospho-specific p38 and total p38 MAPK antibodies as indicated on the right of the panels. Immunoblotting using anti-TRAF6, anti-PKCι and anti-PKCζ antibodies was used to determine knockdown levels. Phosphorylated p38 MAPK levels from three independent replicate experiments were quantified by densitometric analysis and data are shown below the representative immunoblots. Data show the mean±s.e.m. (n = 3); **P≤0.01 (two-way ANOVA). (C) A549 cells transfected as in B were serum deprived and treated with or without 250 pM TGFβ for 48 h. Hoescht 33342 was then used to stain the nuclei of cells prior to image acquisition and cell counting. Scale bar: 10 µm. Quantification of apoptotic nuclei (yellow arrowheads) from three independent experiments is shown below the representative images. Data show the mean±s.e.m. (n = 3); **P≤0.01 (two-way ANOVA).

TRAF6 silencing partially inhibits TGFβ-dependent gene transcription

Finally, given that aPKC knockdown increased TRAF6-dependent p38 MAPK activity and decreased TGFβ-dependent gene transcription, we assessed whether the two observations were linked, i.e. whether TRAF6 modulation would alter TGFβ-dependent gene transcription. To do this, we performed qPCR analysis of SMURF2, SERPINE1, MMP9, CDH-1 and SNAI-1 mRNA levels in control and TRAF6-silenced cells following TGFβ stimulation for 4 and 24 h (Fig. 7). Unlike our previous results regarding SMURF2 mRNA levels in control versus aPKC-silenced cells (Fig. 1B), we observed no differences in SMURF2 transcript levels between control and siTRAF6-treated cells (Fig. 7A). However, consistent with the results observed with aPKC silencing (Fig. 1), TRAF6 silencing decreased TGFβ-dependent SERPINE1, MMP9 and SNAI1 levels (Fig. 7A). Interestingly, the reduction of gene transcription in TRAF6-silenced cells was independent of Smad2 phosphorylation or nuclear accumulation (Fig. 7B,C). Taken together, our results suggest that aPKC isoforms not only regulate TGFβ-dependent gene regulation but also direct NSCLC cells towards EMT versus apoptotic responses using multiple signaling pathways.

Fig. 7.

TRAF6 silencing alters TGFβ-dependent gene induction but not phosphorylation or nuclear accumulation of Smad2. (A) Real-time PCR analysis of TGFβ-induced mRNA levels in A549 control siRNA cells (siControl) versus TRAF6-silenced cells (siTRAF6). RNA was isolated from cells treated for 1 h with TGFβ followed by 4 or 24 h of incubation in the absence of ligand. Two-way ANOVA analysis followed by post-hoc Bonferonni's test was used to determine statistical significance of gene expression changes of SMURF2, SERPINE1 (PAI-1), MMP-9, SNAI1 (Snail) and CDH1 (E-cadherin). Data show the mean±s.e.m. *P≤0.05, **P≤0.01. (B) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 1 h. Lysates were then processed for SDS-PAGE and immunoblotted with anti (α)-phosphorylation (P-) specific Smad2, Smad2/3 or TRAF6 antibodies. (C) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 1 h. The cells were then subjected to subcellular fractionation to isolate cytoplasmic and nuclear fractions. The fractions were subjected to SDS-PAGE and immunoblotted using anti-Smad2, anti-tubulin and anti-histone H3 antibodies to determine the subcellular distribution of Smad2. Histone H3 and tubulin antibodies were used as loading controls for the nuclear and cytoplasmic fractions, respectively. Representative blots from three experiments are shown.

Fig. 7.

TRAF6 silencing alters TGFβ-dependent gene induction but not phosphorylation or nuclear accumulation of Smad2. (A) Real-time PCR analysis of TGFβ-induced mRNA levels in A549 control siRNA cells (siControl) versus TRAF6-silenced cells (siTRAF6). RNA was isolated from cells treated for 1 h with TGFβ followed by 4 or 24 h of incubation in the absence of ligand. Two-way ANOVA analysis followed by post-hoc Bonferonni's test was used to determine statistical significance of gene expression changes of SMURF2, SERPINE1 (PAI-1), MMP-9, SNAI1 (Snail) and CDH1 (E-cadherin). Data show the mean±s.e.m. *P≤0.05, **P≤0.01. (B) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 1 h. Lysates were then processed for SDS-PAGE and immunoblotted with anti (α)-phosphorylation (P-) specific Smad2, Smad2/3 or TRAF6 antibodies. (C) A549 cells transfected with the indicated siRNA were serum starved and treated with 250 pM TGFβ for 1 h. The cells were then subjected to subcellular fractionation to isolate cytoplasmic and nuclear fractions. The fractions were subjected to SDS-PAGE and immunoblotted using anti-Smad2, anti-tubulin and anti-histone H3 antibodies to determine the subcellular distribution of Smad2. Histone H3 and tubulin antibodies were used as loading controls for the nuclear and cytoplasmic fractions, respectively. Representative blots from three experiments are shown.

TGFβ pathways regulate many developmental and homeostatic processes, and aberrant signaling is associated with various pathologies such as fibrosis and cancer. Here, we have found that aPKC knockdown alters both Smad-dependent and the Smad-independent p38 MAPK signaling pathways and regulates whether NSCLC cells undergo TGFβ-dependent apoptosis.

In this report, we examined the transcriptional changes associated with TGFβ signaling in an aPKC-silenced background. In this context, we found that several TGFβ-stimulated genes showed reduced transcriptional activity and this was a result of reduced Smad2 nuclear accumulation. Furthermore, we found that the knockdown of aPKC increased the basal protein levels of SARA. This is an important finding, as it has been reported previously that increased SARA expression is associated with reduced TGFβ receptor degradation (Di Guglielmo et al., 2003), consistent with our previous findings that aPKC knockdown reduces TGFβ receptor degradation (Gunaratne et al., 2012). Moreover, increased SARA expression is associated with the maintenance of epithelial cell phenotype (Runyan et al., 2009), consistent with our findings that aPKC knockdown reduces TGFβ-induced EMT (Gunaratne et al., 2013). Given our previous finding that aPKC alters membrane trafficking of the TGFβ receptors (Gunaratne et al., 2012), it would be interesting to explore whether aPKC alters the function or localization of receptor-bound SARA to control the context under which Smads are signaling. Interestingly, Runyan and colleagues showed that although inhibiting TGFβ receptor internalization from the membrane only slightly altered phosphorylated Smad2 levels, it did significantly impact on the ability of Smad2 to dissociate from SARA (Runyan et al., 2005). Moreover, Smad2–Smad4 complex formation has also been reported to occur in the early endosome (Chen et al., 2007). This suggests that the coordinated function and subcellular localization of SARA and associated Smads are important for mediating TGFβ-dependent transcription properly.

A recent report has implicated SARA in general endocytic processes through classical ESCRT complex machinery (Kostaras et al., 2013). More specifically, the correct subcellular trafficking of the EGFR from the early endosome to late endosomes to regulate EGFR degradation was dependent on SARA, implicating SARA with a more general role in endocytic trafficking than was appreciated previously (Kostaras et al., 2013). This might have important implications with respect to our findings that aPKC knockdown reduces TGFβ receptor degradation and stabilizes particular TGFβ receptor–protein complexes (Gunaratne et al., 2012; Gunaratne et al., 2013). One possibility is that depletion of aPKC leads to an accumulation of SARA and TGFβ receptor complexes. Indeed, aPKC has been reported previously to be involved in the trafficking of membrane proteins, as well as being involved in the passage of EGFR to lysosome-targeted endosomes through the anchoring protein p62 (also known as SQSTM1) (Sanchez et al., 1998). Whether the knockdown of aPKC in our model is causing a reduced passage of receptors to lysosomes is an important area for future study.

Here, we made the novel finding that the knockdown of aPKC increases and extends TGFβ-induced p38 MAPK activation, which sensitizes NSCLC cells to undergo apoptosis. We found that knockdown of aPKC stabilized TβRI–TRAF6 complexes, and that knockdown of TRAF6 in aPKC-silenced cells returned p38 MAPK activation back to control levels. In line with our results, increases in p38 MAPK activity have been reported before upon aPKC silencing (Baldwin et al., 2006), indicating that aPKC might be attenuating p38 MAPK signaling in multiple tumor cell types. Interestingly, when aPKC is knocked down, p38 MAPK is able to elicit an apoptotic response, indicating that in some situations aPKC might be a viable therapeutic target. However, the role of p38 MAPK in cancer is also complex, and context dependent, and in addition to sensitizing cells to a death response, p38 activity is also associated with cancer cell survival and both the stimulation and suppression of EMT (Bakin et al., 2002; Strippoli et al., 2010; Wagner and Nebreda, 2009). We also report here that the enhanced p38 MAPK activation we observed in aPKC-silenced cells was not responsible for the reduction in Smad2 nuclear accumulation. The role of Smad2 linker phosphorylation by MAPK members in TGFβ signaling has yielded mixed results. The original reports show that linker phosphorylation by MAPK blocked Smad2 nuclear accumulation (Grimm and Gurdon, 2002; Kretzschmar et al., 1999); however, nuclear stabilization of Smad2 by linker phosphorylation has also been reported (Alarcón et al., 2009; Burch et al., 2010), suggesting that Smad linker phosphorylation is more complex than originally thought, and requires further examination.

In conclusion, we have found that aPKC plays multiple roles in TGFβ signaling, and the localization and expression patterns of aPKC might dictate how a cell responds to TGFβ. This is especially important given that aPKC isoforms have recently been implicated in cancer progression (Huang and Muthuswamy, 2010; Kojima et al., 2008) and aPKCι has been classified as a human oncogene (Fields and Regala, 2007; Murray et al., 2011). Whether aPKC might be a viable therapeutic target in TGFβ-driven tumor progression remains to be examined.

Antibodies and Reagents

Primary antibodies were as follows: anti-β-actin (Sigma, A2668), anti-PKCι (BD Transduction Laboratories, 610175), anti-PKCζ (Cell Signaling Technology, 9372), anti-phospho-Smad2 (Cell Signaling Technology, 3101), anti-Smad2/3 (BD Transduction Laboratories, 610842), anti-tubulin (Sigma, T4026), anti-H3-histone (Millipore, 05-499), anti- phospho-p38 (Cell Signaling Technology, 9211), anti-p38 (Cell Signaling Technology, 9212), anti phospho-ERK (Cell Signaling Technology, 4370), anti-phospho-JNK (Cell Signaling Technology, 9255s), anti-Smad4 (Abcam, AB40759), anti-SARA (Santa Cruz Biotechnology, sc-9135), anti-Flag (Sigma Aldrich, F3165), anti-Traf6 (Cell Signaling Technology, 8028s), anti-EEA1 (BD Transduction Laboratories, 610457). Horseradish-peroxidase-conjugated secondary goat anti-rabbit-IgG (Thermo Scientific, 31460) and goat anti-mouse-IgG (Thermo Scientific, 31430) were used for immunoblot analysis. Fluorescently conjugated donkey anti-mouse-IgG (Life Technologies, A21206) and donkey anti-rabbit-IgG (Life Technologies, A31572) were used for immunofluorescence studies. Human siRNA constructs were purchased from Life Technologies (siPKCζ, siPKCι and siControl catalog numbers were 10620319-HSS183348, 10620319-HSS183318 and 4390844, respectively). TRAF6 siRNA was purchased from Life Technologies (product number s14389- 4390824). p38 MAPK inhibitor was purchased from Calbiochem (506126).

Cell culture and transfections

A549 and H1299 NSCLC cell lines were maintained in F12K and RPMI-1640 medium, respectively, supplemented with 10% fetal bovine serum. Cells were kept in a humidified tissue culture incubator at 37°C under 5% CO2. siRNA and DNA transfections were conducted using Lipofectamine RNAi max and Lipofectamine LTX (Life Technologies) according to the manufacturer's protocol. TGFβ treatments (250 pM) were conducted in low-serum medium (0.2% FBS) for the indicated times after cells were serum deprived overnight.

Immunoblotting and immunoprecipitation

Cells were lysed (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 0.5% Triton X-100, 1 mM phenylmethylsulfonyl fluoride and a mixture of protease inhibitors) and centrifuged at 20,000 g at 4°C for 10 min. Aliquots of supernatants were collected for analysis of total protein concentration. Protein concentrations were determined using the Lowry method (Fisher). Immunoprecipitations and immunoblotting were conducted as described previously (Gunaratne et al., 2012; Gunaratne et al., 2013).

Cellular fractionation

Cytoplasmic and nuclear cellular fractions were isolated using the Thermo Scientific Kit NE-PER® kit (78833) according to the manufacturer's protocols.

Immunofluorescence microscopy

Cells were fixed with 4% paraformaldehyde, permeabilized with 0.25% Triton X-100, and incubated with primary antibodies at 4°C overnight. Following incubation with the appropriate fluorescent-probe-conjugated secondary antibodies, the probes were visualized by immunofluorescence microscopy using an inverted IX81 Microscope (Olympus, Canada).

RNA quality assessment, probe preparation and GeneChip hybridization

To conduct the microarray analyses, siRNA-treated cells (siControl and siPKCι/ζ double knockdown) were treated with TGFβ for 1 h, followed by washout and further incubation of cells for 24 h in low-serum medium. Total RNA was extracted and processed for microarray analysis as described below.

RNA and GeneChips were processed at the London Regional Genomics Centre (Robarts Research Institute, London, Ontario, Canada; http://www.lrgc.ca) as described previously (Guo et al., 2011). Using Partek, any batch effect due to scan date was removed and an ANOVA (Yijk = μ+Condition×Timeij+eijk) using Method of Moments (Eisenhart, 1947) was run to determine gene-level P-values. Fold change comparisons are expressed relative to untreated siControl cells and represent the average of three separate experiments (three separate GeneChips per condition). A fold change of ±1.6 was considered as the cutoff for induction.

Reverse transcription and qPCR

RNA extraction, reverse transcription and real-time PCR were conducted as described previously (Gunaratne et al., 2013). Primer sequences are shown in supplementary material Table S1. Gene expression in each treatment is expressed relative to the control (siControl, no TGFβ) and is an average of three to six independent experimental trials.

Cell death assays

A549 and H1299 cells transfected with the appropriate siRNA constructs were serum deprived (0.2% FBS) and then incubated with or without TGFβ in low-serum medium in the presence or absence of a p38 MAPK (1 µM) inhibitor for 48 h. After 48 h, apoptosis of A549 and H1299 cells was analyzed by examining nuclear morphology after Hoechst 33342 staining. Hoescht stain (1 µg/ml) was added directly to the medium and samples were incubated for 30 min at 37°C. The cells were then visualized using a fluorescent microscope (Olympus IX71), and ten random images were acquired per condition. Normal and apoptotic nuclei were counted and the apoptotic nuclei (characterized by condensed chromatin) were scored as a proportion of normal healthy cells.

Statistical analysis

One-way or two-way ANOVA analyses followed by post-hoc Bonferonni's test were used to evaluate the significance of the results. Statistical analyses were performed using GraphPad Prism® Software 5.0 and P-values of <0.05 were considered to be statistically significant.

The authors would like to thank the members of the Di Guglielmo laboratory for advice and support.

Author contributions

A.G. conceived and carried out the majority of the work presented in the manuscript. E.C. performed and analyzed the nuclear accumulation of Smad2 and D.C. carried out the microarray analysis. T.H.E.-C. assisted in the acquisition of the phosphorylated p38 and apoptosis data. G.M.D.G. supervised the studies, helped design the overall experimental approach and prepared the final manuscript.

Funding

The work carried out in this study was supported by the Canadian Institutes of Health Research [grant number MOP-93625 to G.M.D.G.].

Alarcón
C.
,
Zaromytidou
A. I.
,
Xi
Q.
,
Gao
S.
,
Yu
J.
,
Fujisawa
S.
,
Barlas
A.
,
Miller
A. N.
,
Manova-Todorova
K.
,
Macias
M. J.
et al. (
2009
).
Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-beta pathways.
Cell
139
,
757
769
.
Attisano
L.
,
Wrana
J. L.
(
2002
).
Signal transduction by the TGF-beta superfamily.
Science
296
,
1646
1647
.
Bakin
A. V.
,
Rinehart
C.
,
Tomlinson
A. K.
,
Arteaga
C. L.
(
2002
).
p38 mitogen-activated protein kinase is required for TGFbeta-mediated fibroblastic transdifferentiation and cell migration.
J. Cell Sci.
115
,
3193
3206
.
Baldwin
R. M.
,
Garratt-Lalonde
M.
,
Parolin
D. A.
,
Krzyzanowski
P. M.
,
Andrade
M. A.
,
Lorimer
I. A.
(
2006
).
Protection of glioblastoma cells from cisplatin cytotoxicity via protein kinase Ciota-mediated attenuation of p38 MAP kinase signaling.
Oncogene
25
,
2909
2919
.
Burch
M. L.
,
Yang
S. N.
,
Ballinger
M. L.
,
Getachew
R.
,
Osman
N.
,
Little
P. J.
(
2010
).
TGF-beta stimulates biglycan synthesis via p38 and ERK phosphorylation of the linker region of Smad2.
Cell. Mol. Life Sci.
67
,
2077
2090
.
Chen
Y. G.
,
Wang
Z.
,
Ma
J.
,
Zhang
L.
,
Lu
Z.
(
2007
).
Endofin, a FYVE domain protein, interacts with Smad4 and facilitates transforming growth factor-beta signaling.
J. Biol. Chem.
282
,
9688
9695
.
Derynck
R.
,
Akhurst
R. J.
(
2007
).
Differentiation plasticity regulated by TGF-beta family proteins in development and disease.
Nat. Cell Biol.
9
,
1000
1004
.
Derynck
R.
,
Zhang
Y. E.
(
2003
).
Smad-dependent and Smad-independent pathways in TGF-beta family signalling.
Nature
425
,
577
584
.
Derynck
R.
,
Akhurst
R. J.
,
Balmain
A.
(
2001
).
TGF-beta signaling in tumor suppression and cancer progression.
Nat. Genet.
29
,
117
129
.
Di Guglielmo
G. M.
,
Le Roy
C.
,
Goodfellow
A. F.
,
Wrana
J. L.
(
2003
).
Distinct endocytic pathways regulate TGF-beta receptor signalling and turnover.
Nat. Cell Biol.
5
,
410
421
.
Edlund
S.
,
Bu
S.
,
Schuster
N.
,
Aspenström
P.
,
Heuchel
R.
,
Heldin
N. E.
,
ten Dijke
P.
,
Heldin
C. H.
,
Landström
M.
(
2003
).
Transforming growth factor-beta1 (TGF-beta)-induced apoptosis of prostate cancer cells involves Smad7-dependent activation of p38 by TGF-beta-activated kinase 1 and mitogen-activated protein kinase kinase 3.
Mol. Biol. Cell
14
,
529
544
.
Eisenhart
C.
(
1947
).
The assumptions underlying the analysis of variance.
Biometrics
3
,
1
21
.
Elliott
R. L.
,
Blobe
G. C.
(
2005
).
Role of transforming growth factor Beta in human cancer.
J. Clin. Oncol.
23
,
2078
2093
.
Fields
A. P.
,
Regala
R. P.
(
2007
).
Protein kinase C iota: human oncogene, prognostic marker and therapeutic target.
Pharmacol. Res.
55
,
487
497
.
Grimm
O. H.
,
Gurdon
J. B.
(
2002
).
Nuclear exclusion of Smad2 is a mechanism leading to loss of competence.
Nat. Cell Biol.
4
,
519
522
.
Griner
E. M.
,
Kazanietz
M. G.
(
2007
).
Protein kinase C and other diacylglycerol effectors in cancer.
Nat. Rev. Cancer
7
,
281
294
.
Gunaratne
A.
,
Benchabane
H.
,
Di Guglielmo
G. M.
(
2012
).
Regulation of TGFβ receptor trafficking and signaling by atypical protein kinase C. Cell.
Signal.
24
,
119
130
.
Gunaratne
A.
,
Thai
B. L.
,
Di Guglielmo
G. M.
(
2013
).
Atypical protein kinase C phosphorylates Par6 and facilitates transforming growth factor β-induced epithelial-to-mesenchymal transition.
Mol. Cell. Biol.
33
,
874
886
.
Guo
F.
,
Carter
D. E.
,
Leask
A.
(
2011
).
Mechanical tension increases CCN2/CTGF expression and proliferation in gingival fibroblasts via a TGFβ-dependent mechanism.
PLoS ONE
6
,
e19756
.
Heldin
C. H.
,
Landström
M.
,
Moustakas
A.
(
2009
).
Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition.
Curr. Opin. Cell Biol.
21
,
166
176
.
Huang
L.
,
Muthuswamy
S. K.
(
2010
).
Polarity protein alterations in carcinoma: a focus on emerging roles for polarity regulators.
Curr. Opin. Genet. Dev.
20
,
41
50
.
Itoh
F.
,
Divecha
N.
,
Brocks
L.
,
Oomen
L.
,
Janssen
H.
,
Calafat
J.
,
Itoh
S.
,
Dijke Pt
P.
(
2002
).
The FYVE domain in Smad anchor for receptor activation (SARA) is sufficient for localization of SARA in early endosomes and regulates TGF-beta/Smad signalling.
Genes Cells
7
,
321
331
.
Kamato
D.
,
Burch
M. L.
,
Piva
T. J.
,
Rezaei
H. B.
,
Rostam
M. A.
,
Xu
S.
,
Zheng
W.
,
Little
P. J.
,
Osman
N.
(
2013
).
Transforming growth factor-β signalling: role and consequences of Smad linker region phosphorylation.
Cell. Signal.
25
,
2017
2024
.
Kojima
Y.
,
Akimoto
K.
,
Nagashima
Y.
,
Ishiguro
H.
,
Shirai
S.
,
Chishima
T.
,
Ichikawa
Y.
,
Ishikawa
T.
,
Sasaki
T.
,
Kubota
Y.
et al. (
2008
).
The overexpression and altered localization of the atypical protein kinase C lambda/iota in breast cancer correlates with the pathologic type of these tumors.
Hum. Pathol.
39
,
824
831
.
Kostaras
E.
,
Sflomos
G.
,
Pedersen
N. M.
,
Stenmark
H.
,
Fotsis
T.
,
Murphy
C.
(
2013
).
SARA and RNF11 interact with each other and ESCRT-0 core proteins and regulate degradative EGFR trafficking.
Oncogene
32
,
5220
5232
.
Kretzschmar
M.
,
Doody
J.
,
Timokhina
I.
,
Massagué
J.
(
1999
).
A mechanism of repression of TGFbeta/ Smad signaling by oncogenic Ras.
Genes Dev.
13
,
804
816
.
Levy
L.
,
Hill
C. S.
(
2005
).
Smad4 dependency defines two classes of transforming growth factor beta (TGF-beta) target genes and distinguishes TGF-beta-induced epithelial-mesenchymal transition from its antiproliferative and migratory responses.
Mol. Cell. Biol.
25
,
8108
8125
.
Massagué
J.
(
1992
).
Receptors for the TGF-beta family.
Cell
69
,
1067
1070
.
Massagué
J.
(
1998
).
TGF-beta signal transduction.
Annu. Rev. Biochem.
67
,
753
791
.
Massagué
J.
(
2003
).
Integration of Smad and MAPK pathways: a link and a linker revisited.
Genes Dev.
17
,
2993
2997
.
Massagué
J.
,
Chen
Y. G.
(
2000
).
Controlling TGF-beta signaling.
Genes Dev.
14
,
627
644
.
Moustakas
A.
,
Heldin
C. H.
(
2005
).
Non-Smad TGF-beta signals.
J. Cell Sci.
118
,
3573
3584
.
Murray
N. R.
,
Kalari
K. R.
,
Fields
A. P.
(
2011
).
Protein kinase Cι expression and oncogenic signaling mechanisms in cancer.
J. Cell. Physiol.
226
,
879
887
.
Pardali
K.
,
Moustakas
A.
(
2007
).
Actions of TGF-beta as tumor suppressor and pro-metastatic factor in human cancer.
Biochim. Biophys. Acta
1775
,
21
62
.
Regala
R. P.
,
Weems
C.
,
Jamieson
L.
,
Khoor
A.
,
Edell
E. S.
,
Lohse
C. M.
,
Fields
A. P.
(
2005
).
Atypical protein kinase C iota is an oncogene in human non-small cell lung cancer.
Cancer Res.
65
,
8905
8911
.
Runyan
C. E.
,
Schnaper
H. W.
,
Poncelet
A. C.
(
2005
).
The role of internalization in transforming growth factor beta1-induced Smad2 association with Smad anchor for receptor activation (SARA) and Smad2-dependent signaling in human mesangial cells.
J. Biol. Chem.
280
,
8300
8308
.
Runyan
C. E.
,
Hayashida
T.
,
Hubchak
S.
,
Curley
J. F.
,
Schnaper
H. W.
(
2009
).
Role of SARA (SMAD anchor for receptor activation) in maintenance of epithelial cell phenotype.
J. Biol. Chem.
284
,
25181
25189
.
Sanchez
P.
,
De Carcer
G.
,
Sandoval
I. V.
,
Moscat
J.
,
Diaz-Meco
M. T.
(
1998
).
Localization of atypical protein kinase C isoforms into lysosome-targeted endosomes through interaction with p62.
Mol. Cell. Biol.
18
,
3069
3080
.
Sanz
L.
,
Diaz-Meco
M. T.
,
Nakano
H.
,
Moscat
J.
(
2000
).
The atypical PKC-interacting protein p62 channels NF-kappaB activation by the IL-1-TRAF6 pathway.
EMBO J.
19
,
1576
1586
.
Shi
Y.
,
Massagué
J.
(
2003
).
Mechanisms of TGF-beta signaling from cell membrane to the nucleus.
Cell
113
,
685
700
.
Siegel
P. M.
,
Massagué
J.
(
2003
).
Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer.
Nat. Rev. Cancer
3
,
807
820
.
Sorrentino
A.
,
Thakur
N.
,
Grimsby
S.
,
Marcusson
A.
,
von Bulow
V.
,
Schuster
N.
,
Zhang
S.
,
Heldin
C. H.
,
Landström
M.
(
2008
).
The type I TGF-beta receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent manner.
Nat. Cell Biol.
10
,
1199
1207
.
Strippoli
R.
,
Benedicto
I.
,
Foronda
M.
,
Perez-Lozano
M. L.
,
Sánchez-Perales
S.
,
López-Cabrera
M.
,
Del Pozo
M. A.
(
2010
).
p38 maintains E-cadherin expression by modulating TAK1-NF-kappa B during epithelial-to-mesenchymal transition.
J. Cell Sci.
123
,
4321
4331
.
Tsukazaki
T.
,
Chiang
T. A.
,
Davison
A. F.
,
Attisano
L.
,
Wrana
J. L.
(
1998
).
SARA, a FYVE domain protein that recruits Smad2 to the TGFbeta receptor.
Cell
95
,
779
791
.
Wagner
E. F.
,
Nebreda
A. R.
(
2009
).
Signal integration by JNK and p38 MAPK pathways in cancer development.
Nat. Rev. Cancer
9
,
537
549
.
Wrana
J. L.
(
2009
).
The secret life of Smad4.
Cell
136
,
13
14
.
Wu
G.
,
Chen
Y. G.
,
Ozdamar
B.
,
Gyuricza
C. A.
,
Chong
P. A.
,
Wrana
J. L.
,
Massagué
J.
,
Shi
Y.
(
2000
).
Structural basis of Smad2 recognition by the Smad anchor for receptor activation.
Science
287
,
92
97
.
Xu
L.
,
Chen
Y. G.
,
Massagué
J.
(
2000
).
The nuclear import function of Smad2 is masked by SARA and unmasked by TGFbeta-dependent phosphorylation.
Nat. Cell Biol.
2
,
261
267
.
Yamashita
M.
,
Fatyol
K.
,
Jin
C.
,
Wang
X.
,
Liu
Z.
,
Zhang
Y. E.
(
2008
).
TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta.
Mol. Cell
31
,
918
924
.
Yu
L.
,
Hébert
M. C.
,
Zhang
Y. E.
(
2002
).
TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses.
EMBO J.
21
,
3749
3759
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information