De-Chang Wu and Hong Wu Liu, Dan Freeman, Jing ...

3 downloads 0 Views 693KB Size Report
Feb 28, 2006 - suppression of the growth and the tumorigenicity of Pten null. MEFs. ... and glucose metabolism through phosphorylation of its substrates (8, 9).
Mechanisms of Signal Transduction: PTEN Deletion Leads to Up-regulation of a Secreted Growth Factor Pleiotrophin Gang Li, Yingchun Hu, Yanying Huo, Minli Liu, Dan Freeman, Jing Gao, Xin Liu, De-Chang Wu and Hong Wu J. Biol. Chem. 2006, 281:10663-10668. doi: 10.1074/jbc.M512509200 originally published online February 28, 2006

Access the most updated version of this article at doi: 10.1074/jbc.M512509200 Find articles, minireviews, Reflections and Classics on similar topics on the JBC Affinity Sites. Alerts: • When this article is cited • When a correction for this article is posted Click here to choose from all of JBC's e-mail alerts This article cites 52 references, 30 of which can be accessed free at http://www.jbc.org/content/281/16/10663.full.html#ref-list-1

Downloaded from http://www.jbc.org/ at UMDNJ on September 11, 2013

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 16, pp. 10663–10668, April 21, 2006 © 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

PTEN Deletion Leads to Up-regulation of a Secreted Growth Factor Pleiotrophin* Received for publication, November 22, 2005, and in revised form, January 11, 2006 Published, JBC Papers in Press, February 28, 2006, DOI 10.1074/jbc.M512509200

Gang Li‡§1, Yingchun Hu§, Yanying Huo§, Minli Liu§, Dan Freeman‡2, Jing Gao‡, Xin Liu‡, De-Chang Wu§, and Hong Wu‡3 From the ‡Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, California 90095-1735 and the §Institute of Radiation Medicine, Beijing 100850, China Tumor suppressor gene PTEN is highly mutated in a wide variety of human tumors. To identify unknown targets or signal transduction pathways that are regulated by PTEN, microarray analysis was performed to compare the gene expression profiles of Pten null mouse embryonic fibroblasts (MEFs) cell lines and their isogenic counterparts. Expression of a heparin binding growth factor, pleiotrophin (Ptn), was found to be up-regulated in Ptenⴚ/ⴚ MEFs as well as Pten null mammary tumors. Further experiments revealed that Ptn expression is regulated by the PTEN-PI3K-AKT pathway. Knocking down the expression of Ptn by small interfering RNA resulted in the reduction of Akt and GSK-3␤ phosphorylation and suppression of the growth and the tumorigenicity of Pten null MEFs. Our results suggest that PTN participates in tumorigenesis caused by PTEN loss and PTN may be a potential target for anticancer therapy, especially for those tumors with PTEN deficiencies.

PTEN4 (phosphatase and tensin homologue deleted on chromosome ten) is the first phosphatase identified as a tumor suppressor (1–3). Loss of function mutations or reduced expression of the PTEN gene are found at high frequencies in a wide variety of human tumors, including glioblastoma, as well as endometrial, prostate, colorectal, lung, and breast cancers. Experimental and clinical evidence demonstrate that PTEN is a critical tumor suppressor (4). PTEN acts primarily as a negative regulator of the phosphoinositide 3-kinase (PI3K) pathway by virtue of its lipid phosphatase activity (5–7). Loss of PTEN leads to an increase in the phosphatidylinositol 3-phosphate level, mimicking the effect of constitutive PI3K activation. Phosphatidylinositol 3-phosphate accumulation results in the activation of various protein kinases, including the PDK1 and PKB/AKT serine/threonine kinases. AKT is the central node in the PTEN-regulated pathway and activated AKT has been shown to promote cell cycle progression,

* This work was supported in part by National Natural Science Foundation of China Grants 3000201 and 30471946, Beijing Natural Science Foundation Grant 7052056 (to G. L.), Department of Defense Grant PC031130 and National Institutes of Health NCI Grants UO1 CA84128 – 06 and RO1 CA107166 (for H. W.). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 Supported by the Howard Hughes Medical Institute. To whom correspondence may be addressed. Tel.: 8610-66932209; E-mail: [email protected]. 2 Supported by a Pre-doctoral Research Training Grant in Pharmacological Sciences. 3 To whom correspondence may be addressed. Tel.: 310-825-5160; Fax: 310-267-0242; E-mail: [email protected]. 4 The abbreviations used are: PTEN, phosphatase and tensin homologue deleted from chromosome 10; PI3K, phosphatidylinositol 3-kinase; PTN, pleiotrophin; PDK1, 3-phosphoinositide-dependent protein kinase-1; PKB/Akt, v-akt murine thymoma viral oncogene homolog 1; MSPB58, 58-kDa microspherule protein; MEFs, mouse embryonic fibroblasts; aa-dUTP, aminoallyl-dUTP; siRNA, small interfering RNA; WT, wild-type; EGFP, enhanced green fluorescent protein; GSK-3␤, glycogen synthase kinase 3␤.

cell growth, cell survival, cell migration, angiogenesis, protein synthesis, and glucose metabolism through phosphorylation of its substrates (8, 9). Aside from acting as a negative regulator of the PI3K pathway, which depends on its lipid phosphatase activity, PTEN also functions independently of its lipid phosphatase activity. Raftopoulou et al. (10) showed that PTEN inhibits cell migration through its C2 domain and dephosphorylates itself depending on its protein phosphatase activity. Freeman et al. (11) demonstrated that PTEN physically associates with p53 in the nucleus, which in turn stabilizes p53 and affects p53 protein levels and transcription activity. Recently, Okumura (12) reports that the PTEN C-terminal domain physically interacts with the oncogenic MSP58 protein and suppresses cell transformation induced by MSP58 expression, independent of its catalytically activity. To further understand the biological functions of PTEN, we undertook an unbiased approach by comparing gene expression profiles of a Pten⫺/⫺ mouse embryonic fibroblast (MEF) cell line with that of its isogenic counterpart (11, 13) using microarray technology. Among genes that are differentially expressed in these cell lines, we found that pleiotrophin (Ptn) was up-regulated in the Pten⫺/⫺ MEFs as well as mammary tumor tissues of Pten conditional knock-out mice (14). Further experiments indicated that the expression of Ptn was regulated by the PI3K pathway, and Ptn overexpression contributes to tumorigenesis caused by PTEN loss.

MATERIALS AND METHODS Cell Culture and Generation of Immortalized Fibroblasts—Fibroblasts were grown in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal calf serum, streptomycin, and penicillin, at 37 °C in 5% CO2. Immortalized wild-type (Pten⫹/⫹) and Pten⫺/⫺ mouse embryonic fibroblast cells were generated independently according to the 3T9 protocol (13). The Pten⌬loxp/⌬loxp (Pten⌬/⌬) cell line was generated by infecting immortalized PtenLoxP/LoxP (PtenL/L) mouse embryonic fibroblasts with an adenovirus vector expressing Cre recombinase (11). Pten⌬/⌬-241 cell was clonally derived from a tumor formed on nude mice by Pten⌬/⌬ MEFs. Microarray Analysis—Mouse cDNA microarrays containing 8,928 elements were printed by the UCLA Microarray Core Facility. Total RNA was isolated using TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. For probe labeling and microarray hybridization, a protocol published by Xiang et al. (15) was adopted with minor modifications. Briefly, cDNA probes that incorporate aminoallyl-dUTP (aa-dUTP; Sigma) were synthesized by reverse transcription using 10 ␮g of total RNA in a 30-␮l reaction volume, containing oligo(dT) primer (0.5 ␮g/␮l, 2 ␮l), RNase inhibitor (Promega, 5 units/␮l, 1 ␮l), 1⫻ first-strand buffer (Invitrogen), 0.5 mM dATP/dGTP/dCTP, 0.3 mM dTTP, 0.2 mM aa-dUTP, 10 mM dithiothreitol, and SuperScript II reverse transcriptase (Invitrogen, 1.9 ␮l). The reactions were incubated at 42 °C for 2 h and

APRIL 21, 2006 • VOLUME 281 • NUMBERDownloaded 16 from http://www.jbc.org/ at UMDNJ on September JOURNAL 11, 2013 OF BIOLOGICAL CHEMISTRY

10663

PTEN Negatively Regulates Pleiotrophin Expression terminated by addition of EDTA (0.5 M, 10 ␮l). The RNA was hydrolyzed with NaOH (1 M, 10 ␮l) and the solution was neutralized with HCl (1 M, 10 ␮l), and then Microcon 30 concentrators (Millipore) were used to clean up the cDNA. Purified cDNA from Pten null and wild-type MEFs was labeled with either Cy5 or Cy3 monoreactive fluors (Amersham Biosciences), combined, and competitively hybridized to microarrays under a coverslip for 16 h at 65 °C. Microarray slides were scanned using the GenePix 4000A fluorescent scanner (Axon Instruments), variable photomultiplier tube voltage settings were used to obtain the maximal signal intensities with ⬍1% probe saturation and balanced intensities between the two channels (ratio of medians is ⬃1). The experiment was performed in duplicate using independently isolated RNA samples. Raw data files were generated and analyzed using the GenePix 3.0 microarray analysis software, then imported into Excel. Features for which R2 values were below 0.4 and less than 50% of feature pixels that were 1 S.D. above background pixels in either channel were filtered out. Features with ratio of medians ⱖ2 were considered to be up-regulated and features with ratio of medians ⱕ0.5 were considered to be down-regulated. Extraction of RNA and Northern Blot Hybridization—RNA was isolated using TRIzol reagent (Invitrogen) according to the procedure provided by the manufacturer. Total RNA was separated on 1.0% agarose gels in the presence of formaldehyde, transferred to Hybond-N⫹ nylon membranes (Amersham Biosciences), and hybridized in QuikHyb威 hybridization solution (Stratagene). Probes were labeled with [32P]dCTP using the random priming kit (Prime-It威 II, Stratagene). DNA Constructs and Transfection—Constructs containing wild-type Pten and the C124S mutant were generated by ligating corresponding Pten cDNA into BglII and SalI sites of the pIRES2-EGFP vector (Clontech). Constructs containing wild-type (WT) Akt1 and dominant negative Akt1 were generated by cutting Akt1 cDNAs in pUSEamp (Upstate) with BamHI and PmeI, and ligating into BglII and SmaI sites of the pIRES2-EGFP vector. For constructs expressing activated Akt1, a PCR fragment containing activated Akt1 (16) was ligated into EcoRI and SmaI sites of the pIRES2-EGFP vector. DNA constructs were transfected into MEFs and 3T3 cells using Superfect transfection reagent (Qiagen). Inhibitor Treatments—MEF1 cells were treated with 10 and 30 ␮M phosphoinositide 3-kinase inhibitor, LY294002 (BIOMOL), and 50 ␮M mitogen-activated protein kinase kinase (MEK)-specific inhibitor, PD98059 (BIOMOL), for 24 h, then subjected to Northern hybridization. siRNA—Three siRNAs were designed to target the open reading frame of the mouse Ptn gene at site A (131–149 nucleotides), B (243– 261 nucleotides), and C (325–343 nucleotides). The oligonucleotides used were: (A) sense, 5⬘-GATCCCAGTCTGACTGTGGAGAATGttcaagagaCATTCTCCACAGTCAGACTttttttGGAAA-3⬘ and antisense, 3⬘-GGTCAGACTGACACCTCTTACaagttctctGTAAGAGGTGTCAGTCTGAaaaaaaCCTTTTCGA-5⬘; (B) sense, 5⬘-GATCCCGACTCAGAGATGTAAGATCttcaagagaGATCTTACATCTCTGAGTCttttttGGAAA-3⬘ and antisense, 3⬘-GGCTGAGTCTCTACATTCTAGaagttctctCTAGAATGTAGAGACTCAGaaaaaaCCTTTTCGA-5⬘; (C) sense, 5⬘-GATCCCGTGTGACCTCAATACCGCCTttcaagagaAGGCGGTATTGAGGTCACAttttttGGAAA-3⬘ and antisense, 3⬘-GGCACACTGGAGTTATGGCGGAaagttctctTCCGCCATAACTCCAGTGTaaaaaaCCTTTTCGA-5⬘. These oligonucleotides were annealed and subcloned to the downstream H1 promoter in pSilencerTM 3.1-H1 hygro vectors (Ambion) using BamHI and HindIII. To test the knocking down abilities of the 3 siRNAs against Ptn, plasmids carrying Ptn-siRNA

A, B, or C, or siRNA negative control (Ambion) were transiently transfected into 3T3 cells using Lipofectamime 2000 (Invitrogen), the cells were subjected to Northern hybridization using Ptn cDNA as probe. To generate cell lines stably expressing Ptn siRNA, 2 ⫻ 105 Pten⌬/⌬-241 cells were transfected with plasmids carrying Ptn-siRNA A, B, C, or control siRNA, respectively, and cultured for 48 h. The cells were then selected with 200 ␮g/ml hygromycin for 2 weeks; the obtained clones were screened for Ptn knocking down by Northern hybridization. Cell Growth Curve—Cells were seeded into 24-well plates in triplicates at a density of 8 ⫻ 103 per well, the number of live cells was determined by trypan blue staining and was scored daily for 7 days with a hematocytometer under an inverted microscope. Statistical Analysis—Microsoft Excel was used to analyze the data and plot curves. Analysis of variance was applied for multiple comparisons. Tumorigenecity in Nude Mice—Athymic female nude mice were each injected subcutaneously with 4 ⫻ 106 Pten⌬/⌬-241 cells stably expressing Ptn siRNA B or control siRNA (n ⫽ 8 for each group). The mice were kept in pathogen-free environments and checked every 2 days. The dates, at which a palpable tumor first arose, and the weights of the tumors were recorded. Western Blot Analysis—Protein was extracted using RIPA buffer (1⫻ phosphate-buffered saline, 1% Nonidet P-40, 0.5% sodium dexoycholate, 0.1% SDS and protease inhibitor mixture tablet (Roche)) and fractionated on SDS-polyacrylamide gels. Western blots were then probed with the antibodies against PTN (R&D Systems), and AKT, phospho-AKT (Thr-308), phospho-GSK-3␤ and ␤-actin, which were all purchased from Cell Signaling Technology. Horseradish peroxidaseconjugated secondary antibodies and LumiGLOTM reagent (Cell Signaling Technology) were used to detect specific binding, and signals were captured by x-ray film.

RESULTS Genes Differentially Expressed in Pten WT and Null MEF Cell Lines— To identify unknown targets or signal transduction pathways that are regulated by PTEN, we compared gene expression profiles of Pten WT and null mouse embryonic fibroblast cell lines (7, 13). Sixteen genes were found to be up-regulated with ratio of medians above 2.0 (Table 1), whereas 30 genes were found to be down-regulated with ratio of medians below 0.5 (Table 2), based on two independent microarray analysis. Eight of 10 differentially expressed genes could be confirmed by other means, such as Northern blot analysis, reverse transcriptase-PCR, or Western blot analysis (data not shown), indicating the high quality of our microarray analysis. We further checked the expression statuses of these genes in another line of Pten null MEFs, which is designated as Pten⌬/⌬. The Pten⌬/⌬ cell line was generated by transfecting immortalized PtenLoxP/LoxP (PtenL/L) mouse embryonic fibroblasts with an adenovirus vector expressing Cre recombinase. Ptn, among several other genes (data not shown), was also up-regulated in Pten⌬/⌬ cells (Fig. 1A). An interesting observation to be noted is that up-regulation of Ptn in Pten-null MEFs is cell density-dependent: the differences in Ptn level between WT and Pten⫺/⫺ MEF cells are more significant when cells reach high density. As shown in Fig. 1B, there was no difference in Ptn expression level between WT and Pten⫺/⫺ MEF cells when they are on log phase, but significant difference was observed when they reached confluence, and a more significant difference was observed 2 days after they reached confluence (Fig. 1B). Because PTN, a secreted growth factor, can promote cell proliferation, migration, and angiogenesis (17–

Paper 8

10664 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 16 • APRIL 21, 2006 Downloaded from http://www.jbc.org/ at UMDNJ on September 11, 2013

PTEN Negatively Regulates Pleiotrophin Expression TABLE 1 Genes that are up-regulated in Ptenⴚ/ⴚMEF cells The cDNA of Pten⫺/⫺ MEF cells was labeled with Cy5, and the cDNA of WT MEF was labeled with Cy3, the ratio of medians is Cy5/Cy3, the ratios of medians from two independent experiments are shown here. The expression statuses of the genes in bold had been confirmed by Northern hybridization. Gene symbol

Description

Unigene No.

Cxcl12 (Sdf1) Ccnd2 Trim10 (Rnf9) Fn1 Timp3 Slpi Zfp36l1 (Brf1) Bicc1 Ptn Rbp1 Serpine2 Sepp1 Slc39a10 Pde7b Snx5 Scamp2

Chemokine (C-X-C motif) ligand 12 Cyclin D2 Tripartite motif protein 10 Fibronectin 1 Tissue inhibitor of metalloproteinase 3 Secretory leukocyte protease inhibitor Zinc finger protein 36, C3H type-like 1 Bicc1: Bicaudal C homolog 1 (Drosophila) Ptn Retinol-binding protein 1, cellular Serine (or cysteine) proteinase inhibitor, clade E, member 2 Selenoprotein P, plasma, 1 Solute carrier family 39 (zinc transporter), member 10 Phosphodiesterase 7B Sorting nexin 5 Secretory carrier membrane protein 2

Mm.303231 Mm.333406 Mm.299155 Mm.193099 Mm.4871 Mm.371583 Mm.235132 Mm.286834 Mm.279690 Mm.279741 Mm.3093 Mm.22699 Mm.233889 Mm.370216 Mm.273379 Mm.286069

Ratio of medians A

B

10.629 4.892 4.499 4.264 3.93 3.797 3.458 2.979 2.966 2.454 2.358 2.293 2.218 2.161 2.148 2.091

11.256 4.088 3.818 3.495 3.265 1.839 2.576 2.713 3.345 2.438 2.226 2.48 2.125 2.124 2.04 3.662

TABLE 2 Genes that are down-regulated in Ptenⴚ/ⴚ MEF cells The cDNA of Pten⫺/⫺ MEF cells was labeled with Cy5, and the cDNA of WT MEF was labeled with Cy3, the ratio of medians is Cy5/Cy3, the ratios of medians from two independent experiments are shown here. The expression statuses of the genes in bold had been confirmed by Northern hybridization. Symbol

Description

Unigene No.

Mmp2 Npepl1 Zfp148 Foxo1 Bat8 Scarb2 E430026A01Rik Sdcbp2 Ly6c Pdlim2 Ap3s2 Lgals3 Gsta3 Cnn1 1110013G13Rik Ube2i Cyp2f2 Rnf144 Pte2a Cyr61 Gpr125 9030409G11Rik: 2310047C17Rik H2-Bf Akp2 Prpf3 Tfpi Dab2ip Hrb2 BC034204

Matrix metalloproteinase 2 Aminopeptidase-like 1 Zinc finger protein 148 Forkhead box O1 HLA-B-associated transcript 8 Scavenger receptor class B, member 2 RIKEN cDNA E430026A01 gene Syndecan-binding protein (syntenin) 2 Lymphocyte antigen 6 complex, locus C PDZ and LIM domain 2 Adaptor-related protein complex 3, ␴-2 subunit Lectin, galactose binding, soluble 3 Glutathione S-transferase, ␣3 Calponin 1 RIKEN cDNA 1110013G13 gene Ubiquitin-conjugating enzyme E2I Cytochrome P450, family 2, subfamily f, polypeptide 2 Ring finger protein 144 Peroxisomal acyl-CoA thioesterase 2A Cysteine rich protein 61 G protein-coupled receptor 125 RIKEN cDNA 9030409G11 gene RIKEN cDNA 2310047C17 gene Histocompatibility 2, complement component factor B Alkaline phosphatase 2, liver PRP3 pre-mRNA processing factor 3 homolog (yeast) Tissue factor pathway inhibitor Disabled homolog 2 (Drosophila) interacting protein HIV-1 Rev-binding protein 2 CDNA sequence BC034204

Mm.29564 Mm.295629 Mm.256809 Mm.29891 Mm.35345 Mm.297964 Mm.29402 Mm.32068 Mm.1583 Mm.283968 Mm.220173 Mm.248615 Mm.14719 Mm.4356 Mm.210305 Mm.240044 Mm.4515 Mm.214932 Mm.202331 Mm.1231 Mm.272974 Mm.255986 Mm.203866 Mm.653 Mm.288186 Mm.279872 Mm.124316 Mm.29629 Mm.34606 Mm.3957

19), properties closely linked to phenotypes associated with PTEN deficiency, we decided to conduct further studies on its regulation. Ptn Is Up-regulated in the Mammary Tumors of Pten-conditional Knock-out Mice—Next we tested if PTEN deficiency could lead to Ptn up-regulation in an in vivo model. PtenLoxp/Loxp;MMTVCre⫹/⫺ mice generated in our laboratory carry Pten conditionally deleted in mammary gland (14), Ptn mRNA expression levels increased strikingly in mammary tumors of the PtenLoxp/Loxp;MMTVCre⫹/⫺ mice, as compared with the WT controls (Fig. 2A). The increase of Ptn expression in mammary tumors of the mutant mice could also be detected by Western blots (Fig. 2B), suggesting that Ptn overexpression may either correlate with or participate in tumorigenesis caused by PTEN loss.

Ratio of medians A

B

0.239 0.26 0.264 0.281 0.282 0.29 0.315 0.349 0.359 0.36 0.366 0.366 0.366 0.372 0.375 0.385 0.398 0.406 0.409 0.414 0.44 0.44 0.444 0.453 0.454 0.457 0.474 0.482 0.486 0.498

0.382 0.274 0.26 0.304 0.421 0.456 0.222 0.182 0.353 0.444 0.217 0.473 0.151 0.297 0.129 0.294 0.309 0.32 0.33 0.259 0.311 0.451 0.278 0.47 0.318 0.475 0.308 0.355 0.334 0.451

Ptn Expression Is Regulated by PI3K/AKT Pathways—We then determined which pathway was involved in Ptn transcription regulation using various pathway-specific inhibitors. As shown in Fig. 3A, Ptn expression was significantly down-regulated by the PI3K inhibitor LY294002 at a concentration of 30 ␮M (Fig. 3A, third lane; quantification shown on the right panel). On the other hand, no significant changes of Ptn expression were seen when the mitogen-activated protein kinase kinase-specific inhibitor PD98059 was used, indicating that the expression of Ptn is regulated by PI3K pathways. Chemical inhibitor treatment can only provide a rough picture on how Ptn is regulated. To further define the pathways that may be involved in Ptn regulation, we transfected various Pten constructs into

Paper 8

APRIL 21, 2006 • VOLUME 281 • NUMBERDownloaded 16 from http://www.jbc.org/ at UMDNJ on September JOURNAL 11, 2013 OF BIOLOGICAL CHEMISTRY

10665

PTEN Negatively Regulates Pleiotrophin Expression

FIGURE 1. Pten deletion leads to up-regulation of Ptn. A, pleiotrophin (Ptn) was found to be up-regulated in 2 independent Pten null MEF cell lines, Pten⫺/⫺ and Pten⌬/⌬. The amount of ribosomal RNAs was used as loading control. B, Ptn expression is subject to cell density control. Total RNAs from Pten⫹/⫹ and Pten⫺/⫺ MEF cells at log phase (Log), 100% confluence (Conf.), and 2 days after confluence (2D after Conf.) were extracted, and subjected to Northern hybridization. The amount of ribosomal RNAs was used as loading control.

FIGURE 2. Ptn is up-regulated in the mammary tumors of Pten conditional knockout mice. A, total RNA was extracted from the virgin mammary glands of 10-week-old control (PtenLoxp/Loxp;MMTVCre⫺/⫺) and the mammary tumors arisen from the PtenLoxp/Loxp; MMTVCre⫹/⫺ mice. The RNAs were blotted to nylon membranes and subjected to Northern hybridization with the Ptn probe. The amount of ribosomal RNAs was used as loading control. B, protein was extracted from the same tissue as in Northern blots and subjected to Western blot analysis, and ␤-actin was used as loading control.

3T3 cells. As shown in Fig. 3B, transfection of wild-type Pten into 3T3 cells could suppress the expression of Ptn, whereas a non-functional, mutated form of Pten (C124S phosphatase-dead mutation) had no effect. Overexpression of wild-type and the activated form of Akt-1 also resulted in up-regulation of Ptn, whereas expression of a dominant-negative form of Akt-1 resulted in a down-regulation of Ptn expression (Fig. 3C), indicating that regulation of Ptn by PTEN is PI3K/AKT-dependent. siRNA against Ptn Could Suppress the Malignant Phenotypes of Pten Null Cells—To assess the tumorigenicities of the Pten null cell lines in vivo, we injected Pten⌬/⌬ MEFs into nude mice subcutaneously, and observed the mice for 4 months. Pten⌬/⌬ MEFs could form tumors in nude mice with a latency of greater than 2.5 months. We then dissociated tumors and cultured the tumor cells in vitro. A clonally derived line, Pten⌬/⌬-241, was used for study as described below because of its shorter tumor forming latency. To test if knocking down the expression of Ptn can reverse the malignant phenotypes of the Pten⌬/⌬-241 cells, we designed 3 siRNAs against the mouse Ptn gene. To test the knocking down abilities of the siRNAs against Ptn, 3T3 cells were transiently transfected with the plasmids expressing the Ptn siRNAs and subjected to Northern blot analysis. As shown in the left panel of Fig. 4A, Ptn-siRNA B and C had better knocking down abilities than Ptn-siRNA A. Plasmids containing Ptn-siRNA B

FIGURE 3. Ptn expression is regulated by PI3K/AKT pathway. A, MEF cells were treated with phosphoinositide 3-kinase inhibitor LY294002 (10 ␮M, LY10; and 30 ␮M, LY30) or mitogen-activated protein kinase kinase (MEK) inhibitor PD98059 (50 ␮M) for 24 h, then subjected to Northern hybridization. B, PTEN suppresses the expression of Ptn. 3T3 cells were transfected with the indicated constructs, and subjected to Northern analysis 24 h after transfection. Pten WT, construct containing wild-type Pten cDNA; Pten CS, construct containing Pten cDNA with the C124S point mutation that lacks phosphatase activity. Empty pIRES2-EGFP vector was transfected as control. C, overexpression of wild-type or the activated form of AKT results in up-regulation of Ptn. 3T3 cells were transfected with the indicated constructs, and subjected to Northern analysis 24 h after transfection. Akt1 WT, construct containing wild-type Akt1 cDNA; Akt1 DN, construct containing dominant negative Akt1 cDNA; Akt1 AC, construct containing activated form of Akt1 cDNA. Empty pIRES2-EGFP vector was transfected as control. The amount of ribosomal RNAs was used as loading control in each panel. Signal intensities of the Northern blot were quantified by Quantity One (Bio-Rad), normalized by the intensities of 28 S ribosomal RNA, and graphed at the right.

or C were then stably transfected into Pten⌬/⌬-241 cells, 3 clones for Ptn-siRNA B and 2 clones for Ptn-siRNA C were picked, and Ptn expression levels were tested by Northern blots. The expression of Ptn in clone 2 of Ptn-siRNA B was almost totally knocked down, whereas clone 2 of Ptn-siRNA C had less reduction (Fig. 4A, right panel). We then measured growth properties of these two clones and compared the control siRNA-transfected cells (Fig. 4B). Consistent with expression levels of Ptn seen in Fig. 4B, the growth rate of clone 2 of Ptn-siRNA B was significantly decreased, whereas the growth rate of clone 2 of Ptn-siRNA C showed mild reduction, indicating that reduction of Ptn expression could inhibit the growth of Pten⌬/⌬-241 cells. We then injected clone 2 of Ptn-siRNA B into nude mice. For the mice injected with Pten⌬/⌬-241 cells transfected with control siRNA, 5 of 8 developed tumors with a latency of 21 to 47 days; whereas in mice injected with clone 2 of Ptn-siRNA B, only one of eight had tumor with latency of more 90 days (Table 3). Taken together, these results indicate that knocking down the expression of Ptn could suppress the tumorigenicity of Pten null cells and overexpression of Ptn may indeed participate in tumorigenesis caused by PTEN loss. Down-regulation of AKT Activity by Ptn siRNA—Previous studies have shown that PTN can induce the activation of the PI 3-kinase pathway in bovine epithelial lens cells (20), U87 glioblastoma cells (21), 3T3 cells (22), and human umbilical vein endothelial cells (23). So we examined the status of the PI 3-kinase pathway in the Pten⌬/⌬-241 cells transfected with Ptn-siRNA vectors. Consistent with previous studies, the phosphorylation levels of AKT and GSK-3␤ were significantly reduced in the Pten⌬/⌬-241 cells with Ptn knocked down (Fig. 4C), indicating that

Paper 8

10666 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 16 • APRIL 21, 2006 Downloaded from http://www.jbc.org/ at UMDNJ on September 11, 2013

PTEN Negatively Regulates Pleiotrophin Expression

FIGURE 4. siRNA against Ptn could suppress tumorigenicity of Ptenⴚ/ⴚ cells. A, testing the knocking down abilities of the siRNAs against Ptn by Northern blots. Left panel, 3T3 cells were transiently transfected with plasmids expressing the indicated Ptn siRNAs and negative control siRNA (provided by Ambion), then subjected to Northern blot analysis. Right panel, Pten⌬/⌬-241 cells were stably transfected with plasmids containing PtnsiRNA B or C or negative control. Three clones for Ptn-siRNA B, 2 clones for Ptn-siRNA C, 1 clone for negative control were picked and their Ptn expression levels were examined by Northern blots. B, reduction of Ptn expression resulted in inhibition of the growth of Pten⌬/⌬-241 cells. Cells were seeded into 24-well plates in triplicates at a density of 8 ⫻ 103 per well and cell numbers were counted daily for 7 days. The data shown are mean ⫾ S.D. from three independent experiments. C, reduction of Ptn expression resulted in the downregulations of AKT and GSK-3␤ phosphorylations. Wild-type MEF cells and Pten⌬/⌬-241 cells stably transfected with the indicated siRNA vector were subjected to Western blot analysis and ␤-actin was used as loading control.

TABLE 3 siRNA directed against Ptn significantly inhibits Pten⌬/⌬-241 cell growth in nude mice SiRNA Negative control Ptn siRNA B a b

Injections 8 8

Tumors 5 1

Tumor weight

Latency

g

Days

0.1239–1.522a 0.4125b

21–47 ⬎90

Weighed at 2 months after injection. Weighed at 4 months and 10 days after injection.

Ptn plays an important positive feedback role in maintaining the activation of the PI 3-kinase pathway in Pten⌬/⌬-241 cells.

DISCUSSION Many laboratories have used microarray analysis to identify PTENregulated genes (24 –30) although the data reported so far are rarely consistent. Of 709 genes identified to be suppressed by Pten overexpression or up-regulated due to Pten deficiency in the above mentioned papers, only Cyclin B1 (27, 30) and MYBL2 (25, 27) showed consistent results by more than one laboratory; whereas totally opposing results about Drg-1 were reported (25, 27, 31). Several possibilities could

explain these discrepancies. First, PTEN could regulate different sets of genes in different cell types, tissues, or systems. Second, additional genetic alterations may be introduced during the process of model establishing, which is not directly related with PTEN expression. For example, alterations of p53 or p19 functions are usually associated with immortalizing MEFs (32). Obviously, shortcomings of the microarray technique itself, because of nonspecific binding caused by homologies between different genes, could contribute to the noise level of microarray analysis. Due to the reasons mentioned above, the genes found to be differentially expressed in different systems is more likely to be the real candidates regulated by PTEN. We found that Ptn was up-regulated in two independent Pten null MEF cell lines as well as in Pten null mammary tumor tissues. We also demonstrated that the expression of Ptn is PI3K/ AKT pathway-dependent. PTN is a heparin binding secreted growth/differentiation factor that has diverse functions: being involved in cell activities of adhesion, migration, survival, growth, and differentiation (for reviews, see Refs. 17–19). Its growth and differentiation promoting activities may play a role in the precocious development observed in skin and mammary glands of Pten conditional knock-out mice, in which precocious hair follicle morphogenesis (33), excessive ductal branching, precocious lobulo-alveolar development, and pregnancy-associated milk-specific protein expression were observed (14). Stable transfection of Ptn resulted in oncogenic transformation of 3T3 cells and highly vascularized tumor formation in nude mice (34, 35). Ribozyme targeting of PTN suppresses the growth, angiogenesis, and metastasis of melanoma (36, 37) and pancreatic cancer cells (38), whereas overexpression of PTN has been observed in a variety of cancers (17–19). Because PTN is a secreted protein and can be detected in serum by enzyme-linked immunosorbent assay (39), PTN might be a tumor marker and has diagnostic value. Indeed, elevated serum PTN levels were found in patients with pancreatic cancer (39, 40), colon cancer (39), testicular cancer (41), lung cancer (42), and astrocytomas (43). Based on our observation PTEN loss led to overexpression of PTN in vitro in cell culture systems and in vivo in the mammary tumors, and PTN plays a critical positive feedback role in controlling AKT activity. Further studies are worthwhile to determine whether PTN can be used as a predictive marker of the PTEN/PI3K/AKT pathway activation or expression status of Pten in animal models and clinical samples. PTEN regulates the expression of the gene through various transcription factors, including hypoxia-inducible factor-1 (44), forkhead transcription factors (45), tumor suppressor p53 (11), NF␬B (46), and ␤-catenin (47). Searching the transcription factor binding sites on the mouse Ptn promoter using the on-line tool Mat Inspector revealed that there are forkhead transcription factor FOXF2 and ␤-catenin/TCF/ LEF-1 binding sites on the Ptn promoter. Whether or not they are involved in the up-regulation of Ptn in the Pten null MEFs and tumors need further investigation. Of note, the difference of PTN expression between Pten null cells and control is marginal when cells were cultured in low densities, whereas there was a significant difference in Ptn expression when cells reached high density, suggesting other signaling pathways sensitive to cell densities, or insensitive to contact inhibition, may be critical for the expression of Ptn. In this paper we also showed knocking down the expression of Ptn could suppress the growth and tumorigenicity of Pten⌬/⌬-241 cells. Knocking down the expression of Ptn also resulted in the reduction of phosphorylation of Akt and GSK-3, suggesting that PTN played an important role in maintaining the activation of the PI 3-kinase pathway in Pten⌬/⌬-241 cells. PTN has been suggested to be a potential new

Paper 8

APRIL 21, 2006 • VOLUME 281 • NUMBERDownloaded 16 from http://www.jbc.org/ at UMDNJ on September JOURNAL 11, 2013 OF BIOLOGICAL CHEMISTRY

10667

PTEN Negatively Regulates Pleiotrophin Expression target for the treatment or/and diagnosis of several types of cancer (48). Gene therapy approaches of targeting PTN in established mouse tumor models using ribozymes (36, 37, 49 –51) or antisense oligonucleotides (52) have been reported. Deficiency of PTEN has been linked with resistance to chemotherapeutic drugs, such as trastuzumab (Herceptin) (53), our data suggests that PTN-targeting therapies might be one of the candidates to overcome this resistance. Acknowledgments—We thank members of our laboratories for critical reading of the manuscript and suggestions and B. Zhang for technical assistance.

REFERENCES 1. Li, J., Yen, C., Liaw, D., Podsypanina, K., Bose, S., Wang, S. I., Puc, J., Miliaresis, C., Rodgers, L., McCombie, R., Bigner, S. H., Giovanella, B. C., Ittmann, M., Tycko, B., Hibshoosh, H., Wigler, M. H., and Parsons, R. (1997) Science 275, 1943–1947 2. Li, D. M., and Sun, H. (1997) Cancer Res. 57, 2124 –2129 3. Steck, P. A., Pershouse, M. A., Jasser, S. A., Yung, W. K., Lin, H., Ligon, A. H., Langford, L. A., Baumgard, M. L., Hattier, T., Davis, T., Frye, C., Hu, R., Swedlund, B., Teng, D. H., and Tavtigian, S. V. (1997) Nat. Genet. 15, 356 –362 4. Parsons, R. (2004) Semin. Cell Dev. Biol. 15, 171–176 5. Maehama, T., and Dixon, J. E. (1998) J. Biol. Chem. 273, 13375–13378 6. Stambolic, V., Suzuki, A., de la Pompa, J. L., Brothers, G. M., Mirtsos, C., Sasaki, T., Ruland, J., Penninger, J. M., Siderovski, D. P., and Mak, T. W. (1998) Cell 95, 29 –39 7. Sun, H., Lesche, R., Li, D. M., Liliental, J., Zhang, H., Gao, J., Gavrilova, N., Mueller, B., Liu, X., and Wu, H. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 6199 – 6204 8. Di Cristofano, A., and Pandolfi, P. P. (2000) Cell 100, 387–390 9. Vivanco, I., and Sawyers, C. L. (2002) Nat. Rev. Cancer 2, 489 –501 10. Raftopoulou, M., Etienne-Manneville, S., Self, A., Nicholls, S., and Hall, A. (2004) Science 303, 1179 –1181 11. Freeman, D. J., Li, A. G., Wei, G., Li, H. H., Kertesz, N., Lesche, R., Whale, A. D., Martinez-Diaz, H., Rozengurt, N., Cardiff, R. D., Liu, X., and Wu, H. (2003) Cancer Cell 3, 117–130 12. Okumura, K., Zhao, M., Depinho, R. A., Furnari, F. B., and Cavenee, W. K. (2005) Proc. Natl. Acad. Sci. U. S. A. 102, 2703–2706 13. Liliental, J., Moon, S. Y., Lesche, R., Mamillapalli, R., Li, D., Zheng, Y., Sun, H., and Wu, H. (2000) Curr. Biol. 10, 401– 404 14. Li, G., Robinson, G. W., Lesche, R., Martinez-Diaz, H., Jiang, Z., Rozengurt, N., Wagner, K. U., Wu, D. C., Lane, T. F., Liu, X., Hennighausen, L., and Wu, H. (2002) Development 129, 4159 – 4170 15. Xiang, C. C., Kozhich, O. A., Chen, M., Inman, J. M., Phan, Q. N., Chen, Y., and Brownstein, M. J. (2002) Nat. Biotechnol. 20, 738 –742 16. Kohn, A. D., Summers, S. A., Birnbaum, M. J., and Roth, R. A. (1996) J. Biol. Chem. 271, 31372–31378 17. Kadomatsu, K., and Muramatsu, T. (2004) Cancer Lett. 204, 127–143 18. Muramatsu, T. (2002) J. Biochem. (Tokyo) 132, 359 –371 19. Deuel, T. F., Zhang, N., Yeh, H. J., Silos-Santiago, I., and Wang, Z. Y. (2002) Arch. Biochem. Biophys. 397, 162–171 20. Souttou, B., Ahmad, S., Riegel, A. T., and Wellstein, A. (1997) J. Biol. Chem. 272, 19588 –19593 21. Powers, C., Aigner, A., Stoica, G. E., McDonnell, K., and Wellstein, A. (2002) J. Biol. Chem. 277, 14153–14158 22. Bowden, E. T., Stoica, G. E., and Wellstein, A. (2002) J. Biol. Chem. 277, 35862–35868 23. Souttou, B., Raulais, D., and Vigny, M. (2001) J. Cell Physiol. 187, 59 – 64 24. Dupont, J., Renou, J. P., Shani, M., Hennighausen, L., and LeRoith, D. (2002) J. Clin. Investig. 110, 815– 825 25. Hong, T. M., Yang, P. C., Peck, K., Chen, J. J., Yang, S. C., Chen, Y. C., and Wu, C. W. (2000) Am. J. Respir. Cell Mol. Biol. 23, 355–363

26. Li, L., He, F., Litofsky, N. S., Recht, L. D., and Ross, A. H. (2003) Mol. Cell Neurosci. 24, 1051–1061 27. Matsushima-Nishiu, M., Unoki, M., Ono, K., Tsunoda, T., Minaguchi, T., Kuramoto, H., Nishida, M., Satoh, T., Tanaka, T., and Nakamura, Y. (2001) Cancer Res. 61, 3741–3749 28. Musatov, S., Roberts, J., Brooks, A. I., Pena, J., Betchen, S., Pfaff, D. W., and Kaplitt, M. G. (2004) Proc. Natl. Acad. Sci. U. S. A. 101, 3627–3631 29. Stolarov, J., Chang, K., Reiner, A., Rodgers, L., Hannon, G. J., Wigler, M. H., and Mittal, V. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 13043–13048 30. Wang, S., Gao, J., Lei, Q., Rozengurt, N., Pritchard, C., Jiao, J., Thomas, G. V., Li, G., Roy-Burman, P., Nelson, P. S., Liu, X., and Wu, H. (2003) Cancer Cell 4, 209 –221 31. Bandyopadhyay, S., Pai, S. K., Hirota, S., Hosobe, S., Tsukada, T., Miura, K., Takano, Y., Saito, K., Commes, T., Piquemal, D., Watabe, M., Gross, S., Wang, Y., Huggenvik, J., and Watabe, K. (2004) Cancer Res. 64, 7655–7660 32. Zindy, F., Eischen, C. M., Randle, D. H., Kamijo, T., Cleveland, J. L., Sherr, C. J., and Roussel, M. F. (1998) Genes Dev. 12, 2424 –2433 33. Suzuki, A., Itami, S., Ohishi, M., Hamada, K., Inoue, T., Komazawa, N., Senoo, H., Sasaki, T., Takeda, J., Manabe, M., Mak, T. W., and Nakano, T. (2003) Cancer Res. 63, 674 – 681 34. Chauhan, A. K., Li, Y. S., and Deuel, T. F. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 679 – 682 35. Bernard-Pierrot, I., Delbe, J., Caruelle, D., Barritault, D., Courty, J., and Milhiet, P. E. (2001) J. Biol. Chem. 276, 12228 –12234 36. Czubayko, F., Riegel, A. T., and Wellstein, A. (1994) J. Biol. Chem. 269, 21358 –21363 37. Czubayko, F., Schulte, A. M., Berchem, G. J., and Wellstein, A. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 14753–14758 38. Weber, D., Klomp, H. J., Czubayko, F., Wellstein, A., and Juhl, H. (2000) Cancer Res. 60, 5284 –5288 39. Souttou, B., Juhl, H., Hackenbruck, J., Rockseisen, M., Klomp, H. J., Raulais, D., Vigny, M., and Wellstein, A. (1998) J. Natl. Cancer Inst. 90, 1468 –1473 40. Klomp, H. J., Zernial, O., Flachmann, S., Wellstein, A., and Juhl, H. (2002) Clin. Cancer Res. 8, 823– 827 41. Aigner, A., Brachmann, P., Beyer, J., Jager, R., Raulais, D., Vigny, M., Neubauer, A., Heidenreich, A., Weinknecht, S., Czubayko, F., and Zugmaier, G. (2003) Ann. Oncol. 14, 1525–1529 42. Jager, R., List, B., Knabbe, C., Souttou, B., Raulais, D., Zeiler, T., Wellstein, A., Aigner, A., Neubauer, A., and Zugmaier, G. (2002) Br. J. Cancer 86, 858 – 863 43. Ulbricht, U., Brockmann, M. A., Aigner, A., Eckerich, C., Muller, S., Fillbrandt, R., Westphal, M., and Lamszus, K. (2003) J. Neuropathol. Exp. Neurol. 62, 1265–1275 44. Zhong, H., Chiles, K., Feldser, D., Laughner, E., Hanrahan, C., Georgescu, M. M., Simons, J. W., and Semenza, G. L. (2000) Cancer Res. 60, 1541–1545 45. Nakamura, N., Ramaswamy, S., Vazquez, F., Signoretti, S., Loda, M., and Sellers, W. R. (2000) Mol. Cell. Biol. 20, 8969 – 8982 46. Agarwal, A., Das, K., Lerner, N., Sathe, S., Cicek, M., Casey, G., and Sizemore, N. (2005) Oncogene 24, 1021–1031 47. Persad, S., Troussard, A. A., McPhee, T. R., Mulholland, D. J., and Dedhar, S. (2001) J. Cell Biol. 153, 1161–1174 48. Papadimitriou, E., Polykratis, A., Hatziapostolou, M., Parthymou, A., Polytarchou, C., and Mikelis, C. (2004) Curr. Cancer Drug Targets 4, 471– 482 49. Czubayko, F., Schulte, A. M., Missner, S. C., Hsieh, S. S., Colley, K. J., and Wellstein, A. (1995) Breast Cancer Res. Treat. 36, 157–168 50. Schulte, A. M., Lai, S., Kurtz, A., Czubayko, F., Riegel, A. T., and Wellstein, A. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 14759 –14764 51. Malerczyk, C., Schulte, A. M., Czubayko, F., Bellon, L., Macejak, D., Riegel, A. T., and Wellstein, A. (2005) Gene Ther. 12, 339 –346 52. Satyamoorthy, K., Oka, M., and Herlyn, M. (2000) Pigment Cell Res. 13, Suppl. 8, 87–93 53. Nagata, Y., Lan, K. H., Zhou, X., Tan, M., Esteva, F. J., Sahin, A. A., Klos, K. S., Li, P., Monia, B. P., Nguyen, N. T., Hortobagyi, G. N., Hung, M. C., and Yu, D. (2004) Cancer Cell 6, 117–127

10668 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 16 • APRIL 21, 2006 Downloaded from http://www.jbc.org/ at UMDNJ on September 11, 2013