Artigo Acesso aberto Revisado por pares

The Tuberin-Hamartin Complex Negatively Regulates β-Catenin Signaling Activity

2003; Elsevier BV; Volume: 278; Issue: 8 Linguagem: Inglês

10.1074/jbc.c200473200

ISSN

1083-351X

Autores

Baldwin Mak, Ken‐Ichi Takemaru, Heidi L. Kenerson, Randall T. Moon, Raymond S. Yeung,

Tópico(s)

Kruppel-like factors research

Resumo

Tuberous sclerosis complex (TSC) is characterized by the formation of hamartomas in multiple organs resulting from mutations in the TSC1 or TSC2 gene. Their protein products, hamartin and tuberin, respectively, form a functional complex that affects cell growth, differentiation, and proliferation. Several lines of evidence, including renal tumors derived from TSC2+/− animals, suggest that the loss or inhibition of tuberin is associated with up-regulation of cyclin D1. As cyclin D1 can be regulated through the canonical Wnt/β-catenin signaling pathway, we hypothesize that the cell proliferative effects of hamartin and tuberin are partly mediated through β-catenin. In this study, total β-catenin protein levels were found to be elevated in the TSC2-related renal tumors. Ectopic expression of hamartin and wild-type tuberin, but not mutant tuberin, reduced β-catenin steady-state levels and its half-life. The TSC1-TSC2 complex also inhibited Wnt-1 stimulated Tcf/LEF luciferase reporter activity. This inhibition was eliminated by constitutively active β-catenin but not by Disheveled, suggesting that hamartin and tuberin function at the level of the β-catenin degradation complex. Indeed, hamartin and tuberin co-immunoprecipitated with glycogen synthase kinase 3 β and Axin, components of this complex in a Wnt-1-dependent manner. Our data suggest that hamartin and tuberin negatively regulate β-catenin stability and activity by participating in the β-catenin degradation complex. Tuberous sclerosis complex (TSC) is characterized by the formation of hamartomas in multiple organs resulting from mutations in the TSC1 or TSC2 gene. Their protein products, hamartin and tuberin, respectively, form a functional complex that affects cell growth, differentiation, and proliferation. Several lines of evidence, including renal tumors derived from TSC2+/− animals, suggest that the loss or inhibition of tuberin is associated with up-regulation of cyclin D1. As cyclin D1 can be regulated through the canonical Wnt/β-catenin signaling pathway, we hypothesize that the cell proliferative effects of hamartin and tuberin are partly mediated through β-catenin. In this study, total β-catenin protein levels were found to be elevated in the TSC2-related renal tumors. Ectopic expression of hamartin and wild-type tuberin, but not mutant tuberin, reduced β-catenin steady-state levels and its half-life. The TSC1-TSC2 complex also inhibited Wnt-1 stimulated Tcf/LEF luciferase reporter activity. This inhibition was eliminated by constitutively active β-catenin but not by Disheveled, suggesting that hamartin and tuberin function at the level of the β-catenin degradation complex. Indeed, hamartin and tuberin co-immunoprecipitated with glycogen synthase kinase 3 β and Axin, components of this complex in a Wnt-1-dependent manner. Our data suggest that hamartin and tuberin negatively regulate β-catenin stability and activity by participating in the β-catenin degradation complex. The phenotype of patients with TSC 1The abbreviations used are: TSC, tuberous sclerosis complex; PI3K, phosphoinositide 3-kinase; Tcf/LEF, T cell factor/lymphoid enhancer factor; APC, adenomatous polyposis coli; GSK3β, glycogen synthase kinase 3 β; Dsh, Disheveled; PMSF, phenylmethylsulfonyl fluoride; ConA, concanavalin A encompasses the development of multiple focal lesions in the brain, heart, kidney, lung, and skin (1Gomez M.R. Brain Dev. 1995; 17 (suppl.): 55-57Google Scholar). While the underlying pathogenic mechanisms are unclear, the histology of these discrete, tumor-like lesions suggests defects in cell proliferation, differentiation, and cell size control. Studies in mammalian cells have shown that overexpression of TSC1 and TSC2 negatively regulates cell proliferation and induces G1/S arrest (2Jin F. Wienecke R. Xiao G.H. Maize Jr., J.C. DeClue J.E. Yeung R.S. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 9154-9159Google Scholar, 3Soucek T. Pusch O. Wienecke R. DeClue J.E. Hengstschlager M. J. Biol. Chem. 1997; 272: 29301-29308Google Scholar, 4Miloloza A. Rosner M. Nellist M. Halley D. Bernaschek G. Hengstschlager M. Hum. Mol. Genet. 2000; 9: 1721-1727Google Scholar). In the case of tuberin, there appears to be an inverse correlation between tuberin level and p27(Kip1) expression and stability (5Soucek T. Yeung R.S. Hengstschlager M. Proc. Natl. Acad. Sci. U. S. A. 1998; 95: 15653-15658Google Scholar). Correspondingly, evidence supports a link between tuberin and cyclin D1 expression. Cortical tubers microdissected from TSC patients showed elevated cyclin D1 mRNA expression in the giant cells (6Crino P.B. Trojanowski J.Q. Dichter M.A. Eberwine J. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 14152-14157Google Scholar). Antisense inhibition of TSC2 in Rat1 fibroblasts resulted in up-regulation of cyclin D1 protein (3Soucek T. Pusch O. Wienecke R. DeClue J.E. Hengstschlager M. J. Biol. Chem. 1997; 272: 29301-29308Google Scholar). Renal cortical tumors from the Eker rat model for TSC express elevated cyclin D1 compared with unaffected kidney tissue (7Kenerson H.L. Aicher L.D. True L.D. Yeung R.S. Cancer Res. 2002; 62: 5645-5650Google Scholar). As an in vivotarget of the β-catenin pathway, cyclin D1 mRNA is responsive to the activity of the Tcf/LEF family of transcription factors (8Shtutman M. Zhurinsky J. Simcha I. Albanese C. D'Amico M. Pestell R. Ben-Ze'ev A. Proc. Natl. Acad. Sci. U. S. A. 1999; 96: 5522-5527Google Scholar, 9Tetsu O. McCormick F. Nature. 1999; 398: 422-426Google Scholar). This raises the possibility that TSC1 and TSC2 negatively regulate β-catenin signaling and, thereby, modulate the expression of cyclin D1. β-Catenin is a highly conserved 95-kDa protein that participates in cell-cell adhesion through its association with members of the membrane-bound cadherin family, and in cell proliferation and differentiation as a key component of the Wnt/Wingless pathway (reviewed in Ref. 10Daniels D.L. Ekolf Spink K. Weis W.I. Trends Biochem. Sci. 2001; 26: 672-678Google Scholar). In its signaling role, β-catenin shuttles between the cytoplasm and the nucleus where it binds the Tcf/LEF family of transcription factors to activate downstream target genes (reviewed in Ref. 11Miller J.R. Hocking A.M. Brown J.D. Moon R.T. Oncogene. 1999; 18: 7860-7872Google Scholar). In the absence of the secreted factor, Wnt, β-catenin is phosphorylated by GSK3β and is targeted for ubiquitination and degradation. Upon Wnt stimulation, Disheveled (Dsh) is activated and blocks the ability of GSK3β to phosphorylate β-catenin. Other components of this degradation complex include Axin, serving as a scaffolding protein, and APC, a tumor suppressor protein. Disruption at multiple levels of this pathway has been shown to be oncogenic in humans and rodents. In this study, β-catenin protein levels were found to be elevated in renal tumors from Eker rats. Overexpression of tuberin and hamartin in cells down-regulated β-catenin levels, its half-life and its activity. Furthermore, we showed that TSC1 and TSC2 proteins co-immunoprecipitated with GSK3β and Axin, supporting a role of hamartin and tuberin in modulating the β-catenin pathway. Eker rats harboring a germ-line TSC2 mutation were described previously (12Yeung R.S. Xiao G.H. Jin F. Lee W.C. Testa J.R. Knudson A.G. Proc. Natl. Acad. Sci. U. S. A. 1994; 91: 11413-11416Google Scholar). Human embryonic kidney cells (HEK293T) were obtained from the American Type Culture Collection (Manassas, VA). All cell culture reagents and transfection reagents (LipofectAMINE and Plus reagents) were purchased from Invitrogen. Monoclonal antibodies directed toward β-catenin and GSK3β were purchased from BD Transduction Laboratories (Los Angeles, CA). Monoclonal antibodies for c-Myc (9E10), actin, and α-tubulin were from Sigma. IgG purified polyclonal antibodies directed toward tuberin (L3–2) and hamartin (4050) were prepared as described earlier (13Xiao G.H. Shoarinejad F. Jin F. Golemis E.A. Yeung R.S. J. Biol. Chem. 1997; 272: 6097-6100Google Scholar, 14Aicher L.D. Campbell J.S. Yeung R.S. J. Biol. Chem. 2001; 276: 21017-21021Google Scholar). Polyclonal antibody for tuberin (C20) was obtained from Santa Cruz (Santa Cruz, CA). Expression vectors included rat TSC2 (14Aicher L.D. Campbell J.S. Yeung R.S. J. Biol. Chem. 2001; 276: 21017-21021Google Scholar), human TSC1 (15Plank T.L. Yeung R.S. Henske E.P. Cancer Res. 1998; 58: 4766-4770Google Scholar), mWnt-1 (Gift from Marian Waterman, University of California, Irvine, CA), constitutively active-β-catenin (CA-β-catenin) (16Yost C. Torres M. Miller J.R. Huang E. Kimelman D. Moon R.T. Genes Dev. 1996; 10: 1443-1454Google Scholar),Xenopus Dsh (17Sokol S.Y. Klingensmith J. Perrimon N. Itoh K. Development (Camb.). 1995; 121: 1637-1647Google Scholar), c-Myc-Xenopus Axin, ΔN-Tcf-4 (18Korinek V. Barker N. Morin P.J. van Wichen D. de Weger R. Kinzler K.W. Vogelstein B. Clevers H. Science. 1997; 275: 1784-1787Google Scholar), c-β-galactosidase (19Turner D.L. Weintraub H. Genes Dev. 1994; 8: 1434-1447Google Scholar), Xenopus GSK-3β (XG73) (20Pierce S.B. Kimelman D. Dev. Biol. 1996; 175: 256-264Google Scholar), and the reporter constructs TOPFLASH and FOPFLASH (18Korinek V. Barker N. Morin P.J. van Wichen D. de Weger R. Kinzler K.W. Vogelstein B. Clevers H. Science. 1997; 275: 1784-1787Google Scholar). Luciferin stock solution was purchased from Pharmingen. Galacton and Emerald stock solutions were obtained from Tropix (Bedford, MA). Immobilon-P (polyvinylidene difluoride) membranes used for Western blots were from Millipore (Bedford, MA). Radiolabeled [35S]methionine (EASYTAGTM express protein labeling mix) was purchased from PerkinElmer (Boston, MA). All other reagents were purchased from Sigma. Eker rats were sacrificed at 12 months of age to collect kidney samples. Kidney lesions were dissected from unaffected tissue. Both were homogenized in cold RIPA buffer (1% Nonidet P-40, 1% sodium deoxycholate, 0.1% SDS, 0.15m NaCl, 10 mm Tris-HCl, pH 7.2, 0.025m β-glycophosphate, pH 7.2, 2 mm EDTA, 50 mm sodium fluoride, 0.05 mm4-(2-aminoethyl)benzenesulfonyl fluoride (AEBSF), 10 μg/ml aprotinin, 10 μg/ml pepstatin, 1 mm orthovanadatate, 10 μg/ml leupeptin, 1 mm microcystin LR), and the protein concentration was determined using the BCA protein assay (Pierce). SDS-PAGE and Western blots were performed using equal amounts of total protein. HEK293T cells were transfected with increasing concentrations of TSC1 and TSC2 (wild-type or ΔY1571H mutant) vectors or with control vector (pcDNA3) (500, 1000, and 1500 ng) either with or without Wnt-1 vector using LipofectAMINE Plus reagents according to the manufacturer's instructions. Empty plasmids were added accordingly to normalize total DNA transfected. The cells were harvested in lysis buffer (10 mm Tris-HCl, pH 7.5, 100 mm NaCl, 0.5% Nonidet P-40, 0.5 μg/ml leupeptin, 1.0 μg/ml pepstatin, 0.2 mmPMSF) and lysed by freeze thawing at 48 h after transfection. Samples were analyzed by Western blotting. Vectors encoding TSC1, TSC2, or TSC2 (ΔY1571H) mutant were co-transfected into HEK293T cells with or without Wnt-1. After 48 h, pulse-chase using [35S]methionine incorporation was performed on transfected cells as outlined by Williams et al. (21Williams B.O. Barish G.D. Klymkowsky M.W. Varmus H.E. Oncogene. 2000; 19: 5720-5728Google Scholar). Protein content in cell lysates was analyzed using the BCA protein assay. Equal amounts of cell lysate were adjusted to equal volumes, pretreated with ConA-Sepharose (1:1 in immunoprecipitation buffer, see below) (Amersham Biosciences, Uppsala, Sweden) and then subjected to immunoprecipitation for β-catenin. Samples were resolved by SDS-PAGE and detected by autoradiography. Radiolabel band intensities were determined using a PhosphorImager Storm 840 (AmershamBiosciences). Cells (HEK293T) were transfected with the Tcf/LEF reporter construct (TOPFLASH) (18Korinek V. Barker N. Morin P.J. van Wichen D. de Weger R. Kinzler K.W. Vogelstein B. Clevers H. Science. 1997; 275: 1784-1787Google Scholar) using LipofectAMINE Plus reagents (Invitrogen). Vectors encoding TSC1, TSC2, or TSC2 (ΔY1571H) mutant were co-transfected with or without Wnt-1, Dsh, or a constitutively active β-catenin mutant (CA-β-cat) construct. A vector encoding β-galactosidase was co-transfected as a transfection control, while a dominant negative Tcf-4 (ΔN-Tcf-4) was used as a negative control for activation. Reporter vector containing mutated Tcf/LEF binding sites (FOPFLASH) (18Korinek V. Barker N. Morin P.J. van Wichen D. de Weger R. Kinzler K.W. Vogelstein B. Clevers H. Science. 1997; 275: 1784-1787Google Scholar) served as a control for background activity. Luciferase activity was analyzed using an EG&G Berthold Autolumet LB953 luminometer (PerkinElmer Instruments, Bad Wildbad, Germany), and normalized to corresponding β-galactosidase values. For immunoprecipitation of endogenous proteins, HEK293T cells were transfected Wnt-1 construct or empty vector. Similarly, for ectopically expressed proteins, HEK293T cells were transfected with TSC1 and TSC2 vectors or pcDNA3 (vector control) in the presence of vectors for GSK3β or c-Myc-tagged Axin. Cells were harvested the following day in lysis buffer (10 mm Tris-HCl, pH 7.5, 100 mm NaCl, 0.5% Nonidet P-40, 0.5 μg/ml leupeptin, 1.0 μg/ml pepstatin, 0.2 mmPMSF). Equal total amounts of lysates were immunoprecipitated using anti-tuberin polyclonal antibody, anti-GSK monoclonal antibody, or anti-c-Myc antibodies as described previously (14Aicher L.D. Campbell J.S. Yeung R.S. J. Biol. Chem. 2001; 276: 21017-21021Google Scholar). The Eker rat contains a germ-line mutation in the TSC2 gene (12Yeung R.S. Xiao G.H. Jin F. Lee W.C. Testa J.R. Knudson A.G. Proc. Natl. Acad. Sci. U. S. A. 1994; 91: 11413-11416Google Scholar) and spontaneously develops renal cortical epithelial tumors that have been shown to possess biallellic inactivation of TSC2 due to loss of heterozygosity, nonsense mutation, or null mutation (22Kajino K. Hino O. Contrib. Nephrol. 1999; 128: 45-50Google Scholar). In a previous study, cyclin D1 levels were shown to be elevated in these kidney tumors compared with unaffected kidney tissue (7Kenerson H.L. Aicher L.D. True L.D. Yeung R.S. Cancer Res. 2002; 62: 5645-5650Google Scholar). Since cyclin D1 gene is a known target of the β-catenin signaling pathway and the accumulation of β-catenin has been shown to activate the transcription of the cyclin D1 gene (8Shtutman M. Zhurinsky J. Simcha I. Albanese C. D'Amico M. Pestell R. Ben-Ze'ev A. Proc. Natl. Acad. Sci. U. S. A. 1999; 96: 5522-5527Google Scholar, 9Tetsu O. McCormick F. Nature. 1999; 398: 422-426Google Scholar). β-Catenin levels were examined in Eker rat kidney tumors. Tumors from three separate Eker rats were dissected from unaffected tissue, homogenized, and analyzed by Western blotting for β-catenin expression. As shown in Fig.1 A, β-catenin levels are higher in tumor samples compared with corresponding unaffected kidney tissue, reflecting the trend observed for cyclin D1. Thus β-catenin appears to accumulate upon TSC2 inactivation implying that tuberin may affect β-catenin levels. To investigate a possible link between tuberin and the β-catenin signaling pathway, we analyzed the effects of tuberin expression on β-catenin steady-state levels. Endogenous β-catenin expression was assessed in HEK293T cells 48 h after transfection with increasing concentrations of TSC1 and TSC2 or control vectors in the presence or absence of Wnt-1 stimulation. In the absence of Wnt-1 stimulation, β-catenin levels were unchanged with or without overexpression of hamartin and tuberin (data not shown). Upon Wnt-1 stimulation, β-catenin accumulated over a 48-h period in vector control samples (Fig. 1 B, compare lanes 1–4). However, β-catenin levels were significantly reduced with overexpression of wild-type tuberin and hamartin (Fig. 1 B, compare lanes 1–7). In contrast, overexpression of a disease-causing TSC2 mutant (ΔY1571H) (14Aicher L.D. Campbell J.S. Yeung R.S. J. Biol. Chem. 2001; 276: 21017-21021Google Scholar) resulted in a modest β-catenin accumulation (Fig. 1 B, compare lanes 8–10). These results show an inverse relationship between β-catenin and hamartin/tuberin expression. With increasing expression of hamartin and wild-type tuberin, β-catenin levels diminished progressively (Fig.1 B, compare lanes 5–7). In contrast, steady-state β-catenin increased modestly with the expression of control vector (Fig. 1 B, compare lanes 2–4) or with mutant tuberin (ΔY1571H) (Fig. 1 B, comparelanes 8–10). Next, we examined the effect of hamartin and tuberin expression on β-catenin half-life. HEK293T cells were transfected as described above and then subjected to pulse-chase following [35S]methionine incorporation. Lysates collected at specific time points were treated with ConA-Sepharose to remove cadherin-bound β-catenin leaving only the free pool of β-catenin for immunoprecipitation. Immunoprecipitates were resolved on SDS-PAGE, and the radiolabel intensity of [35S]methionine incorporated in to β-catenin was measured from the gel to determine the half-life. Without Wnt-1 stimulation, β-catenin half-life was unchanged either with or without expression of hamartin and tuberin (data not shown). With Wnt-1 stimulation, the half-life of β-catenin in vector control samples was 1.7 h (Fig. 1 C). This was the same for β-catenin in samples overexpressing hamartin and mutant tuberin (ΔY1571H) (Fig. 1 C). However, the half-life of β-catenin was reduced to about 1 h upon expression of hamartin and wild-type tuberin (Fig. 1 C). This 41% decrease in β-catenin half-life is consistent with the steady-state data showing an effect of the TSC proteins on β-catenin level. In both situations, modulation of β-catenin by wild-type tuberin occurred under condition of Wnt stimulation. This function is disrupted in the presence of a disease-causing TSC2 mutation. To determine whether modulation of β-catenin levels by hamartin and tuberin affects its transcriptional activity, the ability of Wnt-1 to activate a Tcf/LEF-luciferase reporter construct (TOPFLASH) was examined in transient transfection assays using HEK293T cells. These cells were co-transfected with hamartin and/or tuberin constructs along with a vector for β-galactosidase to account for transfection efficiency. Parallel assays were performed using FOPFLASH, a mutant reporter, to monitor background activity. Upon stimulation with Wnt-1, cells with vector control revealed a 12-fold increase in luciferase activity relative to the non-stimulated cells (Fig.2 A, compare lanes 1and 2). Co-expression of both wild-type tuberin and hamartin in Wnt-1 stimulated cells significantly reduced reporter activity (Fig.2 A, lane 3), while expression of hamartin or tuberin alone had only minor effects (Fig. 2 A, lanes 5 and 6). Importantly, co-expression of the tuberin ΔY1571H mutant with hamartin did not suppress Wnt-1-stimulated TOPFLASH activity (Fig. 2 A, lane 4) consistent with the effects on β-catenin protein levels described above (Fig. 1,B and C). Under the same conditions, hamartin and tuberin had no effects on FOPFLASH activity (Fig. 2 A,lanes 7 and 8). Our data suggest that hamartin and tuberin, functioning as a complex, are capable of inhibiting Wnt-1 stimulated β-catenin-dependent transcriptional activity. To determine at what level in the Wnt/β-catenin signaling pathway hamartin and tuberin act, we examined the effects of TSC1 and TSC2 on TOPFLASH activity when stimulated by different components of the Wnt pathway. The CA-β-catenin mutant with its serine/threonine residues (Ser-33, Ser-37, Thr-41, Ser-45) replaced with alanine residues, thus preventing its phosphorylation and degradation (16Yost C. Torres M. Miller J.R. Huang E. Kimelman D. Moon R.T. Genes Dev. 1996; 10: 1443-1454Google Scholar), acts as a downstream stimulus and activates the TOPFLASH reporter by over 6-fold in HEK293T cells (Fig. 2 B, compare lanes 1 and 2). Co-expression of hamartin and tuberin was ineffective in reducing CA-β-catenin stimulation of the Tcf/LEF reporter (Fig. 2 B, compare lanes 2and 3). Western blot of samples confirmed equal expression of CA-β-catenin (data not shown). As a control, a dominant negative mutant of Tcf-4 (ΔN-Tcf-4) that lacks the N-terminal β-catenin binding domain (18Korinek V. Barker N. Morin P.J. van Wichen D. de Weger R. Kinzler K.W. Vogelstein B. Clevers H. Science. 1997; 275: 1784-1787Google Scholar) was able to inhibit CA-β-catenin activity completely (Fig. 2 B, lane 6). These results suggest that hamartin and tuberin act upstream of β-catenin. Next, TOPFLASH activity was measured in the presence of ectopically expressed Dsh, an effector that is stimulated by the Wnt-Frizzled receptor complex upstream of β-catenin (see Ref. 10Daniels D.L. Ekolf Spink K. Weis W.I. Trends Biochem. Sci. 2001; 26: 672-678Google Scholar). Transient overexpression of Dsh stimulates TOPFLASH activity by ∼3-fold in control vector transfected cells (Fig. 2 C, comparelanes 1 and 2). This activity was inhibited to near baseline levels upon overexpression of hamartin and tuberin (Fig.2 C, lane 3). Hamartin alone did not reduce the activity, while tuberin alone slightly decreased activity (Fig.2 C, lanes 4 and 5). Again, ΔN-Tcf-4 reduced activity toward unstimulated levels (Fig. 2 C,lane 6). Together, these results are consistent with tuberin and hamartin exerting an effect on the Wnt signaling pathway at a level between Dsh and β-catenin (i.e. the β-catenin degradation complex). The β-catenin degradation complex is comprised of several proteins, including APC, Axin, and GSK3β, and is responsible for the regulation of cytoplasmic β-catenin (see Ref. 10Daniels D.L. Ekolf Spink K. Weis W.I. Trends Biochem. Sci. 2001; 26: 672-678Google Scholar). To determine whether hamartin and tuberin physically interact with components of the β-catenin degradation complex, co-immunoprecipitation assays were performed in HEK293T cells ectopically expressing hamartin and tuberin along with GSK3β. Anti-tuberin antibodies brought down GSK3β only in samples where both were overexpressed (Fig. 3 A,panel i, lane 4). This band was not observed in samples where the GSK3β construct was co-expressed with vector control (Fig. 3 A, panel i, lane 2) or in samples without the GSK3β construct (Fig. 3 A,panel i, lanes 1 and 3). As expected, hamartin co-immunoprecipitated with tuberin in sample where both were overexpressed (Fig. 3 A, panel ii, lanes 3 and 4). Conversely, immunoprecipitation of GSK3β brought down both tuberin and hamartin only in samples where all three were overexpressed (Fig. 3 A, panels iv andv, lane 4). The expression of ectopic proteins was verified in cell lysates (Fig. 3 A, panels vii, viii, and ix). Compared with the level of overexpression, the amount of interacting protein was relatively small, suggesting that only a fraction of tuberin/hamartin and GSK3β can associate with one another. If hamartin and tuberin interact with the GSK3β that function in the β-catenin degradation complex, one would predict that other components of the complex such as Axin would co-immunoprecipitate with hamartin and tuberin. To test this hypothesis, hamartin and tuberin were ectopically expressed in the presence of c-Myc-tagged Axin and then subjected to immunoprecipitation analyses. Using anti-tuberin and anti-c-Myc antibodies, tuberin and tagged Axin co-precipitated in samples where they were both overexpressed (Fig. 3 B,panels i and v, lanes 5 and6). Hamartin also co-immunoprecipitated with Axin (Fig.3 B, panel vi, lanes 5 and6). Tuberin, hamartin, and c-Myc-tagged Axin were not detected in vector control samples (Fig. 3 B, panels i, iii, v, and vi, lanes 1–3) or in samples without the tagged construct (Fig.3 B, panels i, iii, v, andvi, lane 4). We conclude that the tuberin-hamartin complex can associate with GSK3β and Axin possibly as part of the β-catenin degradation complex. Finally, to examine whether tuberin interacts with the endogenous GSK3β complex, co-immunoprecipitation assays were performed in HEK293T cells with and without Wnt stimulation. A band corresponding to GSK3β was found to co-immunoprecipitate with tuberin but not in preimmune serum control samples (Fig. 3 C, comparelanes 1 and 3). Furthermore, the amount of co-immunoprecipitated GSK3β was reduced upon Wnt-1 stimulation (Fig.3 C, compare lanes 1 and 2), suggesting that this interaction can be modulated by Wnt-1. The TSC1 and TSC2 tumor suppressor genes have recently been implicated to play a role in negatively regulating mTOR in the PI3K signaling cascade (23Goncharova E.A. Goncharov D.A. Eszterhas A. Hunter D.S. Glassberg M.K. Yeung R.S. Walker C.L. Noonan D. Kwiatkowski D.J. Chou M.M. Panettieri Jr., R.A. Krymskaya V.P. J. Biol. Chem. 2002; 277: 30958-30967Google Scholar, 24Manning B.D. Tee A.R. Logsdon M.N. Blenis J. Cantley L.C. Mol. Cell. 2002; 10: 151-162Google Scholar). As a result, tumors secondary to the inactivation of these genes have elevated levels of p70 S6 kinase activity that is reversible by rapamycin, a specific mTOR inhibitor (7Kenerson H.L. Aicher L.D. True L.D. Yeung R.S. Cancer Res. 2002; 62: 5645-5650Google Scholar). While this pathway may explain some of the complex phenotype exhibited by TSC pathology (i.e. cell size abnormalities), alteration in other cellular functions may involve additional mechanisms. Up-regulation of cyclin D1 mRNA and protein has been noted in CNS and renal lesions (6Crino P.B. Trojanowski J.Q. Dichter M.A. Eberwine J. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 14152-14157Google Scholar, 7Kenerson H.L. Aicher L.D. True L.D. Yeung R.S. Cancer Res. 2002; 62: 5645-5650Google Scholar), but a recent study using rapamycin in the Eker rat model of TSC failed to show a significant change in cyclin D1 or p27 levels despite anti-tumor response (7Kenerson H.L. Aicher L.D. True L.D. Yeung R.S. Cancer Res. 2002; 62: 5645-5650Google Scholar). Here, we describe an alternative pathway that may be relevant to the abnormalities observed in cell proliferation and differentiation. Our data show that in vivo levels of β-catenin are elevated upon disruption of TSC2 in renal tumors from Eker rats. Also, hamartin and tuberin reduce Wnt stimulation of β-catenin half-life and its downstream Tcf/LEF transcriptional activation. The results further suggest that tuberin and hamartin associate with the GSK3β degradation complex in a Wnt-1-dependent manner. These findings are in contrast to those of Kugoh et al. (25Kugoh H. Kleymenova E. Walker C.L. Mol. Carcinog. 2002; 33: 131-136Google Scholar) who reported a lack of change in Tcf-dependent luciferase activities in tuberin-null cells compared with controls. However, these experiments were conducted in the absence of Wnt stimulation. In our model, TSC1 and TSC2 complex with GSK3β to promote β-catenin degradation. Upon Wnt stimulation, tuberin dissociates from this complex resulting in stabilization of cytoplasmic β-catenin. As a known target of β-catenin signaling, the cyclin D1 gene contains Tcf responsive elements in its promoter and its expression is, in part, dependent on β-catenin activity (8Shtutman M. Zhurinsky J. Simcha I. Albanese C. D'Amico M. Pestell R. Ben-Ze'ev A. Proc. Natl. Acad. Sci. U. S. A. 1999; 96: 5522-5527Google Scholar, 9Tetsu O. McCormick F. Nature. 1999; 398: 422-426Google Scholar). We postulate that in the absence of functional tuberin or hamartin, stimulation of the β-catenin pathway will be unopposed resulting in the sustained transcriptional activation of cyclin D1. However, at this point, we cannot exclude the influence of the PI3K/Akt/mTOR pathway on cyclin D1 regulation. There is growing evidence that there exists substantial cross-talk between the Wnt and PI3K signaling pathways. For example, Wnt stimulation increases Akt activity resulting in GSK3β phosphorylation and β-catenin stabilization (26Fukumoto S. Hsieh C.M. Maemura K. Layne M.D. Yet S.F. Lee K.H. Matsui T. Rosenzweig A. Taylor W.G. Rubin J.S. Perrella M.A. Lee M.E. J. Biol. Chem. 2001; 276: 17479-17483Google Scholar). This function of Akt is distinct from its effects on the PI3K pathway and is dependent on the recruitment of Dsh to the GSK3β-β-catenin-Axin complex (26Fukumoto S. Hsieh C.M. Maemura K. Layne M.D. Yet S.F. Lee K.H. Matsui T. Rosenzweig A. Taylor W.G. Rubin J.S. Perrella M.A. Lee M.E. J. Biol. Chem. 2001; 276: 17479-17483Google Scholar). It is conceivable that upon stimulation, activated Akt phosphorylates tuberin and disables it from the complex. Accordingly, tuberin acting downstream of Akt may play a role in coordinating signals tranduced through the Wnt and PI3K pathways, thus providing a mechanism for the pleiotropic effects of the TSC1 and TSC2 genes in tuberous sclerosis. The molecular components and their regulation by which TSC1 and TSC2 interact with the GSK3β complex remain to be identified. We thank David Kimelman for providing the GSK3β construct and members of the Yeung laboratory for critical reading and assistance in the preparation of the manuscript.

Referência(s)