throbber
[CANCER RESEARCH 60, 3504 –3513, July 1, 2000]
`
`A Direct Linkage between the Phosphoinositide 3-Kinase-AKT Signaling Pathway
`and the Mammalian Target of Rapamycin in Mitogen-stimulated and
`Transformed Cells1
`Aleksandar Sekulic´,2 Christine C. Hudson,2 James L. Homme, Peng Yin, Diane M. Otterness, Larry M. Karnitz, and
`Robert T. Abraham3
`Department of Immunology [A. S.], Mayo School of Medicine [J. L. H.], and Division of Oncology Research [L. M. K.], Mayo Clinic, Rochester, Minnesota 55905, and
`Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710 [C. C. H., P. Y., D. M. O., R. T. A.]
`
`ABSTRACT
`
`The microbially derived antiproliferative agent rapamycin inhibits cell
`growth by interfering with the signaling functions of the mammalian
`target of rapamycin (mTOR). In this study, we demonstrate that inter-
`leukin-3 stimulation induces a wortmannin-sensitive increase in mTOR
`kinase activity in a myeloid progenitor cell line. The involvement of
`phosphoinositide 3*-kinase (PI3K) in the regulation of mTOR activity was
`further suggested by findings that mTOR was phosphorylated in vitro and
`in vivo by the PI3K-regulated protein kinase, AKT/PKB. Although AKT
`phosphorylated mTOR at two COOH-terminal sites (Thr2446 and Ser2448)
`in vitro, Ser2448 was the major phosphorylation site in insulin-stimulated
`or -activated AKT-expressing human embryonic kidney cells. Transient
`transfection assays with mTOR mutants bearing Ala substitutions at
`Ser2448 and/or Thr2446 indicated that AKT-dependent mTOR phosphoryl-
`ation was not essential for either PHAS-I phosphorylation or p70S6K
`activation in HEK cells. However, a deletion of amino acids 2430 –2450 in
`mTOR, which includes the potential AKT phosphorylation sites, signifi-
`cantly increased both the basal protein kinase activity and in vivo signal-
`ing functions of mTOR. These results demonstrate that mTOR is a direct
`target of the PI3K-AKT signaling pathway in mitogen-stimulated cells,
`and that the identified AKT phosphorylation sites are nested within a
`“repressor domain” that negatively regulates the catalytic activity of
`mTOR. Furthermore, the activation status of the PI3K-AKT pathway in
`cancer cells may be an important determinant of cellular sensitivity to the
`cytostatic effect of rapamycin.
`
`INTRODUCTION
`
`Rapamycin is a potent immunosuppressive drug and investigational
`anticancer agent, the major mechanism of action of which involves the
`inhibition of lymphoid or tumor cell proliferation, through interfer-
`ence with an event(s) required for G1-to-S phase progression in
`cycling cells. The block to G1 phase progression imposed by rapa-
`mycin occurs prior to the “restriction point,” based on the observa-
`tions that rapamycin inhibits the phosphorylation of the retinoblas-
`toma protein and that rapamycin-treated cells are not fully committed
`to enter S-phase of the cell cycle after release from drug-induced G1
`arrest (1–3). The sensitivity of certain tumor cell lines to the cytostatic
`effects of rapamycin has prompted considerable interest in the possi-
`bility that this drug might be a useful cancer chemotherapeutic agent.
`Indeed, a rapamycin analogue (CCI-779; Wyeth-Ayerst) is now in
`
`Received 1/10/00; accepted 4/27/00.
`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 This work was supported by USPHS Grants CA 76193 and CA 52995 (to R. T. A.)
`from the National Cancer Institute, by a Collaborative Research Program in Cancer
`Research Grant (to R. T. A.) from Glaxo-Wellcome, and by the Mayo Foundation.
`C. C. H. is the recipient of postdoctoral fellowship PF-99-100 – 01-TBE from the Amer-
`ican Cancer Society. R. T. A. is a Glaxo-Wellcome Professor of Molecular Cancer
`Biology.
`2 These authors contributed equally to this work.
`3 To whom requests for reprints should be addressed, at Department of Pharmacology
`and Cancer Biology, Room C333B LSRC, Box 3813, Duke University Medical Center,
`Durham, NC 27710. Phone: (919) 613-8650; Fax: (919) 684-8461 E-mail: abrah008@
`mc.duke.edu.
`
`Phase I clinical trials in cancer patients in the United States and
`Europe.
`The molecular pharmacology underlying the cellular effects of
`rapamycin is now understood in considerable detail. The principal
`rapamycin “receptor” is a widely expressed intracellular protein
`termed FKBP4-12. In mammalian cells, the interaction of rapamycin
`with FKBP12 generates a pharmacologically active complex that
`binds with high affinity to the mTOR [Ref. 4; also named FRAP (5),
`RAFT1 (6), or RAPT1 (7) by others]. This rapamycin target protein is
`a member of a recently described family of protein kinases, termed
`PIKKs. The PIKK family members share a COOH-terminal catalytic
`domain that bears significant sequence homology to the lipid kinase
`domains of PI3Ks (8). Other members of the PIKK family include
`TOR1p and TOR2p, the budding yeast orthologues of mTOR. The
`finding that rapamycin interacts with FPR1p, the budding yeast or-
`thologue of FKBP12, to arrest yeast cell growth in G1 phase (9)
`suggests that the TOR signaling pathway has been at least partially
`conserved during eukaryotic evolution.
`The specificity of rapamycin as an inhibitor of mTOR function
`facilitated the identification of the downstream signaling events gov-
`erned by mTOR in mitogen-stimulated cells. To date, the rapamycin-
`sensitive signaling activities ascribed to mTOR impinge primarily on
`the translational machinery. Rapamycin treatment triggers the rapid
`dephosphorylation and inactivation of p70S6K in mitogen-stimulated
`cells (10 –14). The overall effect of p70S6K activation is to stimulate
`ribosome biogenesis, and, in turn, to increase the capacity of the
`translational machinery, which allows cells to meet the increased
`demand for protein synthesis imposed by cell cycle progression (15–
`17). Although p70S6K activation involves a complex series of phos-
`phorylation events catalyzed by multiple protein kinases (18 –21), the
`prompt reversal of p70S6K activation by rapamycin (11, 14) suggests
`that this protein kinase requires continuous signaling through mTOR
`to both achieve and maintain the activated state. The exact nature of
`the input supplied by mTOR remains unclear; however, recent find-
`ings suggest that mTOR phosphorylates and suppresses the activity of
`a type 2A protein phosphatase bound directly to p70S6K (22). Hence,
`rapamycin treatment may inactivate p70S6K by removing a mTOR-
`dependent inhibitory constraint on the activity of a p70S6K-targeted
`type 2A protein phosphatase PP2A.
`A second downstream protein targeted by mTOR is the transla-
`tional repressor, PHAS-I, also termed 4E-BP1. PHAS-I represses
`translation initiation through association with eIF-4E, the mRNA
`cap-binding subunit of the eIF-4F complex. The binding of PHAS-I to
`eIF-4E blocks assembly of the eIF-4F complex at the 59-cap structure
`of the mRNA template, thereby decreasing the efficiency of transla-
`tion initiation (23). Stimulation of cells with insulin or growth factors
`
`4 The abbreviations used are: FKBP, FK506-binding protein; mTOR, mammalian
`target of rapamycin; PI3K, phosphoinositide 39-kinase; PIKK, PI3K-related kinases;
`p70S6K, ribosomal p70 S6 kinase; eIF, eukaryotic initiation factor; FBS, fetal bovine
`serum; IL, interleukin; HEK, human embryonic kidney; HA, hemagglutinin; mAb, mono-
`clonal antibody; PMSF, phenylmethylsulfonyl fluoride; PDK, phosphoinositide-depen-
`dent kinase; PKC, protein kinase C.
`3504
`
`.
`
`Roxane Labs., Inc.
`Exhibit 1021
`Page 001
`
`

`
`mTOR REGULATION BY THE PI3K-AKT PATHWAY
`
`MATERIALS AND METHODS
`
`suggest that deregulated signaling through mTOR may contribute to
`the transformed phenotype of PTEN-deficient cancer cells.
`
`causes the phosphorylation of PHAS-I at five sites, which leads to the
`release of eIF-4E, and, in turn, an increase in eIF-4F-dependent
`translation initiation (24 –29). The phosphorylation of PHAS-I in-
`duced by hormonal stimuli is strongly inhibited by rapamycin (27, 30,
`31). Earlier results suggested that mTOR is directly responsible for the
`Plasmids, Reagents, and Antibodies. The expression vectors encoding
`phosphorylation of PHAS-I in intact cells (32, 33), although addi-
`AU1-tagged wild-type mTOR (AmTORwt) and catalytically inactive (“kinase-
`tional proline-directed kinases appear to be required for full phospho-
`dead”) mTOR (AmTORkd) were described previously (32). The rapamycin-
`rylation of PHAS-I in insulin-stimulated cells (34).
`resistant versions of mTOR contain an additional Ser (2035)3 Ile mutation
`The signaling pathway that couples growth factor receptor occu-
`and are usually designated with the suffix “SI” (e.g., AmTOR-SI). PCR-based
`pancy to mTOR activation is only partially understood. However,
`mutagenesis was used to construct single and double mTOR point mutants in
`accumulating evidence places mTOR downstream of both PI3K and
`which Thr2446 and Ser2448 were changed to alanine (A) residues. The internal
`the PI3K-regulated protein kinase, AKT (also termed PKB), in growth
`deletion mutant, AmTORD, which lacks amino acids 2430 –2450, was pre-
`factor-stimulated cells. This model is based in part on genetic and
`pared by the PCR-based SOEing technique (40). The expression vectors for
`HA-tagged wild-type AKT (cAKT), catalytically inactive AKT (AKT-kd) and
`pharmacological evidence that links activation of PI3K and/or AKT to
`the constitutively active myristylated form of AKT (myrAKT), were kind gifts
`the two intracellular events known to be governed by mTOR, the
`from P. N. Tsichlis (Fox-Chase Cancer Center, Philadelphia, PA). The cDNA
`activation of p70S6K and the phosphorylation of PHAS-I (35). The
`encoding p70S6K (kindly provided by Dr. Naohiro Terada, National Jewish
`notion that mTOR participates in signaling downstream from PI3K
`Medical and Research Center, Denver, CO) was appended with nucleotides
`may be particularly relevant to the antitumor activity of rapamycin.
`encoding an NH2-terminal FLAG epitope tag and was cloned into pcDNA3
`Recent studies have identified a negative regulator of PI3K-mediated
`using EcoRI and XbaI restriction sites. All PCR products were subcloned and
`signaling, PTEN, as a tumor suppressor gene product (36). The tumor
`then sequenced to ensure the fidelity of the amplification step.
`suppressor function of PTEN is attributed to its activity as a phos-
`Recombinant murine IL-3 was purchased from R&D Systems, Inc. (Min-
`phoinositide 3-phosphatase, which effectively terminates PI3K-
`neapolis, MN). Recombinant human insulin (Recombulin) and G418 (Geneti-
`mediated signaling via dephosphorylation of the second messengers,
`cin) were obtained from Life Technologies, Inc. (Gaithersburg, MD), and
`FuGene transfection reagent was purchased from Boehringer Mannheim (In-
`phosphatidylinositol-3,4,5-trisphosphate
`and phosphatidylinositol-
`dianapolis, IN). Wortmannin (Sigma) was dissolved in DMSO (Me2SO) to
`3,4-bisphosphate. Thus, loss of PTEN function leads to hyperactiva-
`yield a 1.2 mM stock solution. The wortmannin stock solution was aliquoted
`tion of the PI3K signaling cascade, which promotes abnormal cell
`and stored at 270°C. Rapamycin (Sigma) was prepared as a 10 mM stock
`growth, survival, and migration.
`solution in ethanol and aliquoted and stored as described above.
`The importance of PI3K signaling during tumorigenesis is under-
`The a-AU1 and 12CA5 (a-HA) mAbs were purchased from Babco (Rich-
`scored by observations that mutations in the PTEN gene occurs
`mond, CA), and the a-mTOR monoclonal antibody, 26E3, was a generous gift
`frequently in a variety of human cancers, including prostate cancer
`from Dr. Peter Houghton (St. Jude Children’s Research Hospital, Memphis,
`and glioblastoma (37). If mTOR also resides downstream of PI3K
`TN). Peptides corresponding to amino acid residues 2433–2450 in mTOR were
`and/or AKT, then mTOR activity should also be deregulated in
`synthesized (Research Genetics, Huntsville, AL) with or without phosphate at
`PTEN-deficient tumor cells, and consequently, PTEN status might be
`either or both of the underlined residues in the sequence CDTNAKGNKRSR-
`TRTDSYS. Polyclonal antibodies directed against the nonphosphorylated pep-
`an important predictor of cancer cell sensitivity to the mTOR inhib-
`tide were raised by immunizing rabbits with the peptide coupled to keyhole
`itor, rapamycin. Given these speculative arguments, it becomes in-
`limpet hemocyanin. The antiserum (designated a-mTOR 367) was affinity-
`creasingly important to define the interactions among PI3K, AKT, and
`purified over a peptide-coupled Sulfolink bead column according to the man-
`mTOR as the rapamycin analogue, CCI-779, moves into clinical trials
`ufacturer’s procedure (Pierce, Rockford, IL). Phosphospecific antibodies were
`in patients with different types of cancer.
`prepared in a similar fashion, except that a keyhole limpet hemocyanin-
`At the inception of this study, the most direct evidence for epistatic
`coupled, dually phosphorylated peptide (containing phosphate at both Thr2446
`relationships among PI3K, AKT, and mTOR stems from results
`and Ser2448 served as the immunogen. The resulting antiserum was first passed
`obtained with a polyclonal antibody, termed mTAb1, which recog-
`over a column consisting of nonphosphorylated peptide immobilized on Sul-
`nizes a COOH-terminal peptide sequence in mTOR (residues 2433–
`folink beads, and the flow-through fraction was then passed through a second
`2450; Ref. 38). The authors noted that cellular stimulation with
`column containing the immobilized, dually phosphorylated peptide. The bound
`antibodies (designated a-mTORp2) were eluted at low pH and were stored in
`insulin, or expression of mutationally activated AKT, caused a de-
`PBS containing 0.05% azide.
`crease in the immunoreactivity of mTOR in anti-mTAb1 immunoblot
`Cell Culture and Transfections. The murine IL-3-dependent myelomono-
`analyses (39). The loss of mTAb1 binding activity was reversed by
`cytic progenitor cell line, FDC-P1, was cultured in standard growth medium
`treatment of the immunoprecipitated mTOR with a protein phospha-
`[RPMI 1640 supplemented with 10% (v/v) FBS (Hyclone, Logan, UT), 2 mM
`tase prior to immunoblot analysis. Collectively, these results sug-
`L-glutamine, 50 mM 2-mercaptoethanol, 10 mM HEPES (pH 7.2), and 10%
`gested that insulin or AKT stimulation caused the phosphorylation of
`(v/v) WEHI-3 cell-conditioned medium as a source of IL-3]. Stably transfected
`mTOR at a site(s) that resulted in a decrease in the recognition of this
`FDC-P1 cells expressing AmTORwt were prepared by suspending 1 3 107
`protein by the mTAb1 antibody.
`exponentially growing cells in 350 ml of standard growth medium at 4°C. The
`The goal of the present study was to further understand the role of
`cells were mixed with a total of 45 mg of plasmid DNA suspended in the same
`the PI3K-AKT signaling pathway in the regulation of mTOR function
`medium. Mock transfectants received 45 mg of pcDNA3 only, whereas mTOR
`transfectants were electroporated with 25 mg of mTOR-encoding plasmid plus
`by extracellular stimuli. We demonstrate that stimulation of myeloid
`20 mg of pcDNA3 as filler. Prior to electroporation, the cell-DNA mixtures
`progenitor cells with IL-3 triggers a rapid increase in the protein
`were incubated for 10 min at room temperature. The cells were electroporated
`kinase activity of mTOR. The IL-3-dependent increase in mTOR
`with a BTX model T820 square-wave electroporator (San Diego, CA) at a
`activity is blocked by low concentrations of the PI3K inhibitor,
`setting of 350 V (10-ms pulse duration). The electroporated cells were mixed
`wortmannin. Furthermore, we provide in vitro and in vivo evidence
`gently and then allowed to stand at room temperature for an additional 10 min.
`that AKT phosphorylates mTOR at a site(s) located in a region that
`The cells were then diluted into 20 ml of standard growth medium and cultured
`represses the catalytic activity of the mTOR kinase domain. Deletion
`for 24 h, at which time the cells were transferred into fresh growth medium
`of this repressor domain generates a mTOR mutant bearing a consti-
`containing 800 mg of G418/ml. Stable clones that expressed AmTORwt and
`tutively elevated level of protein kinase activity. These findings out-
`AmTORkd were isolated by limiting dilution, and expression levels of the
`line a direct linkage between the PI3K-AKT pathway and mTOR and
`transfected proteins were assessed by immunoblotting with the AU1 mAb.
`3505
`
`Roxane Labs., Inc.
`Exhibit 1021
`Page 002
`
`

`
`mTOR REGULATION BY THE PI3K-AKT PATHWAY
`
`transfected cells were transferred into serum-free DMEM and cultured for
`24 h. Cellular extracts were prepared by removal of the culture medium,
`followed by addition of 400 ml of buffer P per dish [50 mM Tris-HCl, 100 mM
`NaCl, 50 mM b-glycerophosphate (pH 7.4), containing 10% (w/v) glycerol, 1%
`Triton X-100, 1 mM DTT, 50 nM microcystin, 1 mM PMSF, and protease
`inhibitor cocktail]. The detached cells were disrupted by sonication, and
`cleared extracts from the HA-tagged AKT-expressing cells and the AmTORwt-
`expressing cells were mixed at a total protein ratio of 1:9. The epitope-tagged
`mTOR and AKT proteins were coimmunoprecipitated with 1 ml of AU1 mAb
`and 5 mg of 12CA5 mAb bound to protein A-Sepharose beads that had been
`precoupled to rabbit antimouse immunoglobulin antibodies. The immunopre-
`cipitates were washed three times in buffer N [25 mM HEPES (pH 7.6), 0.5 M
`NaCl, 10% glycerol, 1 mM Na3VO4, and 0.2% Tween 20] and two times in
`kinase buffer F [50 mM Tris (pH 7.5), 10 mM MgCl2, 50 mM Na3VO4, and 1
`mM DTT]. The coimmunoprecipitated proteins were incubated for 50 min at
`30°C in 20 ml of kinase buffer F supplemented with 10 mM ATP and 20 mCi
`of [g-32P]ATP (specific activity, 4500 Ci/mmol). The reaction products were
`separated by SDS-PAGE and transferred to an Immobilon-P membrane. The
`incorporation of 32P into wild-type or mutated forms of AmTOR was detected
`by autoradiography and quantitated by phosphorimager analysis as described
`above.
`To measure the activity of transiently transfected FLAG-p70S6K, serum-
`deprived HEK 293 cells were prepared as described above. The cells were
`stimulated with 1 mM insulin and then lysed in TNEE buffer [50 mM Tris-HCl,
`150 mM NaCl, 2.5 mM EDTA, 2 mM EGTA, 25 mM b-glycerophosphate, 25
`mM NaF (pH 7.5), containing, 0.5% Triton X-100, 100 mM sodium orthovana-
`date, 2 mM DTT, and protease inhibitor cocktail]. The epitope-tagged p70S6K
`was immunoprecipitated from cellular extracts with anti-FLAG M2 affinity
`resin (Sigma Chemical Co., St. Louis, MO), and protein kinase activity
`was determined with a p70S6K assay kit (Upstate Biotechnology, Inc., Lake
`Placid, NY).
`
`For experiments, exponentially growing FDC-P1 cells (2 3 107 cells/
`sample were washed twice in PBS. The cells were resuspended in 20 ml of
`starvation medium [RPMI 1640 containing 100 mg/ml BSA, 2 mM L-gluta-
`mine, and 50 mM 2-mercaptoethanol, buffered to pH 7.2 with 10 mM HEPES].
`After 4 – 6 h in culture, the factor-deprived cells were treated for 30 min with
`the indicated pharmacological inhibitors and then were restimulated with either
`30 ng/ml IL-3 or 20% FBS.
`HEK 293 and 293T cells were maintained in monolayer cultures in DMEM
`(Life Technologies, Inc.) supplemented with 10% FBS or 5% FBS, respec-
`tively. Prior to transfection, 6 3 105 cells were seeded into a 60-mm tissue
`culture dish. The cells were cultured for 24 h under standard conditions and
`then were transfected with a total of 5 mg of plasmid DNA mixed with 8 ml of
`FuGene transfection reagent/dish. The standard amounts of plasmid DNAs
`used for each transfection were: AKT, 0.25 mg; mTOR, 3 mg; and p70S6K, 2
`mg. When necessary, the total amount of plasmid DNA was brought to 5 mg
`by addition of the empty pcDNA3 expression vector. The transfected cells
`were cultured for 16 h, washed one time in PBS, and arrested for 24 h in
`serum-free DMEM. The serum-deprived cells were pretreated for 30 min with
`wortmannin or rapamycin and then stimulated with insulin for the indicated
`times. The procedures for transfection of HEK 293T cells were similar, except
`that the cells were deprived of serum for 2 h prior to drug treatment.
`DU 145 and PC-3 prostate cancer cells were maintained in monolayer
`cultures in RPMI 1640 supplemented with 10% FBS. Prior to assay, 2 3 105
`cells were seeded in 60-mm tissue culture dishes. After 24 h, the cells were
`transferred into serum-free RPMI 1640 and were cultured for an additional
`20 h. The cells were washed in PBS and lysed in LB buffer [25 mM Tris-HCl,
`pH 7.4, 50 mM NaCl, 10% (w/v) glycerol, 1% Triton X-100, 50 mM b-glyc-
`erophosphate, 20 nM microcystin- LR, 100 mg/ml PMSF, and protease inhib-
`itor cocktail (5 mg/ml aprotinin, 5 mg/ml pepstatin, and 10 mg/ml leupeptin)].
`The lysates were cleared of insoluble material, and the cleared extracts were
`assayed for total protein to equalize sample loading prior to SDS-PAGE.
`Immunoprecipitations. Mock-transfected or AmTOR-transfected FDC-P1
`cells (2 3 107 cells/sample) were growth factor deprived and restimulated as
`described above. The cells were washed in PBS and lysed by sonication in 1
`Stimulation of mTOR Catalytic Activity by Serum or IL-3. Our
`ml of buffer L [50 mM Tris-HCl, 50 mM b-glycerophosphate, 100 mM NaCl
`initial objective was to determine whether mTOR activity was regu-
`(pH 7.4), containing 10% glycerol, 0.2% Tween 20, 1 mM DTT, 1 mM
`Na3VO4, 1 mM MgCl2, 50 nM microcystin-LR, 1 mM PMSF, and protease
`lated in a PI3K-dependent fashion by IL-3, a cytokine that promotes
`inhibitor cocktail]. The lysates were cleared of insoluble material by centrif-
`the proliferation and survival of myeloid lineage progenitor cells. To
`ugation, and the extracts were immunoprecipitated with 1 ml of a-AU1 mAb
`facilitate the analyses of mTOR kinase activity, we stably expressed
`for 2 h at4°C. The immunoprecipitates were washed three times in buffer W
`AU1-tagged wild-type or catalytically inactive (“kinase-dead”) ver-
`[50 mM Tris-HCl, 50 mM b-glycerophosphate, 100 mM NaCl (pH 7.4), con-
`sions of mTOR (AmTORwt and AmTORkd, respectively) in IL-3-
`taining 10% glycerol, 0.2% Tween 20, and 1 mM DTT] and twice in buffer K
`dependent FDC-P1 cells. Importantly, the stable cell lines selected for
`[10 mM HEPES, 50 mM NaCl, 50 mM b-glycerophosphate (pH 7.4), 50 nM
`these studies were not overexpressing the recombinant protein, as
`microcystin-LR, and the protease inhibitor cocktail].
`indicated by immunoblot analyses of the transfected clones for total
`Immunoblot Analyses. For immunoblot analyses with a-mTOR 367 or
`mTOR protein levels with antibodies that recognize both the endog-
`a-mTORp2 antibodies, recombinant AmTOR was immunoprecipitated with
`the tag-specific a-AU1 mAb from transfected FDC-P1, HEK 293, or HEK
`enous and transfected proteins (results not shown). The tagged Am-
`293T cells. The immunoprecipitated proteins were separated by SDS-PAGE
`TOR proteins therefore serve as a “tracer” subpopulation, the behavior
`through 6% polyacrylamide gels. After transfer to Immobilon-P, the mem-
`of which in response to physiological stimuli should reflect that of the
`branes were blocked and probed with 5 mg per ml affinity-purified antibodies
`endogenous mTOR.
`in Tris-buffered saline-0.2% Tween 20 (TBST) containing 2% (w/v) BSA (for
`In the initial studies, AmTORwt-expressing cells were deprived of
`a-mTORp2 antibodies) or 5% milk (for a-mTOR 367 antibodies). Immuno-
`serum and IL-3 for 6 h and then were restimulated for 10 min with
`reactive proteins were detected with horseradish peroxidase-conjugated to
`IL-3 prior to the preparation of cellular extracts. The extracts were
`protein A and the Renaissance reagent (New England Nuclear, Boston, MA).
`immunoprecipitated with a-AU1 mAb, and mTOR kinase activities
`The blots were then stripped and reprobed with the a-AU1 mAb in TBST-milk
`were determined with PHAS-I as the substrate. Parallel samples were
`solution. The phosphorylation state of endogenous mTOR in DU 145 or PC-3
`prepared from identically treated cells that expressed the AmTORkd
`prostate cancer cells was analyzed by immunoblotting with a-mTORp2 as
`mutant. Stimulation of AmTORwt-expressing FDC-P1 cells with IL-3
`described above, followed by reblotting of the same membrane with the
`a-mTOR mAb, 26E3, in TBST-milk solution.
`significantly increased the in vitro kinase activity of the immunopre-
`Kinase Assays. The protein kinase activity of immunoprecipitated mTOR
`cipitated AmTORwt but did not change the amount of AmTORwt in
`was assayed with recombinant PHAS-I as the substrate (Stratagene, La Jolla,
`these immunoprecipitates (Fig. 1A, left panel). The activation of
`CA; Ref. 32). The samples were separated by SDS-PAGE, and radiolabeled
`mTOR by IL-3 was maximal at 5–10 min after cytokine stimulation
`PHAS-I was detected by autoradiography. Incorporation of 32P into PHAS-I
`and then dropped to a lower, but still elevated, plateau level of activity
`was quantitated with a Molecular Dynamics Storm 840 Phosphorimager
`that was sustained for at least 4 h after cytokine addition (Fig. 1B and
`(Sunnyvale, CA) and ImageQuant software.
`data not shown). In contrast, AU1 immunoprecipitates from either
`Phosphorylation of mTOR by AKT in vitro was performed by transfection
`mock-transfected or AmTORkd-expressing cells contained low levels
`of AmTORwt and HA-tagged myrAKT, c-AKT, or catalytically inactive AKT
`of background protein kinase activity that was not substantially in-
`into separate populations of HEK 293 cells. The cells were plated in 60-mm
`creased by cellular stimulation with IL-3. Interestingly, serum-starved
`culture dishes and were transfected as described above. After 16 h, the
`3506
`
`RESULTS
`
`Roxane Labs., Inc.
`Exhibit 1021
`Page 003
`
`

`
`mTOR REGULATION BY THE PI3K-AKT PATHWAY
`
`noreactivity of AU1-tagged mTOR isolated from IL-3-stimulated
`FDC-P1 cells (Fig. 3, upper panel). Interestingly, the time course of
`the alteration in anti-mTOR antibody reactivity corresponded closely
`to the changes in mTOR kinase activity induced by IL-3 (Fig. 1B).
`The IL-3-dependent decrease in mTOR immunoreactivity was abro-
`gated by pretreatment of the cells with 100 nM wortmannin, suggest-
`ing that this alteration was mediated through the activation of PI3K.
`As will be described below, parallel immunoblot analyses with a
`phospho-mTOR-specific antibody (a-mTORp2) indicated that
`the
`decrease in a-mTOR 367 reactivity induced by IL-3 stimulation is
`attributable to the phosphorylation of at least one amino acid (Ser2448)
`located within the a-mTOR 367 epitope (Fig. 3, middle panel; see
`below for description).
`Examination of the peptide sequence recognized by a-mTOR 367
`antibodies revealed that this region contained two consensus phos-
`phorylation sites (Thr2446 and Ser2448) for AKT (Fig. 4). To determine
`whether mTOR was an in vitro substrate for AKT, we expressed
`AmTORwt, HA-tagged wild-type AKT (cAKT), activated AKT
`(myrAKT), or a catalytically inactive AKT (AKTkd) in different
`populations of HEK 293 cells. Cellular extracts were then mixed, and
`
`Fig. 1. Stimulation of mTOR kinase activity by IL-3 or serum. FDC-P1 cells were
`stably transfected with wild-type AU1-tagged mTOR or a catalytically inactive version of
`AmTOR. Transfected clones were cultured for 6 h inmedium without serum and IL-3. A,
`left panel, factor-deprived FDC-P1 cells were stimulated for 10 min with IL-3 or medium
`only (2). The protein kinase activities of wild-type (wt) AmTOR or the “kinase-dead”
`(kd) AmTOR were determined in immune complex kinase assays with [g-32P]ATP and
`recombinant PHAS-I as the substrate (lower panels). Incorporation of 32Pi into PHAS-I
`was quantitated with a Molecular Dynamics Storm 840 phosphorimaging system and
`reported as cpm. The amounts of immunoprecipitated mTOR were assessed by immuno-
`blotting with AU1 mAb (upper panels). Right panel, AmTORwt-expressing FDC-P1 cells
`were restimulated for 10 min with IL-3 or serum, and AmTORwt kinase activities were
`measured as above. B, mTORwt-expressing FDC-P1 cells were restimulated for the
`indicated times with IL-3. The protein kinase activities in AmTORwt immunoprecipitates
`were determined as described in A. Incorporation of 32Pi into PHAS-I was normalized to
`the basal level of phosphorylation in the immunoprecipitate prepared from unstimulated
`cells.
`
`Fig. 2. Inhibition of IL-3 dependent mTOR activation by cellular treatment with
`wortmannin. AmTORwt-transfected FDC-P1 cells were deprived of growth factors as
`described in Fig. 1. AmTORwt-expressing cells were treated for 30 min with the indicated
`concentrations of wortmannin (Wm). After stimulation of the cells for 10 min with IL-3,
`AmTORwt was immunoprecipitated, and immune complex kinase assays were performed
`as described in the Fig. 1 legend. The amount of AmTORwt in each immunoprecipitate
`was determined by a-AU1 immunoblotting (upper panel).
`
`FDC-P1 cells also displayed a clear increase in AmTORwt activity
`after a 10-min exposure to fresh serum (Fig. 1A, right panel). Thus,
`ligation of receptors for IL-3, as well as undefined serum components
`(possibly insulin-like growth factors), initiates a signaling pathway
`leading to mTOR activation in FDC-P1 cells.
`Role of PI3K in IL-3-dependent mTOR Activation. Earlier stud-
`ies implicated the PI3K pathway in the activation of mTOR-depend-
`ent signaling events in HEK 293 cells and 3T3-L1 preadipocytes (35,
`39). Stimulation of the IL-3 receptor also triggers a rapid increase in
`PI3K activity (41), which suggested that PI3K might be responsible
`for the activation of mTOR in IL-3-stimulated FDC-P1 cells. If the
`activation of mTOR by IL-3 is dependent on PI3K, then this response
`should be inhibited by pretreatment of the cells with wortmannin at
`drug concentrations #100 nM (42). As shown in Fig. 2, the activation
`of AmTORwt by IL-3 was virtually abrogated by pretreatment of the
`FDC-P1 cells with 10 nM wortmannin. Thus, the sensitivity of IL-3-
`dependent mTOR activation to wortmannin strongly suggests that this
`response is dependent on the activation of PI3K.
`Direct Phosphorylation of mTOR by the PI3K-regulated Ki-
`Fig. 3. IL-3-dependent alterations in the reactivity of AmTORwt with R domain-
`nase, AKT. An earlier report provided evidence that activation of the
`directed a-mTOR antibodies. AmTORwt-expressing FDC-P1 cells were deprived of
`growth factors and then stimulated for the indicated times with medium only (2) or with
`PI3K-AKT pathway led to the phosphorylation of the mTAb1 anti-
`IL-3. Wortmannin (100 nM) was added to the indicated sample 30 min prior to IL-3
`body epitope located in the COOH-terminal region of mTOR (39).
`stimulation. The cells were lysed, and AmTORwt was immunoprecipitated from cleared
`extracts with a-AU1 mAb. The control lane (Co) represents an a-AU1 immunoprecipitate
`Using independently derived polyclonal antibodies (a-mTOR 367)
`from mock-transfected (empty plasmid only) FDC-P1 cells. The immunoprecipitates were
`specific for the same region of mTOR (amino acid residues 2433–
`resolved by SDS-PAGE and sequentially immunoblotted with a-mTOR 367 antibodies,
`2450), we observed a similar time-dependent decrease in the immu-
`phosphospecific a-mTORp2 antibodies (see Figs. 4 and 5 for details), and a-AU1 mAb.
`3507
`
`Roxane Labs., Inc.
`Exhibit 1021
`Page 004
`
`

`
`mTOR REGULATION BY THE PI3K-AKT PATHWAY
`
`Fig. 4. Schematic diagram of mTOR functional domains and mutants used in this
`study. The NH2-terminal AU1 epitope tag is followed by an extended NH2-terminal
`region of unknown function. The FKBP12zrapamycin binding (FRB) domain is labeled, as
`is the Ser (2035)3 Ile mutation used to generate the rapamycin-resistant AmTOR-SI
`mutants. The FRB domain is followed by the catalytic region (CAT). Finally, the putative
`“repressor” (R) domain (residues 2430 –2450) is shown together with the newly identified
`AKT phosphorylation sites at Thr2446 and Ser2448.
`
`observed that IL-3 stimulation caused a prompt increase in the reac-
`tivity of mTOR with these phosphospecific antibodies (Fig. 3, middle
`panel). Notably, the time-dependent increase in a-mTORp2 immuno-
`reactivity mirrored precisely the decrease in a-mTOR 367 binding
`provoked by IL-3 stimulation (Fig. 3, upper panel). Pretreatment of
`these cells with 100 nM wortmannin blocked the increase in
`a-mTORp2 binding stimulated by IL-3, indicating that this alteration
`was dependent on the activation of PI3K.
`Transient transfection studies in HEK 293 cells revealed that
`insulin stimulation also provoked a rapid increase in the
`a-mTORp2 reactivity of the rapamycin-resistant AmTOR-SI mutant
`(Fig. 6A). AmTOR-SI contains a single amino acid substitution [Ser
`(2035)3 Ile] that renders the hor

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket