All EMBO Press journals Open Access as of 1 January 2024 - read the FAQs

Article
15 December 2005
Free access

Novel roles of Akt and mTOR in suppressing TGF‐β/ALK5‐mediated Smad3 activation

The EMBO Journal
(2005)
25: 58 - 69
Insulin‐like growth factor‐I inhibits transforming growth factor‐β (TGF‐β) signaling by blocking activation of Smad3 (S3), via a phosphatidylinositol 3‐kinase (PI3K)/Akt‐dependent pathway. Here we provide the first report that the kinase activity of Akt is necessary for its ability to suppress many TGF‐β responses, including S3 activation and induction of apoptosis. Wild‐type and myristoylated Akts (AktWT and AktMyr) suppress TGF‐β‐induced phospho‐activation of S3 but not Smad2 (S2), whereas kinase‐dead Akt1 (Akt1K179M) or dominant‐negative PI3K enhances TGF‐β‐induced phospho‐activation of both S2 and S3. Using siRNA, rapamycin (Rap), and adenoviral expression for FKBP12‐resistant and constitutively active TGF‐β type I receptor (ALK5), we demonstrate that mammalian target of Rap (mTOR) mediates Akt1 suppression of phospho‐activation of S3. These and further data on Akt1‐S3 binding do not support a recently proposed model that Akt blocks S3 activation through physical interaction and sequestration of S3 from TGF‐β receptors. We propose a novel model whereby Akt suppresses activation of S3 in an Akt kinase‐dependent manner through mTOR, a likely route for loss of tumor suppression by TGF‐β in cancers.

Introduction

Transforming growth factor‐β (TGF‐β) is a 25 kDa multifunctional autocrine/paracrine growth regulator belonging to the large TGF‐β superfamily (Roberts and Sporn, 1990; Massague, 1992), and may function as either a tumor suppressor in normal or preneoplastic epithelia or a tumor promoter in a variety of late‐stage carcinomas (Guo and Kyprianou, 1999; Tang et al, 1999; Wakefield and Roberts, 2002; Song et al, 2003a; Danielpour, 2005). TGF‐β1 signals mainly through two cell surface signaling receptors, TGF‐β receptor type I (TβRI) and TβRII, whereby binding of ligand to TβRII promotes receptor heteromerization (Massague, 1992; ten Dijke et al, 1996), allowing the TβRII kinase to activate TβRI (Wieser et al, 1995). TβRI then activates S2 and S3 by phosphorylating their C‐terminal SSXS serines (Abdollah et al, 1997), a process shown to involve accessory proteins (Tsukazaki et al, 1998). This causes the receptor‐activated Smads to multimerize (Wu et al, 1997) and then translocate to the nucleus, where they activate gene transcription (Xiao et al, 2000).
Akt, which relays signals downstream of phosphatidylinositol 3‐kinase (PI3K), is emerging as a central player in the tumorigenesis and pathogenesis of a variety of human cancers (Vivanco and Sawyers, 2002). Akt activation is a multistep process, involving both membrane translocation and phosphorylation. PI3K, which is commonly activated by receptor tyrosine kinases, catalyzes the addition of a phosphate moiety to the D3 position of phosphatidylinositol‐4‐phosphate. This generates phosphatidylinositol 3,4‐diphosphate, which is necessary for the membrane anchor of Akt and PDK1, two interacting PH domain proteins. PDK1 phosphorylates Akt at Thr308, whereas a yet unidentified kinase (PDK2) or a mammalian target of Rap (mTOR) complex phosphorylates Akt at Ser473, leading to full activation of Akt (Downward, 1998; Nicholson and Anderson, 2002; Sarbassov dos et al, 2005). Recent studies have unveiled a growing list of Akt substrates (Nicholson and Anderson, 2002) that cooperate to prevent apoptosis and/or promote cell proliferation (Plas and Thompson, 2003; Xu et al, 2004).
We previously reported that physiological levels of insulin‐like growth factor‐I (IGF‐I) function through a PI3K‐dependent pathway to block several TGF‐β‐mediated responses, including gene transcription, apoptosis, and Smad3 (S3) activation (Song et al, 2003b; Danielpour, 2005), using the well‐established NRP‐152 non‐tumorigenic rat prostate epithelial cell line model developed in our laboratory (Danielpour et al, 1994; Danielpour, 1996). The LR3 analog of IGF‐I (LR3‐IGF‐I), which binds poorly to IGF‐I binding proteins, blocks TGF‐β responses by suppressing the phospho‐activation of S3 independent of changes in TGF‐β receptor expression or changes in the expression of Smad(S)s 2, 3, or 4 (Song et al, 2003b). Recently, two other groups also reported that Akt suppresses TGF‐β responses, through a mechanism involving the direct binding of Akt to S3, which blocks activation of S3 by sequestering S3 from TβRI (Conery et al, 2004; Remy et al, 2004). In the current report, we show a different mechanism of Akt suppression of S3, involving the Akt target mTOR.
mTOR is an in vivo target for the complex of rapamycin (Rap) with its intracellular receptor, FKBP12 (Fingar and Blenis, 2004). As a member of the PIK‐related family of large protein kinases, mTOR controls the phosphorylation of at least two regulators of protein synthesis and cell growth, S6 kinase 1 (S6K) and eIF‐4E binding protein (4E‐BP1) (Bjornsti and Houghton, 2004; Fingar and Blenis, 2004). Along with PI3K/Akt axis, mTOR pathway is emerging as a pivotal regulator of cell growth in response to hormones, nutrition, and growth factors. PI3K‐dependent signaling has been implicated in the regulation of mTOR and S6K, and Akt‐dependent phosphorylation has also been reported to result in the phosphorylation of common downstream target proteins (Gao et al, 2002; Manning and Cantley, 2003; Plas and Thompson, 2003; Tee et al, 2003; Fingar and Blenis, 2004).
In our model, Akt blocks phospho‐activation of S3 by an Akt kinase‐dependent mechanism through mTOR and also blocks TGF‐β signals downstream of S3 activation, but through a mechanism that does not require the kinase activity of Akt or mTOR. This is the first direct evidence for roles of Akt kinase and mTOR as suppressors of S3 phospho‐activation.

Results

IGF‐I, which is strongly implicated in the pathogenesis of prostate cancer (Grimberg, 2003; Renehan et al, 2004), is critical to survival and growth of NRP‐152 cells, a non‐tumorigenic rat prostatic epithelial cell line established in our laboratory (Danielpour et al, 1994; Hsing et al, 1996; Danielpour, 1999; Hayward et al, 1999). Similar to normal prostate epithelium, NRP‐152 cells are exquisitely sensitive to the induction of apoptosis by TGF‐β (Hsing et al, 1996; Stewart et al, 2003). Moreover, dominant‐negative (DN)‐TβRII promotes malignant transformation of these cells, supporting a role of TGF‐β in tumor suppression of the prostate (Tang et al, 1999). Importantly, IGF‐I blocks the ability of TGF‐β to induce their apoptosis. We have recently reported that IGF‐I specifically blocks TGF‐β‐induced phospho‐activation of Smad3 (S3) but not Smad2 (S2), and we have confirmed roles for PI3K and Akt in mediating this IGF‐I effect (Song et al, 2003b). We now show that this effect does not require de novo protein synthesis, as cycloheximide does not block the ability of IGF‐I to suppress TGF‐β‐induced phospho‐S3 (Supplementary Figure 1S).
We have further analyzed the mechanism by which Akt1 kinase inhibits TGF‐β responses, using the NRP‐152 cell model. We first compared the abilities of wild‐type (WT), constitutively active (CA) myristoylated (Myr; N‐terminal fusion with src aa 1–11), and kinase‐dead (KD) (K179M mutant) Akt1 constructs to control transcriptional responses by TGF‐β1. These cells were transiently co‐transfected with the above expression constructs along with a plasminogen activator inhibitor‐I (PAI‐I) promoter reporter construct, 3TP‐lux (Figure 1A and B). Enforced expression of both Akt1WT and Akt1Myr inhibited TGF‐β‐induced PAI‐I promoter activity by about two‐ and seven‐fold, respectively, whereas Akt1K179M, which functions as a DN of Akt kinase, instead slightly enhanced the response to TGF‐β (Figure 1A). Increased expression of Akt1K179M by greater transfection efficiency more effectively enhanced TGF‐β‐induced 3TP‐lux activity (Figure 1B), similar to DN‐PI3K (Song et al, 2003b), indicating that the Akt1 kinase is required for suppression of this promoter activity by Akt1. Suppression of TGF‐β‐induced 3TP‐lux promoter activity also occurred by the two other Akt isoforms (Figure 1C). Consistent with the dependence on Akt kinase activity, the PI3K inhibitor LY294002 (LY) reversed the suppression of TGF‐β‐induced 3TP‐lux by either Akt1WT or Akt1Myr (Figure 1D). Rap, an mTOR inhibitor, which we previously used to reverse the IGF‐I suppression of TGF‐β‐induced 3TP‐lux and S3 activation in NRP‐152 cells (Song et al, 2003b), also reversed Akt1 suppression of TGF‐β‐induced 3TP‐lux (Figure 1D). Together, these results support a role for the kinase domain of Akt1 in the suppression of TGF‐β responses.
image
Figure 1. Akt kinase activity is essential for suppression of TGF‐β signaling by Akt in NRP‐152, NRP‐154, and Mv1Lu cells. (A–F) NRP‐152 cells were transiently transfected in 12‐well plates with 1 μg of DNA, which included 3TP‐lux, CMV‐renilla plasmids, and either pUSE empty vector, pUSE‐Myc‐Akt1s (WT, Myr, or kinase‐dead (K179M)) (A, B, D), pCDNA3 empty vector, or pCDNA3‐Myc‐Akts 1–3 (C), followed by treatment with LY (10 μM) or Rap (200 nM) (D). Expression of Myc‐Akt1s and phospho‐p70S6 kinase (Thr389) is shown in panel D. Separately, cells were transfected with Akt1WT, Akt1Myr, Akt1WT mutants (K179M, T308A, S473A) (E), or Akt1Myr mutants (Myr/K179M, Myr/T308A, Myr/S473A, Myr/T308A/S473A) (F). Expression of these Akt mutants and their kinase activity were confirmed by immunoblotting for Myc and phospho‐GSK‐3α/β (Ser21/9), respectively. (G, H) NRP‐154 (G) and Mv1Lu (H) were co‐transfected with pUSE empty vector, pUSE‐Akt1Myr, or pUSE‐Akt1Myr/K179M. For the Mv1Lu cells, IGF‐I or insulin pretreatments were added after transfection (H). Cells were then incubated overnight and cultured for an additional 24 h in the presence or absence of TGF‐β1 (10 ng/ml) before assay. A dual luciferase assay was performed, and data shown are averages (±s.d.) of triplicate independent measurements of Firefly/Renilla luciferase readings normalized to untreated controls. Results are representative of two to three different experiments (A–H).
To further confirm that the kinase activity of Akt1WT and Akt1Myr is necessary for the suppression of TGF‐β‐induced 3TP‐lux promoter activity, we developed additional constructs of Akt1WT and Akt1Myr with mutations at the kinase domain (K179M) and/or phosphorylation sites (T308A and S473A). Akt1WT mutated at K179M or at T308A (Akt1K179M or Akt1T308A, respectively) were unable to block TGF‐β‐induced 3TP‐lux, whereas the S473A mutant (Akt1S473A), similar to Akt1WT, suppressed 3TP‐lux activity induced by TGF‐β1 (Figure 1E). Akt1Myr constructs mutated at K179M (Akt1Myr/K179M) or T308A (Akt1Myr/T308A) were kinase‐inactive (as measured by phosphorylation of an Akt1 substrate, GSK‐3α/β) and were unable to suppress TGF‐β‐induced 3TP‐lux, whereas the S473A form (Akt1Myr/S473A) was just as active as Akt1Myr in suppressing the induction of 3TP‐lux by TGF‐β and phosphorylating GSK‐3α/β (Figure 1F). Akt1Myr/T308A/S473A was biologically indistinguishable from Akt1Myr/T308A, indicating that S473 does not contribute significantly to suppression of this TGF‐β response or phosphorylation of GSK‐3α/β. Importantly, Akt1K179M, Akt1Myr/K179M, and Akt1Myr/T308A significantly enhanced this TGF‐β activity (Figure 1E–G and Supplementary Figure 2S), in contrast to that of a previous report, where the ‘kinase‐dead’ form of AktMyr was functionally indistinguishable from kinase‐active AktMyr in Hep3B cells (Conery et al, 2004). Our observations that the kinase activity of Akt is required for suppressing this TGF‐β response extend to other highly TGF‐β responsive cell lines, such as NRP‐154 prostatic line (Figure 1G) and Mv1Lu mink lung line (Figure 1H). Together, our data provide strong support that the kinase activity of Akt1 is critical for its suppression of TGF‐β responses.
To study the individual roles of PI3K and Akt1 in suppressing the phospho‐activation of S3, we used adenoviral constructs for efficient and rapid delivery of Akt1WT, Akt1Myr, Akt1K179M, DN‐PI3K, or CA‐PI3K in NRP‐152 cells. Akt1Myr, CA‐PI3K, LR3‐IGF‐I, and, to a lesser extent, Akt1WT all suppressed the TGF‐β1 activation of S3 but not of S2, whereas DN‐PI3K and Akt1K179M substantially enhanced both phospho‐S3 and phospho‐S2 levels (Figure 2A–D). The expression levels of total Akt1, phospho‐Akt1 (Ser473), phospho‐Akt1 (Thr308), and phospho‐GSK‐3α/β (Ser21/9) were as expected for each treatment. None of these constructs altered expression of total S2, S3, and S4. The enhanced phospho‐activation of S2 by either DN‐PI3K or Akt1K179M was unexpected, as Akts did not suppress phospho‐S2 levels even at early TGF‐β1 treatment times (Figure 2B and C). Neither DN‐PI3K nor Akt1K179M alone induced phosphorylation of S3 or S2 (Figure 2D). These results showed that activation of either PI3K or Akt alone is sufficient for suppression of S3 activation, and that the kinase activity of Akt is essential for such suppression. Moreover, S2 is also regulated by the Akt kinase, but likely through a mechanism or stoichiometry different from that of S3. Collectively, these data strongly support that the Akt suppression of the phospho‐activation of S3 and possibly that of S2 occurs downstream of the Akt kinase.
image
Figure 2. Akt abolishes TGF‐β‐induced S3 phosphorylation and apoptosis and reverses growth arrest in NRP‐152 cells. (A) Cells were infected with control virus, Admax‐Myc‐tagged Akt1 (Akt1WT, Akt1Myr, Akt1K179M), Admax‐DN‐PI3K, or Admax‐Myc‐CA‐PI3K, and incubated overnight in the presence or absence of 2 nM LR3‐IGF‐I, followed by treatment with 10 ng/ml of TGF‐β1 for 4 h. (B, C) The ability of Akt1Myr to block the phospho‐activation of S2 and S3 at various time points after TGF‐β1 addition was examined. (D) Effect of Akt1K179M or DN‐PI3K alone or together with TGF‐β1 on phospho‐activation of S2 and S3. (E) TGF‐β1 failed to downregulate cyclin D2 expression in NRP‐152 cells overexpressing Akt1Myr or CA‐PI3K. Whole‐cell lysates (50 μg protein) were subjected to Western blot analysis (A–E). (F) Cells were infected with control virus, Admax‐Myc‐Akt1Myr, or Admax‐Myc‐CA‐PI3K for 2 h and incubated overnight followed by 48 h with TGF‐β1. Cell numbers were determined with a Coulter Counter. Data shown are averages (±s.d.) of triplicate independent measurements. (G) The intranucleosomal DNA fragmentation induced in NRP‐152 cells by 24 h of TGF‐β1 treatment was blocked by preinfection with adenovirus delivering Akt1Myr or CA‐PI3K, but not by Akt1K179M. (H) Cell cycle analysis was performed on NRP‐152 cells infected with control virus or Admax‐Myc‐Akt1Myr following treatment with vehicle or TGF‐β1 for 48 h. Cells fixed with 90% methanol were stained with propidium iodide and analyzed by flow cytometry, based on a reference count of 20000 cells at G1/G0. The distributions of cycling cells (S+G2/M), undergoing DNA synthesis (S), and dead cells (sub‐G1) are shown. Results are representative of two to three experiments per treatment.
Adenoviral‐mediated gene delivery was also used to assess the biological end points of PI3K and Akt on TGF‐β1‐induced growth suppression and apoptosis. Impressively, CA‐PI3K and Akt1Myr blocked TGF‐β1‐induced downregulation of cyclin D2 (Figure 2E), cell death (Figure 2F), and apoptosis (by intranucleosomal DNA fragmentation; Figure 2G), whereas Akt1K179M was ineffective in suppressing the above responses of TGF‐β1 (Figure 2). Rather, Akt1K179M alone killed cells, especially >24 h of infection, consistent with Akt kinase being antiapoptotic (Yamaguchi and Wang, 2001; Jetzt et al, 2003). The ability of Akt to reverse the suppression of cyclin D2 expression by TGF‐β suggests that Akt also reverses growth suppression by TGF‐β. We used flow cytometry to analyze how Akt affected TGF‐β responses on cell cycle distribution. However, under the above growth conditions, NRP‐152 cells are essentially growth arrested (G1/G0), and these cells require both high serum and insulin (⩾1 μM) or IGF‐I (⩾2 nM) to proliferate (Hsing et al, 1996). Thus, to test the effect of Akt on TGF‐β‐induced growth suppression, NRP‐152 cells were cultured in GM3 containing 5% fetal bovine serum (FBS). Under these conditions, only 8% of the viable cells were in G2/M+S. TGF‐β suppressed the G2/M+S population by 60%, whereas Akt1Myr partially reversed this suppression (52% reversal at S; 40% reversal of G2/M+S) and the TGF‐β‐induced sub‐G1 (35% reversal) (Figure 2H). The above results are consistent with our previous report that IGF‐I suppresses TGF‐β responses downstream of Akt (Song et al, 2003b).
We next determined whether the ability of the Akt1 to suppress TGF‐β1 responses is limited to inhibition of S3 activation or also acts downstream of active S3. To address the latter possibility, we studied the ability of Akt1Myr or CA‐PI3K to suppress 3TP‐lux activity induced by CA‐S3 (C‐terminal DDVD; Chipuk et al, 2002) versus that induced by TGF‐β1 or by transfection of S3WT. CA‐PI3K suppressed 3TP‐lux activity induced by TGF‐β1 or to a lesser extent by CA‐S3 (Figure 3A and B). In confirmation of the biological significance of these results, we showed that IGF‐I similarly inhibits the activation of 3TP‐lux by CA‐S3, but not by S3WT (not activated by TGF‐β) (Figure 3C). Overexpressed S3WT activates transcription by nuclear import of S3 without its phospho‐activation. Thus, our data suggest that Akt1Myr or CA‐PI3K blocks not only phospho‐activation of S3 but also the biological responses of S3 downstream of this activation. However, neither LY nor Rap reversed Akt suppression of CA‐S3, as indicated by both 3TP‐lux and SBE4‐luc reporters (Figure 3D and E). This contrasts with the reversal of Akt's effect on the above response by TGF‐β1 (Figure 1), suggesting that Akt suppresses CA‐S3 by an Akt kinase‐independent mechanism. To test this, we compared the effects of Akt1K179M, Akt1Myr/K179M, and Akt1Myr on 3TP‐lux by CA‐S3 (Figure 3F). Whereas the kinase‐dead Akt mutants did not suppress this activity of TGF‐β1 (Figure 1), they suppressed that of CA‐S3 (Figure 3F). In contrast, kinase‐active or kinase‐dead Akts did not suppress 3TP‐luciferase activated by transfection of S3WT (Figure 3G), which is consistent with our demonstration that the overexpression of S3WT does not increase phospho‐S3 levels (Figure 3H and I). However, the enhanced 3TP‐lux activity by co‐transfection of S4 with S3WT, over that by S3WT alone, was reduced by Akt (Supplementary Figure 3S). Together, the above experiments suggest that suppression of TGF‐β signaling by Akt1 occurs not only through suppression of S3 activation via an Akt kinase‐dependent pathway, but also by transcriptional suppression of TGF‐β‐activated S3 by means of an Akt kinase‐independent mechanism.
image
Figure 3. Transcriptional activity of CA‐S3 but not S3WT is blocked by an Akt‐kinase‐independent mechanism in NRP‐152 cells. (A–C) CA‐PI3K, LR3‐IGF‐I, and Akt1Myr block 3TP‐Lux transcriptional activity induced by TGF‐β and CA‐S3 in NRP‐152 cells. (D, E) Inhibition of CA‐S3‐induced 3TP‐Lux (D) and SBE4BV‐Luc (E) by Akt was not reversed by LY (10 μM) or Rap (200 nM). (F) Akt1Myr, Akt1Myr/K179M, and Akt1K179M inhibit 3TP‐Lux induced by CA‐S3 activity. (G) Akt1Myr and Akt1Myr/K179M do not block 3TP‐Lux induced by S3WT activity, similar to the absence of a suppressive effect of LR3‐IGF‐I on S3WT activity. (H, I) Demonstration that activation of 3TP‐lux by S3WT does not involve the phospho‐activation of S3 (H), and that overexpression of S3WT does not enhance its phospho‐activation, by Western blot analysis (I). Cells were pretreated with the TβRI kinase inhibitor SB‐431542 (20 μM) to specifically block the phospho‐activation of receptor Smads. Firefly luciferase values were normalized to Renilla luciferase. Each bar represents the average of triplicate determinations±s.e. (A–H). Results are representative of two to three experiments.
The kinase independence of Akt1 on the suppression of CA‐S3 suggests that Akt1 may associate with S3, although in a kinase‐independent manner. To examine this possibility, NRP‐152 cells were infected with an adenovirus for Flag‐S3WT and various forms of Akt1, either alone or with DN‐PI3K, followed by treatment with TGF‐β, IGF‐I, LY, or Rap. Co‐immunoprecipitation (Co‐IP) assays showed that S3WT physically associates with WT, Myr, and KD forms of Akt1 (Figure 4A). Moreover, these physical interactions are not suppressed by TGF‐β1, LY, or Rap, and not enhanced by IGF‐I, supporting that the association of Akt1 with S3 is not sufficient for IGF‐I or Akt1 to suppress phospho‐activation of S3 (Figure 4B), in contrast to results of another group (Conery et al, 2004). Interestingly, DN‐PI3K slightly reduced the amount of the complex formed between Akt1 and S3, whereas LY enhanced such complex formation (Figure 4B). On close inspection of inputs, however, changes in complex formation actually reflected changes in the expression of S3 and/or Akt1 by the above treatments (Figure 4B). Similarly, although lesser Akt1Myr and Akt1K179M were immunoprecipitated than Akt1WT, these differences also reflected their relative expression levels (input, Figure 4A).
image
Figure 4. Akt1 binds to multiple Smad proteins in an Akt‐kinase‐independent mechanism. (A, B) NRP‐152 cells were coinfected with Admax‐Myc‐Akts (WT, Myr, KD), Admax‐Flag‐S3, and Admax‐DN‐PI3K, followed by washing with PBS. Cells were then incubated for 24 h with/without LY (10 μM) or Rap (200 nM) in GM3 medium followed by treatment with LR3‐IGF‐I (2 nM, 5 h), TGF‐β1(10 ng/ml, 4 h), or both LR3‐IGF‐I and TGF‐β1 before cell lysis. (C, D) Akt1s associate with not only S3 (at the MH2 domain) but also with S2, S4, S7, CA‐S2 (S2*), and CA‐S3 (S3*) in HEK293 cells. (E) Akt1 associates with S2WT and S3WT in HEK293T cells cultured in serum‐free DMEM/F12 medium (SFM) or in DMEM/F12 containing 5% FBS. (F) Endogenous Akt1 associates with endogenous Smads 2 and 3 in NRP‐152 cells. Whole‐cell lysate (500 μg (A–E) or 1 mg (F)) was subjected to immunoprecipitation with mouse anti‐Flag M2 IgG (A–E) or with either anti‐Smad2 or anti‐Smad3 IgGs (F), and expression of co‐immunoprecipitated Akts was detected with rabbit anti‐Myc IgG (A–E) or with anti‐Akt1 IgG (F). Results are representative of two to three different experiments.
We further studied the physical interaction of S3 with Akt1, by defining the domains of S3 required for such binding. For this, we used truncations of S3 containing MH1, MH1+middle linker, MH2, and MH2+middle linker. Co‐IP experiments revealed that S3 MH2 domain bound to Akt1WT with the highest affinity, whereas the S3 MH1 domain associated also with Akt1WT, albeit with much lower affinity. The middle linker was inhibitory to the interaction of Akt1 with MH1 or MH2 domains (Figure 4C). Thus, the suppression by the middle linker may negatively regulate S3's interaction with Akt.
To examine the association of other Smad proteins with Akt1, HEK293 cells were transiently co‐transfected with Flag‐tagged Smads (S2, S3, S4, S7, CA‐S3, or CA‐S2), and either Myc‐Akt1WT or Myc‐Akt1Myr. Co‐IP assays showed that all these Smads physically interact with Akt1, with the CA Smads (S3* and S2*) having slightly less affinity than WT Smads to Akt1 (Figure 4D). These data suggest that Akt may regulate TGF‐β signaling through interaction with multiple Smads, in contrast to the reports that Akt1 binds only the S3 isoform in HEK293T cells (Conery et al, 2004; Remy et al, 2004). We therefore compared the ability of transfected S2 and S3 to co‐IP Akt1 in HEK293T cells grown in either serum‐containing or serum‐free conditions (Figure 4E). Under both conditions, S2 and S3 each comparably pulled down Akt1. Moreover, we also showed that endogenous S2 and S3 could co‐IP endogenous Akt in NRP‐152 cells (Figure 4F).
In our previous report, we showed that Rap reverses the IGF‐I inhibition of TGF‐β responses including the phospho‐activation of S3 (Song et al, 2003b). Here, we show that Rap can also reverse the ability of either CA‐PI3K or Akt1Myr to inhibit TGF‐β‐induced phospho‐activation of S3 (Figure 5A and B), suggesting a role for mTOR in this mechanism. However, Rap has also been implicated in activation of TGF‐β signaling through reversing the inhibitory action of FKBP12 on activation of TβRI by TβRII (Stockwell and Schreiber, 1998). To test whether the above results with Akt1 and Rap could instead be explained by suppression of TβRI by FKBP12, we tested the effects of Akt1 and Rap on activation of phospho‐S3 and 3TP‐lux promoter activity induced by TβRIL193A/P194A/T204D (LPT), which is a constitutively active and FKBP12‐dead ALK5 (Charng et al, 1996). Adenoviral transduction of LPT led to phospho‐activated S3 that was inhibited by coinfection with Akt1Myr, and this Akt1Myr suppression was reversed in cells pretreated with Rap (Figure 5C). Similarly, activation of 3TP‐lux by LPT was significantly suppressed by both Akt1WT and Akt1Myr (Figure 5D) and such suppression was Rap reversible (Figure 5E), suggesting that such suppression by Akt1 is independent of the interaction of FKBP12 with TβRI. Interestingly, shorter treatment times of Rap or LY enhanced the induction of 3TP‐lux by LTP, indistinguishable from that by TβRIT204D (Supplementary Figure 4S). Together, these results support that Rap reverses the Akt suppression of receptor‐mediated S3 activation by blocking mTOR. We further tested this model by silencing mTOR with siRNA (si‐mTOR). Consistent with the dependence of Akt kinase activity, silencing of mTOR by siRNA reversed the Akt1 suppression of LPT‐induced 3TP‐lux activity (Figure 5F), but not that induced by CA‐S3 (Figure 5G). We next examined whether the activation of S3 by LPT could be blocked by Akt1Myr in an mTOR‐dependent manner. The activation of S3 with LPT was clearly suppressed by Akt1Myr, and silencing of mTOR by siRNA completely reversed the Akt block on such S3 activation (Figure 5H). mTOR expression was downregulated about three‐fold by si‐mTOR, and TGF‐β‐induced S3 activation was slightly enhanced by silencing mTOR (Figure 5H). Lastly, we showed that Akt1Myr can block the induction of apoptosis by LPT, and that the suppression of LPT‐induced apoptosis was reversed by Rap (Figure 5I). These results firmly support a role of mTOR as a key mediator of Akt kinase‐dependent effects on a number of TGF‐β responses, including S3 activation, transcription, and apoptosis.
image
Figure 5. mTOR is the critical mediator of inhibition of TGF‐β signaling by Akt in NRP‐152 cells. (A–C) Effect of Rap on suppression of either TGF‐β‐induced or LPT‐induced phospho‐Smad3 by either CA‐PI3K or Akt1Myr. (D, E) 3TP‐lux activity induced by LPT in NRP‐152 cells was inhibited by Akt and this inhibition was Rap (200 nM)‐reversible. (F, G) Silencing mTOR expression by siRNA reverses the Akt suppression of LPT‐induced 3TP‐lux. Cells were transiently transfected with siRNA against mTOR (si‐mTOR, 80 nM), scrambled control siRNA (80 nM), pUSE‐Myc‐Akt1Myr, 3TP‐Lux reporter construct, and either pCMV5‐LPT or pCMV5‐CA‐S3. Data shown are relative values of Firefly luciferase normalized to Renilla luciferase. Each bar represents the average of triplicate determinations±s.e. (D–G). (H) si‐mTOR (80 nM) reversed the inhibition by Akt of phospho‐activation of S3 induced by LPT, and phosphorylation of S3 by TGF‐β was enhanced by blocking mTOR expression with si‐mTOR. Results are representative of two to three experiments. (I) Rap reverses the ability of Akt1Myr to block apoptosis induced by LPT (upper panel, DNA ladder; lower panel, DNA ladder was quantified by a phosphoimager). Adenoviral transduction was used to deliver CA‐PI3K, AktMyr, and LPT (A–C, H, I). (J) A schematic representation of our model showing that TGF‐β signaling is blocked by both Akt kinase‐dependent and Akt kinase‐independent mechanisms, with mTOR as the downstream substrate of Akt that mediates suppression of S3 phospho‐activation. The kinase‐independent function of Akt inhibits gene transcription by activated S3, but does not block the activation of S3 by TGF‐β.
We propose a novel and unifying model by which Akt/mTOR suppresses TGF‐β signaling (Figure 5J), whereby Akt1 inhibits S3 activity through both Akt kinase‐dependent and Akt kinase‐independent mechanisms. Our data support that mTOR is the downstream target of Akt kinase, which mediates the suppression of S3 phospho‐activation. These findings further suggest that enhanced activation of mTOR may be pivotal to loss of TGF‐β responses during tumor progression. The mechanism by which mTOR is able to suppress S3 activation may occur either by an interaction with mTOR or via downstream signals. In the kinase‐independent mechanism, Akt blocks downstream of S3 activation but not phospho‐activation of S3. Thus, our data do not support the mechanism proposed by a recent study (Conery et al, 2004) that Akt blocks S3 activation by sequestering it from TGF‐β receptors. Rather, this molecular association may indirectly inhibit signals downstream of receptor‐activated S3.

Discussion

We published the first report that IGF‐I, acting through the IGF‐I receptor/PI3K/Akt signaling pathway, selectively blocks the ability of TGF‐β to activate S3 but not S2, induce apoptosis, and mediate gene expression (Song et al, 2003b). Two subsequent publications reported similar suppression of S3 activation in other cell lines including the Hep3B hepatocarinoma cell line (Conery et al, 2004; Remy et al, 2004), using insulin, but only at super‐physiological concentrations, levels that are well established to cross‐activate IGF receptors (Megyesi et al, 1975; Rosenfeld and Hintz, 1980). To specifically focus on the IGF‐IR signaling, our approach was to use physiologically relevant doses of IGF‐I and an analog of this growth factor (LR3‐IGF‐I) that is essentially unable to bind to IGF‐I binding proteins (Hsing et al, 1996). We found that physiological concentrations of insulin or IGF‐I were not able to suppress such TGF‐β‐induced responses in Hep3B cells (data not shown), in contrast to NRP‐152, DP‐153, and Mv1Lu cells. Insulin receptor signaling was reported to block the phospho‐activation of S3 by inducing the membrane localization of Akt, which directly binds to and sequesters S3 away from TβRI (Conery et al, 2004; Remy et al, 2004). In their model, the Akt kinase is not involved in suppression of S3 activation, as they reported that AktK179A was just as effective as AktMyr in suppressing S3 activation. However, we show that AktK179M and AktMyr have opposite effects on both biological responses of TGF‐β, and these Akts or IGF‐I are ineffective in suppressing transcriptional responses of overexpressed S3WT (without TGF‐β), evidence against the above sequestration model. Moreover, our data (not shown) with Hep3B cells, which are weakly responsive to TGF‐β, suggest that the underlying discrepancies result from differences in experimental design rather than cell type. For example, in our study, we measured changes in the level of phospho‐S3 by direct Western blot analysis. In contrast, the other studies detected phospho‐S3 only indirectly in the material precipitated with anti‐S3 antibodies, without assessing input levels of phospho‐S3. Moreover, cell lines that stably express KD‐Akt were derived without the use of an inducible expression system (Conery et al, 2004). As DN‐Akt blocks the downstream survival pathways of Akt (Jetzt et al, 2003), stable overexpression of DN‐Akt in this way would be selected against. Neither group showed data to confirm suppression of Akt kinase activity in KD‐Akt‐expressing cells, leaving open the possibility of compensation by activation of endogenous Akt kinase or a downstream target. To avoid such secondary phenotypic changes, our approach was to use adenoviral gene delivery for rapid and efficient gene expression. We also clearly show that cells expressing KD‐Akt are actually kinase dead, by the absence of GSK‐3β phosphorylation (Ser21/9).
Consistent with the role of S3 as a major mediator of TGF‐β responses, our results suggest that Akt1 suppresses multiple TGF‐β signals, including apoptosis, PAI‐1 induction, growth suppression, and downregulation of cyclin D2. Recent studies with neuroepithelial and glioblastoma cells suggest that Akt kinase may more directly reverse the growth suppressive effects of TGF‐β by blocking the induction of p21Cip1 promoter activated by the association of S3 and S4 with FoxO Forkhead transcription factors (Seoane et al, 2004). In contrast, Conery et al (2004) reported that Akt's suppression of TGF‐β responses is more limited, as they exclude TGF‐β‐mediated growth suppression, c‐myc suppression, or induction of p21Cip1 promoter activity.
The role of the physical interaction of S3 with Akt1 in the ability of Akt1 to block phospho‐activation of S3 is not clear. Our results show that the ability of Akt1 to block S3 activation does not correlate with the strength of its association to S3. For example, AktWT, AktK179M, and AktMyr similarly associate with S3; however, only AktWT and AktMyr block phospho‐activation of S3. The PI3K inhibitor, LY, enhances the formation of a complex between S3 and Akt1WT by elevating the levels of Akt1, whereas DN‐PI3K reduces the level of Akt1 precipitating with S3 (Figure 4B), and PTEN neither affects the levels of Akt or S3 nor alters complex formation between S3 and Akt1 (data not shown). Unexpectedly, our results show that Akt1K179M and DN‐PI3K each enhances the level of phospho‐S2 induced by TGF‐β, although kinase‐active Akt1 does not suppress S2 activation, in contrast to S3 (Figure 2A–D). These results suggest that S2 activation may be fully suppressed by basal levels of Akt1 activity, unlike S3, which can be suppressed further or completely by induced levels of Akt1 kinase. It is thus likely that different mechanisms are involved in the Akt suppression of S3 versus S2.
We show that Akt1 can also inhibit TGF‐β signals downstream of receptor‐activated S3, as shown by the induction of 3TP‐lux activity following transfection with CA‐S3, a disparity with Conery et al (2004), in which Akt1 was reported to not suppress 3TP‐luciferase activity induced by CA‐S3. We show that Akt1 can suppress signals downstream of S3 activation through a mechanism that is independent of the kinase activity of Akt1, in contrast to its suppression of S3 activation that requires Akt1 kinase activity. Although the mechanism behind this kinase‐independent suppression is not known, our results suggest that this may occur through a physical association of Akt1 with phospho‐S3. We also show novel associations of Akt1WT or Akt1Myr with Smads 2, 3, 4, and 7 (Figure 4D), in contrast to the other groups (Conery et al, 2004; Remy et al, 2004), who claim that S3 is the only Smad that binds Akt1. These discrepancies cannot be due to the variant of HEK293 cells used (we used HEK293 cells versus HEK293T), as we showed that S2 interacted with Akt1 in HEK293T cells (used by the other groups) by co‐IP experiments, and endogenous S2 and S3 can also interact with endogenous Akt1 as shown by co‐IP experiments with NRP‐152 cells.
Our data strongly support a new model of S3 suppression by Akt, in which mTOR is essential for the ability of Akt to suppress biological responses of TGF‐β and the phospho‐activation of S3. Our studies using LPT indicate that such suppression is independent of the association of FKBP12 to ALK5. There has been increased interest in understanding the role of mTOR in cancer, particularly in mediating Akt effects on tumor growth and angiogenesis (Humar et al, 2002; Chan, 2004). Rap, which has gained broad interest as a useful drug for therapeutic intervention of a number of late‐stage cancers, may function by preventing the Akt survival signals activated during cancer (Rao et al, 2004). Thus, a better understanding of the function of mTOR in TGF‐β signaling is likely to impact on the therapeutics of a variety of cancers.

Materials and methods

Materials

The following were used: recombinant human TGF‐β1 (R&D Systems); phospho‐Akt1 (Ser473, #9271), anti‐Akt1 (#9272), phospho‐GSK‐3α/β (#9331, Ser21/9), phospho‐S3 (Ser433/435, #9514), phospho‐S2 (#3101), and mTOR (#2972) antibodies (Cell Signaling); anti‐Smad3 (sc‐8332), anti‐cyclin D2 (sc‐593), and anti‐Myc (sc‐789) antibodies (Santa Cruz); anti‐Flag M2 (F‐3165), anti‐Flag M1 (F‐3040), and anti‐β‐actin (A‐5441) antibodies (Sigma); DMEM/F12 (1:1, v/v) and calf serum (Invitrogen); LR3‐IGF‐I, insulin (BioSource International); dexamethasone (Sigma); LY and Rap (BioMol); pCMV5‐Flag‐CA‐S3 (Chipuk et al, 2002; Song et al, 2003b), pSG5‐DN‐PI3K (p85αΔSH2N) and pSG5‐CA‐PI3K (p110αCAAX) (Downward, 1998), pUSE‐Myc‐Akt1WT, pUSE‐Myc‐Akt1Myr, and pUSE‐Myc‐Akt1K179M (Upstate), LPT (TβRIL193A/P194A/T204D) (Charng et al, 1996); pcDNA3‐Flag‐S3 truncations (Wang et al, 2005); characterized FBS (HyClone); SB‐431542 (Tocris).

Cell culture

NRP‐152 and NRP‐154 prostatic epithelial cell lines (Danielpour et al, 1994; Danielpour, 1996) were maintained in GM2.1 culture medium (DMEM/F12 supplemented with 5% FBS, 5 μg/ml insulin, and 0.1 μM dexamethasone) in 100‐mm Falcon culture dishes. The cells were kept at 37°C in a 95% air/5% CO2 environment and passaged every 3–4 days (at subconfluence), plating at a seeding density of 1:40. All experiments were performed in GM3 medium (DMEM/F12 supplemented with 1% calf serum, 15 mM HEPES (pH 7.5), and 0.1 μM dexamethasone). HEK293, HEK293T, Hep3B, and Mv1Lu cells were cultured in DMEM/F12 medium supplemented with 5% FBS.

Western blot analysis

Western blot analyses were performed essentially as described (Song et al, 2003a, 2003b). In brief, NRP‐152 cells were plated overnight at a density of 4 × 105 cells/2 ml of GM3 media/well in six‐well plates and infected for 2 h by adenovirus expressing Akt, DN‐PI3K, or CA‐PI3K, and washed once with PBS followed by addition of 2 ml GM3 medium. Following treatment, cells were washed with PBS and lysed at 4°C with ice‐cold RIPA buffer (PBS, 1% Nonidet P‐40, 0.1% sodium dodecylsulfate, 0.5% sodium deoxycholate) containing 1 mM sodium orthovanadate, Complete Mini‐EDTA‐free Protease Inhibitor Mixture™, and 1 mM phenylmethylsulfonyl fluoride. Lysates were clarified at 16 000 r.c.f. for 20 min (at 4°C) and supernatants were quantified by the BCA protein assay (Pierce). Lysates were boiled for 5 min in SDS–PAGE loading buffer containing 5% 2‐mercaptoethanol and electrophoresed through 4–12% NuPAGE BisTris gel (Invitrogen) and electrophoretically transferred to nitrocellulose membranes. Membranes were blocked for 1 h in TBS (10 mM Tris–HCl, pH 8.0, and 150 mM NaCl) containing 5% nonfat dry milk and 0.05% thimerosal, and incubated with the indicated primary antibodies and the appropriate horseradish peroxidase‐conjugated secondary antibody (1:5000; Jackson Laboratory). The SuperSignal Chemiluminescence Substrate System (Pierce) was used to visualize protein bands.

Immunoprecipitation

All these procedures were performed as described (Chipuk et al, 2002; Wang et al, 2005). NRP‐152 cells were infected by adenovirus for 2 h to introduce the indicated genes and cultured in GM3 medium. Separately, HEK293 and HEK293T cells were plated overnight in 5% FBS‐DMEM/F12 in six‐well plates at a density of 5 × 105 cells/2 ml/well and transfected with 2 μg of DNA per well by a calcium phosphate precipitation method (Chipuk et al, 2002). The tagged proteins were co‐immunoprecipitated with anti‐Flag M2 IgG1 monoclonal antibody (Sigma) and subjected to Western blot analysis.

Transient transfection and luciferase assay

All the procedures were performed essentially as described (Chipuk et al, 2002; Song et al, 2003b). In brief, NRP‐152, NRP‐154, or Mv1Lu cells were plated overnight at a density of 1 × 105 cells/1 ml/well in 12‐well plates. A total of 1 μg of DNA including reporter construct was transfected using a standard calcium phosphate co‐precipitation method, and 3 h later cells were glycerol‐shocked (15% glycerol in 1 × HEPES‐buffered saline) for 90 s. Cells were washed twice and incubated overnight in GM3 medium either in the presence or absence of signaling inhibitors and then cultured with or without TGF‐β1 for 24 h. Luciferase activity was measured using the Promega's Dual Luciferase Assay Kit and an ML3000 Microtiter Plate Luminometer.

siRNA

Rat mTOR siRNA oligonucleotide (si‐mTOR), 5′‐CCCAGCCUUUGUCAUGCCUTT‐3′, and its complementary RNA strand harboring TT at the 3′ end were synthesized by Dharmacon, annealed in vitro, and co‐transfected into NRP‐152 cells (80 nM per well, six‐well plates) with indicated plasmids by either Lipofectamine 2000 reagent (Invitrogen) or calcium phosphate precipitation method. siRNA pair of identical length and design of a scrambled sequence not found in the rat genome, 5′‐AGCUGUGCCCUUCAUCCCUTT‐3′, was used as a negative control. Cell extracts were prepared 24 or 48 h after siRNA transfection and subjected to Western blot analysis or dual luciferase assay.

Adenovirus

Adenovirus vectors that direct the expression of Myc‐Akt1WT, Myc‐Akt1Myr, Myc‐Akt1K179M, DN‐PI3K, CA‐PI3K, and Flag‐Smad3WT (AdMax‐Myc‐Akt1WT, Admax‐Myc‐Akt1Myr, Admax‐Myc‐Akt1K179M, Admax‐DN‐PI3K, Admax‐Myc‐CA‐PI3K, and Admax‐Flag‐Smad3WT) were constructed using the AdMax system (Microbix Biosystems). The regions corresponding to the coding sequence of each gene were subcloned from pUSE‐Myc‐Akt1WT, pUSE‐Myc‐Akt1Myr, pUSE‐Myc‐Akt1K179M, pSG5‐DN‐PI3K, pSG5‐Myc‐CA‐PI3K, and pCMV5‐Flag‐Smad3WT into the adenovirus shuttle vector pDC515. Constructs were confirmed by DNA sequencing (Cleveland Genomics) and protein expression (in HEK293 cells) by Western blot analysis with two different primary antibodies (epitope‐tagged antibody and one against expressed protein). pDC515‐Myc‐Akt1WT, pDC515‐Myc‐Akt1Myr, pDC515‐Myc‐Akt1K179M, pDC515‐DN‐PI3K, pDC515‐Myc‐CA‐PI3K, or pDC515‐Flag‐Smad3WT (1 μg) was co‐transfected with 1 μg of the genomic vector pBHGfrtDE1,3FLP into HEK293 cells in six‐well plates using the calcium phosphate precipitation method. Transfected cells were maintained in DMEM/F12 containing 2% FBS. Approximately 10 days after transfection, when viral cytopathic effect was observed, the cells were lysed by freeze–thaw and then expanded according to the manufacturer's protocol. The ability of viral preparations to deliver the desired functional protein was evaluated by overnight infection of NRP‐152 and NRP‐154 cells followed by Western blot analysis of cell lysates and, in certain cases, by direct immunostaining of the infected cells. To assay viral titers, aliquots of virus stocks were diluted 100‐fold in lysis solution (0.1% sodium dodecylsulfate, 10 mM Tris–HCl (pH 7.4), 1 mM EDTA) and incubated for 10 min at 56°C. The optical density of the samples at 260 nm was used to calculate the virus content using the relationship of 1012 virus particles/ml/OD260 unit.

DNA fragmentation assay

All the procedures were performed essentially as described (Hsing et al, 1996; Danielpour, 1999). Internucleosomal DNA fragmentation was detected using the TACS apoptosis DNA ladder kit (Trevigen). NRP‐152 cells were infected for 2 h with control virus, Admax‐Myc‐Akt1Myr, Admax‐Myc‐Akt1Myr/K179M, Admax‐Myc‐Akt1K179M, or Admax‐Myc‐CA‐PI3K in the absence or presence of LPT. Cells were then washed once with 1 × PBS and cultured for 24 or 48 h in GM3 medium, with the inclusion of 10 ng/ml TGF‐β1 or vehicle control for another 24 h. Cells were then lysed and DNA was purified according to the manufacturer's instructions. The nicked ends of 0.5 μg of DNA were labeled using 2.5 U of Klenow fragment of DNA polymerase I with 0.5 μCi [α‐32P]dCTP (NEN) for 30 min at room temperature. The labeled DNA was electrophoresed through a 1.5% Trevigel 500 gel in 1 × TAE at 85 V for 2 h. The gel was dried and directly exposed to an X‐Omat AR film (Kodak).

Cell cycle distribution with propidium iodide

In brief, NRP‐152 cells infected with Admax‐Myc‐Akt1Myr were treated with TGF‐β1 or vehicle for 48 h and then fixed with 90% methanol (final concentration) at −20°C. Fixed cells were washed free of methanol and incubated with RNase A for 20 min at 37°C. Cells were placed on ice for 10 min followed by staining with propidium iodide solution (final concentration 50 μg/ml) for 30 min at 4°C and analyzed by flow cytometry using Coulter XL (Coulter Electronics).

Supplementary data

Supplementary data are available at The EMBO Journal Online.

Acknowledgements

We thank Drs Joan Massagué, Bert Vogelstein, Michael Schneider, and Julian Downward for providing 3TP‐lux, SBE4BV‐luciferase, pCMV5‐LPT, and various PI3K constructs, respectively, and Dr Anthony Zeleznik for adenoviral protocols and suggestions. This study was supported by the NCI Grants R01CA092102 and R01CA102074.

Supporting Information

References

Abdollah S, Macias‐Silva M, Tsukazaki T, Hayashi H, Attisano L, Wrana JL (1997) TbetaRI phosphorylation of Smad2 on Ser465 and Ser467 is required for Smad2–Smad4 complex formation and signaling. J Biol Chem 272: 27678–27685
Bjornsti MA, Houghton PJ (2004) The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4: 335–348
Chan S (2004) Targeting the mammalian target of rapamycin (mTOR): a new approach to treating cancer. Br J Cancer 91: 1420–1424
Charng MJ, Kinnunen P, Hawker J, Brand T, Schneider MD (1996) FKBP‐12 recognition is dispensable for signal generation by type I transforming growth factor‐beta receptors. J Biol Chem 271: 22941–22944
Chipuk JE, Cornelius SC, Pultz NJ, Jorgensen JS, Bonham MJ, Kim SJ, Danielpour D (2002) The androgen receptor represses transforming growth factor‐beta signaling through interaction with Smad3. J Biol Chem 277: 1240–1248
Conery AR, Cao Y, Thompson EA, Townsend Jr CM, Ko TC, Luo K (2004) Akt interacts directly with Smad3 to regulate the sensitivity to TGF‐beta induced apoptosis. Nat Cell Biol 6: 366–372
Danielpour D, Kadomatsu K, Anzano MA, Smith JM, Sporn MB (1994) Development and characterization of nontumorigenic and tumorigenic epithelial cell lines from rat dorsal–lateral prostate. Cancer Res 54: 3413–3421
Danielpour D (1996) Induction of transforming growth factor‐beta autocrine activity by all‐trans‐retinoic acid and 1 alpha, 25‐dihydroxyvitamin D3 in NRP‐152 rat prostatic epithelial cells. J Cell Physiol 166: 231–239
Danielpour D (1999) Transdifferentiation of NRP‐152 rat prostatic basal epithelial cells toward a luminal phenotype: regulation by glucocorticoid, insulin‐like growth factor‐I and transforming growth factor‐beta. J Cell Sci 112: 169–179
Danielpour D (2005) Functions and regulation of transforming growth factor‐beta (TGF‐beta) in the prostate. Eur J Cancer 41: 846–857
Downward J (1998) Mechanisms and consequences of activation of protein kinase B/Akt. Curr Opin Cell Biol 10: 262–267
Fingar DC, Blenis J (2004) Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 23: 3151–3171
Gao X, Zhang Y, Arrazola P, Hino O, Kobayashi T, Yeung RS, Ru B, Pan D (2002) Tsc tumour suppressor proteins antagonize amino‐acid‐TOR signalling. Nat Cell Biol 4: 699–704
Grimberg A (2003) Mechanisms by which IGF‐I may promote cancer. Cancer Biol Ther 2: 630–635
Guo Y, Kyprianou N (1999) Restoration of transforming growth factor beta signaling pathway in human prostate cancer cells suppresses tumorigenicity via induction of caspase‐1‐mediated apoptosis. Cancer Res 59: 1366–1371
Hayward SW, Haughney PC, Lopes ES, Danielpour D, Cunha GR (1999) The rat prostatic epithelial cell line NRP‐152 can differentiate in vivo in response to its stromal environment. Prostate 39: 205–212
Hsing AY, Kadomatsu K, Bonham MJ, Danielpour D (1996) Regulation of apoptosis induced by transforming growth factor‐beta1 in nontumorigenic rat prostatic epithelial cell lines. Cancer Res 56: 5146–5149
Humar R, Kiefer FN, Berns H, Resink TJ, Battegay EJ (2002) Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)‐dependent signaling. FASEB J 16: 771–780
Jetzt A, Howe JA, Horn MT, Maxwell E, Yin Z, Johnson D, Kumar CC (2003) Adenoviral‐mediated expression of a kinase‐dead mutant of Akt induces apoptosis selectively in tumor cells and suppresses tumor growth in mice. Cancer Res 63: 6697–6706
Manning BD, Cantley LC (2003) United at last: the tuberous sclerosis complex gene products connect the phosphoinositide 3‐kinase/Akt pathway to mammalian target of rapamycin (mTOR) signalling. Biochem Soc Trans 31: 573–578
Massague J (1992) Receptors for the TGF‐beta family. Cell 69: 1067–1070
Megyesi K, Kahn CR, Roth J, Neville Jr DM, Nissley SP, Humbel RE, Froesch ER (1975) The NSILA‐s receptor in liver plasma membranes. Characterization and comparison with the insulin receptor. J Biol Chem 250: 8990–8996
Nicholson KM, Anderson NG (2002) The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal 14: 381–395
Plas DR, Thompson CB (2003) Akt activation promotes degradation of tuberin and FOXO3a via the proteasome. J Biol Chem 278: 12361–12366
Rao RD, Buckner JC, Sarkaria JN (2004) Mammalian target of rapamycin (mTOR) inhibitors as anti‐cancer agents. Curr Cancer Drug Targets 4: 621–635
Remy I, Montmarquette A, Michnick SW (2004) PKB/Akt modulates TGF‐beta signalling through a direct interaction with Smad3. Nat Cell Biol 6: 358–365
Renehan AG, Zwahlen M, Minder C, O'Dwyer ST, Shalet SM, Egger M (2004) Insulin‐like growth factor (IGF)‐I, IGF binding protein‐3, and cancer risk: systematic review and meta‐regression analysis. Lancet 363: 1346–1353
Roberts AB, Sporn MB (1990) The Transforming Growth Factor Beta. New York: Springer‐Verlag
Rosenfeld RG, Hintz RL (1980) Characterization of a specific receptor for somatomedin C (SM‐C) on cultured human lymphocytes: evidence that SM‐C modulates homologous receptor concentration. Endocrinology 107: 1841–1848
Sarbassov dos D, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation of Akt/PKB by the rictor–mTOR complex. Science 307: 1098–1101
Seoane J, Le HV, Shen L, Anderson SA, Massague J (2004) Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117: 211–223
Song K, Cornelius SC, Danielpour D (2003a) Development and characterization of DP‐153, a nontumorigenic prostatic cell line that undergoes malignant transformation by expression of dominant‐negative transforming growth factor beta receptor type II. Cancer Res 63: 4358–4367
Song K, Cornelius SC, Reiss M, Danielpour D (2003b) Insulin‐like growth factor‐I inhibits transcriptional responses of transforming growth factor‐beta by phosphatidylinositol 3‐kinase/Akt‐dependent suppression of the activation of Smad3 but not Smad2. J Biol Chem 278: 38342–38351
Stewart LV, Song K, Hsing AY, Danielpour D (2003) Regulation of trespin expression by modulators of cell growth, differentiation, and apoptosis in prostatic epithelial cells. Exp Cell Res 284: 301–313
Stockwell BR, Schreiber SL (1998) TGF‐beta‐signaling with small molecule FKBP12 antagonists that bind myristoylated FKBP12‐TGF‐beta type I receptor fusion proteins. Chem Biol 5: 385–395
Tang B, de Castro K, Barnes HE, Parks WT, Stewart L, Bottinger EP, Danielpour D, Wakefield LM (1999) Loss of responsiveness to transforming growth factor beta induces malignant transformation of nontumorigenic rat prostate epithelial cells. Cancer Res 59: 4834–4842
Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J (2003) Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase‐activating protein complex toward Rheb. Curr Biol 13: 1259–1268
ten Dijke P, Miyazono K, Heldin CH (1996) Signaling via hetero‐oligomeric complexes of type I and type II serine/threonine kinase receptors. Curr Opin Cell Biol 8: 139–145
Tsukazaki T, Chiang TA, Davison AF, Attisano L, Wrana JL (1998) SARA, a FYVE domain protein that recruits Smad2 to the TGFbeta receptor. Cell 95: 779–791
Vivanco I, Sawyers CL (2002) The phosphatidylinositol 3‐kinase AKT pathway in human cancer. Nat Rev Cancer 2: 489–501
Wakefield LM, Roberts AB (2002) TGF‐beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev 12: 22–29
Wang H, Song K, Sponseller TL, Danielpour D (2005) Novel function of androgen receptor‐associated protein 55/Hic‐5 as a negative regulator of Smad3 signaling. J Biol Chem 280: 5154–5162
Wieser R, Wrana JL, Massague J (1995) GS domain mutations that constitutively activate T beta R‐I, the downstream signaling component in the TGF‐beta receptor complex. EMBO J 14: 2199–2208
Wu RY, Zhang Y, Feng XH, Derynck R (1997) Heteromeric and homomeric interactions correlate with signaling activity and functional cooperativity of Smad3 and Smad4/DPC4. Mol Cell Biol 17: 2521–2528
Xiao Z, Liu X, Lodish HF (2000) Importin beta mediates nuclear translocation of Smad 3. J Biol Chem 275: 23425–23428
Xu G, Zhang W, Bertram P, Zheng XF, McLeod H (2004) Pharmacogenomic profiling of the PI3K/PTEN–AKT–mTOR pathway in common human tumors. Int J Oncol 24: 893–900
Yamaguchi H, Wang HG (2001) The protein kinase PKB/Akt regulates cell survival and apoptosis by inhibiting Bax conformational change. Oncogene 20: 7779–7786

Information & Authors

Information

Published In

The EMBO Journal cover image
Read More
The EMBO Journal
Vol. 25 | No. 1
11 January 2006
Table of contents
Pages: 58 - 69

Article versions

Submission history

Received: 25 May 2005
Accepted: 24 November 2005
Published online: 15 December 2005
Published in issue: 11 January 2006

Permissions

Request permissions for this article.

Keywords

  1. Akt
  2. IGF‐I
  3. mTOR
  4. PKB
  5. prostate
  6. rapamycin
  7. Smad

Authors

Affiliations

David Danielpour* [email protected]
Case Comprehensive Cancer Center Research Laboratories and the Department of Pharmacology Case Western Reserve University Cleveland OH USA

Notes

*
Corresponding author. Ireland Cancer Center Research Labs, Case Western Reserve University, Wolstein Research Building, Room 3‐532, 2103 Cornell Avenue, Cleveland, OH 44106, USA. Tel.: +1 216 368 5670; Fax: +1 216 368 8919; E‐mail: [email protected]

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

View Options

View options

PDF

View PDF

Get Access

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media