Mtor Review

download Mtor Review

of 9

Transcript of Mtor Review

  • 8/2/2019 Mtor Review

    1/9

    Review

    The mTOR pathway and its role in human genetic diseases

    Margit Rosner, Michaela Hanneder, Nicol Siegel, Alessandro Valli,Christiane Fuchs, Markus Hengstschlager *

    Medical Genetics, Obstetrics and Gynecology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090 Vienna, Austria

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285

    1.1. The mTOR pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285

    1.2. Localization of mTOR pathway components . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

    2. The mTOR pathway and specific human genetic diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

    2.1. Tuberous sclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

    2.2. Peutz-Jeghers syndrome. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287

    2.3. Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Proteus syndrome, Lhermitte-Duclos disease. . . . . . . . . . . . . . . . . . . . . . 287

    2.4. von Hippel-Lindau disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287

    2.5. Neurofibromatosis type 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    2.6. Polycystic kidney disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    3. The mTOR pathway in Alzheimers syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    Mutation Research 659 (2008) 284292

    A R T I C L E I N F O

    Article history:

    Received 15 May 2008Received in revised form 29 May 2008

    Accepted 3 June 2008

    Available online 11 June 2008

    Keywords:

    PI3K

    Akt

    TSC

    mTOR

    Ras

    Human genetic disease

    Cancer

    A B S T R A C T

    The signalling components upstream and downstream of the protein kinase mammalian target of

    rapamycin (mTOR) are frequently altered in a wide variety of human diseases. Upstream of mTOR keysignalling molecules arethe small GTPaseRas, the lipid kinasePI3K, theAkt kinase, andthe GTPaseRheb,

    whichare knownto be deregulated in many humancancers. Mutations in themTOR pathway component

    genes TSC1, TSC2, LKB1, PTEN, VHL, NF1 and PKD1 trigger the development of the syndromes tuberous

    sclerosis, Peutz-Jeghers syndrome, Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Lhermitte-

    Duclos disease, Proteus syndrome, von Hippel-Lindau disease, Neurofibromatosis type 1, and Polycystic

    kidney disease, respectively. In addition, the tuberous sclerosis proteins have been implicated in the

    development of several sporadic tumors and in the control of the cyclin-dependent kinase inhibitor p27,

    known to be of relevance for several cancers. Recently, it has been recognized that mTOR is regulated by

    TNF-a andWnt, both of whichhave been shownto play critical roles in thedevelopmentof many human

    neoplasias. In addition to all these human diseases, the role of mTOR in Alzheimers disease, cardiac

    hypertrophy, obesity and type 2 diabetes is discussed.

    2008 Elsevier B.V. All rights reserved.

    * Corresponding author. Tel.: +43 1 40400 7847; fax: +43 1 4040 7848.

    E-mail address: [email protected] (M. Hengstschlager).

    Abbreviations: 4EBP1, eukaryotic initiationfactor 4E binding protein-1; ADPKD, autosomal dominant polycystickidney disease;AMPK, 50AMP-activated protein kinase; Cdk,

    cyclin-dependent kinase; EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; FGFR, fibroblasts growth factor receptor; FKBP, FK506-binding

    protein;GAP, GTPase activating protein;GSK3b, glycogen synthasekinase 3b; HIF,hypoxia-inducibletranscriptionfactor; IGF-1R, insulin-like growth factor 1 receptor; IGF-

    1, insulin-like growth factor 1; IKK, inhibitor of kB kinase; IRS1, insulin receptor substrate 1; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of

    rapamycin; NF1, neurofibromatosis type 1; p70S6K, ribosomal p70S6 kinase; PC1, polycystin-1; PDGFb, plateled-derived growth factor b; PDGFR, platelet-derived growth

    factor receptor; PDK1, phosphoinositide-dependent kinase-1; PI3K, phosphatidylinositol 3-kinase; PIP2, lipid phosphatidylinositol-4,5-biphosphate; PIP3, phosphatidy-

    linositol-3,4,5-triphosphate; PML, promyelocytic leukaemia; PRAS40, proline-rich AKT substrate 40 kDa; protor, protein observed with rictor; PTEN, phosphatase and tensin

    homolog; raptor, regulatory associated protein of mTOR; Rheb, Ras homolog enriched in brain; rictor, rapamycin-insensitive companion of mTOR; RTK, receptor tyrosine

    kinase; siRNA, small interfering RNA; sin1, stress-activated protein kinase-interacting protein; TGF-a, and transforming growth factor a; TNF-a, tumor necrosis factor-a;TSC, tuberous sclerosis complex; TSC1, tuberous sclerosis complex gene 1; TSC2, tuberous sclerosis complex gene 2; VEGF, vascular endothelial growth factor; VEGFR,

    vascular endothelial growth factor receptor; VHL, von Hippel-Lindau disease.

    Contents lists available at ScienceDirect

    Mutation Research/Reviews in Mutation Research

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / r e v i e w s m rC o m m u n i t y a d d r e s s : w w w . e l s e v i e r . c o m / l o c a t e / m u t r e s

    1383-5742/$ see front matter 2008 Elsevier B.V. All rights reserved.

    doi:10.1016/j.mrrev.2008.06.001

    mailto:[email protected]://www.sciencedirect.com/science/journal/13835742http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://www.sciencedirect.com/science/journal/13835742mailto:[email protected]
  • 8/2/2019 Mtor Review

    2/9

    4. The mTOR pathway and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    4.1. Upstream and downstream of mTOR in tumor development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    4.2. Ras, mTOR and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    4.3. TNF-a activates mTOR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    4.4. Wnt regulates mTOR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    4.5. mTOR and renal cell carcinoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    4.6. The TSC proteins in sporadic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    4.7. The TSC proteins and p27 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

    5. The mTOR pathway and cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2906. The mTOR pathway in obesity and type 2 diabetes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

    7. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

    Acknowledgements. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

    References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

    1. Introduction

    1.1. The mTOR pathway

    The mammalian target of rapamycin (mTOR) is a member of the

    phosphoinositide-3-kinase-related kinase family, which is centrally

    involved in growth regulation, proliferation control and cancer cell

    metabolism. In mammalian cells, two structurally and functionallydistinct mTOR-containingcomplexes have been identified. mTORC1

    contains raptor (regulatory associated protein of mTOR), mLST8

    (also known as GbL) and PRAS40 (proline-rich Akt substrate40 kDa). Whereas the functionof mLST8 is not really clarified, raptor

    regulates mTORC1 functioning as a scaffold for recruiting TORC1

    substrates. PRAS40 is phosphorylated by Akt at T246 releasing its

    inhibitory effects on mTORC1. The major substrates of mTORC1

    known so far are 4EBP1 (eukaryotic initiation factor 4E binding

    protein-1) and p70S6K (ribosomal p70S6 kinase), both regulators of

    protein translation. mTORC1 phosphorylates and activates p70S6K

    at T389 to activate the ribosomal protein S6 via phosphorylation at

    S240/244 (Fig. 1) [15].

    mTORC2 also contains the mLST8 protein, but instead of raptor,

    mTORC2 contains rictor (rapamycin-insensitive companion ofmTOR) and sin1 (stress-activated protein kinase-interacting pro-

    tein) proteins. Rictor and sin1appearto stabilize eachother through

    binding, building the structural foundation for mTORC2. The

    interaction between rictor and mTOR is not blocked by the drug

    rapamycin nor affected by nutrient levels, which are conditions

    known to regulate mTORC1. mTORC2 additionally contains protor

    (protein observed with rictor), which lacks any obvious functional

    domains. The role of protorfor mTORC2 activity(if any)still remains

    elusive. mTORC2 phosphorylates the oncogenic kinase Akt (also

    known as protein kinase B) at S473, what in conjunction with the

    PDK1 (phosphoinositide-dependent kinase-1)-mediated phosphor-

    ylation of Akt at T308 drives full activationof Akt(Fig.1) [15]. Over

    100 putative Akt substrates have been reported, although many of

    those have not been characterized in more detail [6].One important substrate of Akt has been shown to function

    upstream of mTORC1: the TSC1/TSC2 protein complex. The TSC1

    gene on chromosome 9q34 encodes hamartin, and TSC2 on

    chromosome 16p13.3 encodes tuberin [7,8]. Mutations in either

    the TSC1 or TSC2 gene causetuberous sclerosis (TSC), a multisystem

    autosomal dominant disorder (see below) [9,10]. Activated

    receptor tyrosine kinases activate the phosphatidylinositol-3-

    kinase (PI3K) through phosphorylation of adaptors, such as the

    insulin receptor substrate 1 (IRS1). Phosphorylation of the

    membrane lipid phosphatidylinositol-4,5-biphosphate (PIP2) by

    PI3K produces the second messenger phosphatidylinositol-3,4,5-

    triphosphate (PIP3) in a reaction that can be reversed by the

    phosphatase PTEN (phosphatase and tensin homolog) (Figs. 1 and

    2). PDK1 and Akt bind to PIP3 at the plasma membrane, and PDK1

    phosphorylates Akt at T308. Akt-mediated phosphorylation on

    S939 and T1462 downregulates tuberins GTPase activating (GAP)

    potential toward Rheb (Ras homolog enriched in brain), which is a

    potent regulator of mTOR. It was recently shown that Rheb

    regulates mTOR through FKBP38, a member of the FK506-binding

    protein (FKBP) family. FKBP38 binds to mTOR and inhibits its

    activity and Rheb interacts directly with FKBP38 and prevents its

    association with mTOR (Fig. 1) [1,2,4,11].p70S6K has been demonstrated to phosphorylate IRS1 on

    multiple inhibitory sites promoting its degradation (Fig. 1) [12].

    This finding is one possible explanation for the observation that

    loss of TSC1/2 function results in downregulated Akt phosphor-

    ylation [13,14]. It has been suggested that tumors in TSC patient

    are less aggressive because of this S6K-dependent negative

    feedback inhibition. Furthermore, it has recently been reported

    that Rheb has a negative effect on mTORC2. This study provides

    evidence that Rheb may affect mTORC2 indirectly probably

    through this S6K-dependent negative feedback loop [15]. In

    addition, recently it was demonstrated that the TSC1TSC2

    Fig. 1. mTORC1 and mTORC2 in the insuling signalling pathway. For details see the

    text.

    M. Rosner et al./ Mutation Research 659 (2008) 284292 285

    http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001http://dx.doi.org/10.1016/j.mrrev.2008.06.001
  • 8/2/2019 Mtor Review

    3/9

    complex can physically associate with mTORC2 but not mTORC1,and that the TSC protein complex positively regulates mTORC2in a

    manner independent of its GTPase-activating protein activity

    toward Rheb (Fig. 1) [16].

    1.2. Localization of mTOR pathway components

    IRS1, PI3K, PDK1 and Akt function at the plasma membrane. In

    agreement with its role in the regulation of translation, all the

    other components of the described pathway including TSC1/2,

    Rheb, mTOR andp70S6K have also been localizedto thecytoplasm.

    Besides regulation of translation,mTORhas also been implicated in

    the regulation of ribosome biogenesis, macro-autophagy or

    transcription [17]. Accordingly, it is not surprising that the

    proteins involved in the PI3K signalling pathway are also localizedwithin thenucleus. PI3K has been shown to be nuclear [18] PDK1 is

    a nucleo-cytoplasmic shuttling protein [19]. Akt translocates also

    to the nucleus [2023] and it was reported that IGF-1 (insulin-like

    growth factor1) stimulates phosphorylationof AktT308 andof Akt

    S473 in the cytoplasm and in the nucleus [24]. It has even recently

    been shown that the PML (promyelocytic leukaemia) tumor

    suppressor prevents cancer by inactivating Akt in the nucleus [25].

    Several recent reports have brought conclusive evidence that the

    tumor suppressor PTEN, once considered as a strictly cytoplasmic

    protein, also shuttles to the nuclear compartment, where it joins

    the components PI3K, PDK1 and Akt [26,27]. Tuberin has been

    reported to be cytoplasmic and nuclear [2831]. Furthermore,

    mTOR and its substrate p70S6K have been found to be localized to

    both the cytoplasm and the nucleus, and cytoplasmic nuclearshuttling of mTOR has been shown to be involved in rapamycin-

    sensitive signalling and translation initiation [3236]. The p70S6K

    target S6 is dispersed throughout the cytoplasm. Within the

    nucleus S6 protein is concentrated to the nucleoli and almost

    absent from the nucleoplasm, what is a consequence of the fact

    that eukaryotic ribosomes are assembled in the nucleolus before

    export to the cytoplasm [37,38].

    2. The mTOR pathway and specific human genetic diseases

    2.1. Tuberous sclerosis

    Hamartin and tuberin are believed to function in the same

    complex, because they associate physically in vivo with high

    affinity to form heterodimers [39,40]. Until now, over 50 proteinshave been demonstrated to interact with hamartin and/or tuberin

    anda wide variety of functionshave been describedfor this protein

    complex [41]. However, in addition, evidence for functional

    differences of these two proteins comes from knockout studies

    and from microarray and proteomicanalyses [4246]. As described

    above (Fig. 1), a major function of the hamartin/tuberin (TSC1/

    TSC2) protein complex is to antagonize the mTOR pathway. Akt-

    mediated phosphorylation on S939 and T1462 downregulates

    tuberins GTPase activating (GAP) potential toward Rheb (Figs. 1

    and 2). In summary, Akt stimulates mTOR signalling by inhibiting

    the function of tuberin [4752]. Via this pathway mTOR controls

    the translation rate of RNAs specifically regulated via the

    translation initiation factor eIF4E. Cap-dependent translation is

    facilitated by mTOR phosphorylation and inactivation of 4E-BPs,which are suppressors of eIF4E. In addition, mTOR regulates the

    translation rates of specific messages (mainly from genes involved

    in ribosome biogenesis) via its effects on the phosphorylation of

    the 40S ribosomal protein S6 by S6K. S6K is considered to be

    involved in translation control of a small subset of mRNAs that

    contain a 50-terminal oligopyrimidine tract [1,2,53].

    The majority of TSC disease-causing mutations occur de novo in

    either TSC1 or TSC2. Linkage analysis suggests that about half of

    large families are linked to TSC1 and half to TSC2, whereas in the

    sporadic TSC population mutations in TSC2 are about five times

    more common than mutations in TSC1. Themutation spectra of the

    TSC genes are very heterogenous and no hotspots of mutations

    have been found. No significant genotypephenotype correlations

    have been established. Patients with TSC2 mutations seem to bemore severely affected than patients with mutations in the TSC1

    gene, which appears to be due to a higher rate of second hit events.

    TSC patients carry a mutant TSC1 or TSC2 gene in each of their

    somatic cells and loss of heterozygosity has been documented in a

    wide variety of TSC tumors. Tumor development is assumed to be

    the result of somatic second hit mutations according to

    Knudsons tumor suppressor model. Accordingly, although TSC

    is a disease, which is transmitted in an autosomal dominant

    fashion, mutations in the TSC genes are believed to be recessive at

    the level of the affected cell [710,53]. TSC affects about 1 in 6000

    live births and is characterized by the development of tumor-like

    growths, named hamartomas, in the kidneys, heart, skin and brain.

    Primary diagnostic criteria for TSC include facial angiofibromas,

    peringual fibromas, calcified retinal hamartomas, cortical tubers or

    Fig.2. Regulatory components upstreamof theTSC1/TSC2 complexThe phosphorylation sites on tuberinor hamartinare shown under thecorresponding enzymes.For details

    see the text.

    M. Rosner et al./ Mutation Research 659 (2008) 284292286

  • 8/2/2019 Mtor Review

    4/9

    renal angiomyolipomas. In addition, nearly all patients exhibit skin

    signs, such as, e.g. hypomelanotic macules, forehead fibrous

    plaques, facial angiofibromas or shagreen patches. In summary, the

    major features of TSC include dermatologic manifestations, renalangiomyolipomas, and neurologic manifestations, such as epi-

    lepsy, mental retardation, and autism. The severity of TSC and its

    impact on the quality of life is extremely variable. Many patients

    have minimal signs and symptoms with no neurologic disability.

    The greatest source of morbidity is the brain tumors, named

    cortical tubers, causing seizures in 8090% of affected individuals,

    and behavioural abnormalities (mostly autism) in over half of

    affected individuals [9,10,53].

    Lymphangiomyomatosis (also known as lymphangioleiomyo-

    matosis) is characterized by widespread pulmonary proliferation

    of abnormal smooth-muscle cells and cystic changes within the

    lung parenchyma manifested by dyspnea or pneumothorax. It

    affects women almost exclusively. Among women with TSC the

    incidence for lymphangiomyomatosis is 2639% with many ofthese women being asymptomatic [9,53] (Table 1).

    The mTORC1 inhibitor sirolimus has already been studied in

    clinical trials analysing its effects in TSC and lymphangiomyoma-

    tosis therapy justifying the need for larger, well-powered trials to

    test whether sirolimus therapy shouldbecome thestandardof care

    for such patients [54,55].

    2.2. Peutz-Jeghers syndrome

    In the recent past, in addition to Akt other enzymes have been

    identified to regulate tuberins functions. The LKB1 tumor

    suppressor gene is responsible for the hamartomatous Peutz-

    Jeghers syndrome and encodes a serine/threonine kinase, which

    phosphorylates and activates AMPK (50

    AMP-activated proteinkinase). Under energy starvation conditions phosphorylation by

    AMPK activates tuberin via phosphorylationat T1227 andS1345, is

    required for the regulation of cell size control, and protects cells

    from energy deprivation-induced apoptosis [56,57]. Recently, it

    was demonstrated that Wnt inhibits the GSK3b (glycogensynthase kinase 3b)-mediated phosphorylation of tuberin. GSK3inhibits the mTOR pathway via phosphorylating tuberin at S1337

    and at S1341. This phosphorylation depends on AMPK-priming

    phosphorylation of tuberin at S1345 and triggers activation of

    tuberins potential to inhibit mTOR. Accordingly, Wnt inhibits the

    GSK3-mediated activation of tuberin to block mTOR [58] (Fig. 2).

    The Peutz-Jeghers syndrome, caused by mutation in the LKB1

    tumor suppressor gene, is characterized by hamartomas primarily

    in the intestine, where they grow as polyps, by abnormal

    mucocutaneous pigmentation and an increased risk of malignant

    tumors in the intestine and elsewhere [59,60].

    2.3. Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome,

    Proteus syndrome, Lhermitte-Duclos disease

    The tumor suppressor gene PTEN encodes a phosphatase that

    catalyzes the conversion of PIP3 to PIP2. Loss of PTEN increases Akt

    activity, which downregulates tuberins function (Fig. 2). Germline

    PTEN mutations trigger a wide variety of different clinical

    syndromes, such as Cowden syndrome, Bannayan-Riley-Ruvalcaba

    syndrome, Proteus syndrome, and Lhermitte-Duclos disease. Since

    these four diseases are all autosomal dominant hamartoma

    syndromes it has been suggested that these syndromes should

    be grouped together as PTEN-hamartoma tumor syndromes

    [59,60]. As described above, LKB1, associated with the hamarto-

    matous Peutz-Jeghers syndrome,activates tuberin via AMPK. Since

    PTEN, LKB1 and tuberin have been demonstrated to be involved inthe regulation of HIF and VEGF, it has been speculated that

    increasedHIF andVEGF levels may be a commonfeature of familial

    hamartoma syndromes. It was reported that tuberin regulates the

    expression of the vascular endothelial growth factor (VEGF)

    through mTOR-dependent and -independent mechanisms. Loss

    of functional tuberin triggers the accumulation of the hypoxia-

    inducible transcription factor (HIF) and upregulation of the

    expression of HIF-responsive genes including VEGF. TSC2-negative

    cells, in contrast to normal cells, fail to downregulate HIF in

    response to growth factor deprivation. Expression of a disease-

    causing mutation of TSC2 fails to normalize HIF in TSC2-deficient

    cells, suggesting that HIF regulation plays a role in TSC2 tumor

    suppressor function (Table 1; Figs. 2 and 3) [5962].

    2.4. von Hippel-Lindau disease

    Mutations in the VHL tumor suppressor gene cause von Hippel-

    Lindau disease (VHL). This genetic multisystem disorder is

    characterized by the abnormal growth of tumors (angiomas) in

    certain parts of the body. Hemangioblastomas (tumors of the

    central nervous system) can develop in the brain, the retina of the

    eyes, and other areas of the nervous system. Other types of tumors

    develop in the adrenal glands, the kidneys, or the pancreas.

    Patients can have headaches, problems with balance and walking,

    dizziness, weakness of the limbs, vision problems, and high blood

    pressure. Individuals with VHL also harbor increased risks for

    certain types of cancer, especially renal carcinomas. VHL encodes a

    protein, which is part of a multiprotein complex involved in

    Table 1

    mTOR pathway components in specific human genetic diseases

    Gene Protein: function Disease: clinical characteristics

    TSC1 Hamartin: binding partner for tuber in Tub erou s scleros is: hamartomas , epileps y, mental retardatio n

    Lymphangiomyomatosis: lung cysts, dyspnea, pneumothorax

    TSC2 Tuberin: Rheb GTPase activation, regulation of mTORC2, p27, etc. Tuberous sclerosis: hamartomas, epilepsy, mental retardation

    Lymphangiomyomatosis: lung cysts, dyspnea, pneumothorax

    LKB1 LKB1: ser/thr kinase Peutz-Jeghers syndrome: hamartomas in the intestine

    PTEN PTEN: phosphatase Cowden syndrome: hamartomas in multiple organs

    Bannayan-Riley-Ruvalcaba syndrome: hamartomas in multiple organs

    Lhermitte-Duclos disease: hamartomas in brain

    Proteus syndrome: hamartomas in multiple organs

    VHL VHL: ubiquitination of HIF von Hippel-Lindau disease: angiomas, hemangioblastomas, renal carci nomas

    NF1 Neu rofi bro min: Ra s-GTPa se activa ting Neur ofib romatos is type 1: neu rofi bromas , h amar toma s

    PKD1 Po lycys tin- 1: interactio n, b lock o f mTOR Polycystic kidney diseas e: cys ts in b oth kidn eys

    M. Rosner et al./ Mutation Research 659 (2008) 284292 287

  • 8/2/2019 Mtor Review

    5/9

    ubiquitination and degradation of the transcription factor HIF

    leading to the dysregulation of a variety of genes, involved in

    growth control. As described above, mTOR is a key upstreamregulator of HIF. However, the interactive roles of mTOR and VHL

    for the control of HIF activity is still under investigation (Fig. 3;

    Table 1) [59,60,63,64].

    2.5. Neurofibromatosis type 1

    The familial cancer syndrome neurofibromatosis type 1 (NF1)

    affects about 1 in 3500 individuals. It is characterized by the

    development of benign (neurofibromas) and malignant peripheral

    nerve sheath tumors. Patients can exhibit cognitive deficits, bone

    deformations and hamartomatous lesions of the iris. Neurofibro-

    matosis type 1 is often grouped clinically with TSC as a common

    neurocutaneous syndrome. It is caused by mutations in NF1. NF1

    encodes neurofibromin, which functions as a Ras-GTPase-activat-ing protein. Accordingly, Ras is aberrantly activated in NF1-

    deficient tumors. Rashas many functionsin thecell, oneof which is

    to activate the PI3K-TSC1/TSC2-mTOR cascade. It has been

    demonstrated that in NF1-deficient cells mTOR is constitutively

    activated. This activation depends on Ras, PI3K and TSC2.

    Furthermore, it has been shown that activation of mTOR is

    essential for in vivo NF1-associated tumorigenesis (Fig. 2; Table 1)

    [65,66].

    2.6. Polycystic kidney disease

    Renal cysts are the hallmark of the autosomal dominant

    polycystic kidney disease (ADPKD) and are also characteristic for

    TSC. Mutations in the PKD1 gene, which encodes polycystin-1(PC1), account foralmost 90%of ADPKD. ADPKDis considered to be

    one of the most common monogenic inherited diseases. The

    growth of thousands of cysts in both kidneys in a progressive

    manner during adulthood results in replacement of normal renal

    tissue and significant overall growth of the organs. A synergistic

    role of tuberin and PC1 is suggested by the fact that patients with

    mutations in both, TSC2 and PKD1, have earlier-onset and more

    severe polycystic kidney disease than patients harboring only

    PKD1 mutations. It has been demonstrated that tuberin is required

    for the localization of PC1 to the plasma membrane [67].

    Furthermore, the cytoplasmic tail of PC1 can interact, directly or

    indirectly, with tuberin and mTOR. This interaction would appear

    to normally result in inhibition of mTOR activity. Disruption of PC1

    triggers high levels of mTOR activity (Fig. 3) [68]. In addition,

    another protein known to be involved in the etiology of polycystic

    kidney disease was found to interact with tuberin. Mutations in

    NEK1 cause the formation of kidney cysts in mice. Performing a

    yeast two-hybrid screen NEK1 has been identified as interacting

    partner of tuberin. It will be of great interest for the future to

    further investigate the role of this interaction for the formation of

    renal cysts (Table 1) [69]. Recent results demonstrate that PC1

    regulates mTOR and is involved in mechanotransduction by

    primary cilia measuring the degree of luminal fluid flow. It is

    currently under discussion whether a critical function of PC1 and

    primary cilia in the kidney may be to sense renal injury by

    detecting changes in luminal flow [70].

    3. The mTOR pathway in Alzheimers syndrome

    Hamartin has been demonstrated to interact with neurofila-

    ment-light chain and hamartin and tuberin co-localize with

    neurofilament-light chain preferentially in the proximal to central

    growth cone region of cultured cortical neurons suggesting that

    hamartin could function as an integrator of the neuronal

    intermediate filament and the actin cytoskeletal network [71].

    In TSC the central nervous system lesions, such as cortical tubers,

    result in a variety of neurological manifestations, including mentalretardation and seizures. Furthermore, tuberin has been reported

    to be involved in the regulation of neuronal differentiation [72].

    Alzheimers disease, the most common form of dementia in the

    elderly, is a progressive neurodegenerative disorder characterized

    by global cognitive decline involving memory, orientation,

    judgment, and reasoning. Upon autopsy abundant amounts of

    the typical lesions, extracellular deposits of b-amyloid andintracellular deposits of neurofibrillary tangles, are necessary for

    a confirmed diagnosis of Alzheimers disease. There is a very high

    incidence of Alzheimer-type neuropathology that is observed in

    the brains of middle-aged patients with Down syndrome (Trisomy

    21). A genetic link between these two diseases has been suggested

    since it was shown that the b-amyloid precursor protein gene

    mapped to chromosome 21. Beside the b-amyloid precursorprotein gene recentgenetic studies have identifiedalso other genes

    associated with inherited risk for Alzheimers disease, such as

    presenilin 1, presenilin 2, and apolipoprotein E. However,

    Alzheimers disease is a multifactorial illness with both genetic

    and non-genetic causes [73]. Tuberin levels have been found to be

    decreased in samples of brains positive for b-amyloid plaques and

    neurofibrillary tangles of patients with Alzheimers disease or with

    Down syndrome [74]. Deregulation of PTEN and Akt [75,76],

    alteration of mTOR activity [77,78] and deregulated p70S6K

    activity [79] has also already been found to be associated with

    Alzheimers disease pathology.

    4. The mTOR pathway and cancer

    4.1. Upstream and downstream of mTOR in tumor development

    The signalling components upstream and downstream of mTOR

    are frequently altered in a wide variety of human tumors. As

    described above, mutations in the tumor suppressor genes TSC1,

    TSC2, LKB1, PTEN, VHL, NF1 and PKD1 trigger the development of the

    hamartoma syndromes Tuberous sclerosis, Peutz-Jeghers syn-

    drome, Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome,

    Lhermitte-Duclos disease, Proteus syndrome, von Hippel-Lindau

    disease, Neurofibromatosis type 1, and Polycystic kidney disease,

    respectively (Table 1). PI3K itself can function as protooncogene,

    since increased PI3K activity has been shown to induce cell

    transformation and progression and has been found in many

    human cancers, such as, e.g. ovarian and gastrointestinal cancer.

    Fig. 3. The mTOR pathway downstream of the TSC1/TSC2 complex. For details see

    the text.

    M. Rosner et al./ Mutation Research 659 (2008) 284292288

  • 8/2/2019 Mtor Review

    6/9

    Mutations in PI3K upstream elements, such as epidermal growth

    factor, ErbB2 or insulin-like growth factor 1 receptor trigger

    increased PI3K activity in various cancer cell types [1,3,17,80]. The

    protooncogene Akt is amplified in different tumors, such as, e.g.

    breast and ovarian cancer [1,6,17]. Rheb expression is upregulated

    in many human cancers [81] and p70S6K often is amplified or

    overexpressed in breast cancers correlating with poor prognosis

    [82].

    4.2. Ras, mTOR and cancer

    Growth factors activate receptor tyrosine kinases (RTKs),

    including epidermal growth factor receptor (EGFR), platelet-

    derived growth factor receptor (PDGFR), fibroblasts growth factor

    receptor (FGFR), insulin-like growth factor 1 receptor (IGF-1R),

    interleukin receptors, vascular endothelial growth factor receptor

    (VEGFR), interferon receptors, and integrin receptors, which then

    activate two key signalling molecules. As described above, one is

    the lipid kinase PI3K and the other one is the small GTPase Ras

    (Fig. 2). In fact, most human tumors harbor activating mutations in

    K-ras, H-ras, N-ras, the p110a PI3K subunit or in RTKs [17,83].Tuberin is also phosphorylated by ERK at S664 (Fig. 2). Mitogenic

    stimuli or oncogenic Ras via Raf activate the mitogen-activatedprotein kinase (MAPK)/Erk pathway leading to phosphorylation of

    tuberin and inactivation of the TSC protein complex to regulate

    p70S6K [84]. Furthermore, the MAPK-activated kinase RSK1

    interacts with tuberin and phosphorylates it at S1798 triggering

    inactivation of tuberin and increased mTOR signalling to p70S6K

    (Fig. 2) [85]. Very recent data provide evidence that the function of

    Ras to trigger inactivation of tuberin via the MAPK/Erk pathway

    plays a major role in theregulation of cell survival upon mutational

    activation of the oncogene Ras [86,87].

    4.3. TNF-a activates mTOR

    Dysregulation of the tumor necrosis factor-a (TNF-a) signalling

    pathway contributes to the development of many human cancersdue to enhanced IKK activity and constitutive activation of the

    transcription factor NF-kB. Very recently, this pathway of

    transcription control has also been demonstrated to affect the

    regulation of translation. TNF-a activates mTOR through phos-phorylation and inactivation of hamartin by the kinase IKKb(Fig. 2) [88].

    4.4. Wnt regulates mTOR

    Components of the mTOR pathway may also be therapeutic

    targets for diseases linked to hyperactive Wnt signalling. The Wnt

    protein family regulates a wide range of cellular functions

    including cell growth, proliferation, polarity, differentiation and

    development. Deregulation of the Wnt/b-catenin pathway is acommon feature in many humanneoplasms [89,90]. Inthe absence

    of the secreted factor Wnt, GSK3 phosphorylates b-catenintriggering ubiquitination and degradation of b-catenin. Wntstimulation leads to activation of the protein Dishevelled (Dsh)

    to inhibit the ability of GSK3 to phosphorylate b-catenin. Anothercomponent of this degradation complex is the scaffold protein

    Axin. Both, hamartin and tuberin, have been reported to co-

    immunoprecipitate with GSK3 and Axin [91,92]. As already

    described above, Wnt inhibits the GSK3b-mediated phosphoryla-tion of tuberin. GSK3 inhibits the mTOR pathway via phosphor-

    ylating tuberin, what depends on AMPK-priming phosphorylation

    of tuberin and triggers activation of tuberins potential to inhibit

    mTOR. Accordingly, Wnt inhibits the GSK3-mediated activation of

    tuberin to block mTOR (Fig. 2) [58].

    4.5. mTOR and renal cell carcinoma

    In about 60% of renal cell carcinomas the VHL gene is

    inactivated [63,64]. As already described, inactivation of VHL

    triggers increased activity of the transcription factor HIF leading to

    overexpression of vascular endothelial growth factor (VEGF),

    plateled-derived growth factor b (PDGFb) and transforminggrowth factor a (TGF-a). A role of mTOR for renal cell carcinoma

    development has been suggested because mutations in TSC1 or

    TSC2 lead to activation of mTOR, confer a predisposition to renal-

    cell carcinoma and are accompanied by upregulated HIF activity.

    mTOR is a key upstream regulator of HIF (Fig. 3). Recent data

    suggest that inhibition of mTOR results in clinical benefit in renal

    cell carcinoma patients with poor prognostic features [5964,93].

    4.6. The TSC proteins in sporadic cancer

    The finding that the TSC genes are involved in regulation of the

    mTOR signalling network already initiated clinical trials for the

    treatment with rapamycin, a negative regulator of mTOR, of renal

    tumors and lung cysts found in tuberous sclerosis [15,17].

    Approaches like that have clear implications for the devastating

    disease, tuberous sclerosis, but beyond that to sufferers fromcancers that may also involve the TSC proteins, such as, e.g.

    sporadic bladder cancer, ovarian and gall bladder carcinoma, non-

    small-cell carcinoma of the lung and breast cancer [94]. Mutations

    in TSC1 have been found to be associated with the development of

    human bladder carcinoma [9597]. Evidence has been provided

    that reduced expression of tuberin might be involved in the

    progression of human pancreatic cancer [98]. It was also reported

    that tuberin and hamartin are aberrantly expressed and linked to

    clinical outcome in human breast cancer. The TSC1 gene promoter

    was seen to be methylated in human breast tumor tissues [99].

    Human xanthoastrocytomas consistently show low TSC1 transcript

    levels [100]. And recently, it was demonstrated that inactivation of

    tuberin via loss of expression or phosphorylation occurs frequently

    in endometrial carcinoma [101].

    4.7. The TSC proteins and p27

    A major regulator of mammalian cell cycle control is the cyclin-

    dependent kinase (Cdk) inhibitor p27. The transition from G0/G1

    to S phase is regulated via Cdk4 and Cdk6 activated by D-type

    cyclins andvia Cdk2 activatedby cyclin E andA. Dueto itspotential

    to control Cdk activity p27 is a very potent regulator of this

    transition [102,103]. Another important cell cycle regulator is

    tuberin. Downregulation of endogenous tuberin expression

    induces quiescent fibroblasts to enter the cell cycle and TSC2-

    negative cells exhibit a shortened G1 phase. Overexpression of

    hamartin or tuberin negatively regulates cell cycle progression

    accompanied by an increase of G1 cells and of p27 protein levels.Tuberin negatively regulates the activity of Cdk2 [104107]. p27 is

    known to be regulated on the level of protein stability and via

    subcellular localization. Binding of tuberin to p27 inhibits p27

    degradation by sequestering p27 from the Skp2-containing E3

    ubiquitin ligase [108]. Tuberin also induces nuclear p27 localiza-

    tion by inhibiting its 14-3-3-mediated cytoplasmic retention [109]

    (Fig. 3). In bladder tumors TSCgene mutations are correlated with

    reduced p27 expression [9597]. Cytoplasmic localization of p27

    was found in breast cancer and it was demonstrated that tuberin

    andhamartin expression is downregulated in breastcancer. Due to

    this connection between tuberin and p27, one could assume that

    under certain circumstances p27 or Cdk2 could also be considered

    as targets in addition to mTOR for therapeutics of hamartoma

    diseases and other tumor diseases [99,110,111].

    M. Rosner et al./ Mutation Research 659 (2008) 284292 289

  • 8/2/2019 Mtor Review

    7/9

    5. The mTOR pathway and cardiac hypertrophy

    Under energy starvation conditions tuberin is phosphorylated

    and thereby activated by AMPK [56,57]. Dependent on AMPK-

    priming phosphorylation of tuberin, GSK3b phosphorylatestuberin and triggers activation of its potential to inhibit mTOR

    [58] (Fig. 2). AMPKg2, an important regulatory subunit of AMPK, isencoded by the gene PRKAG2. Mutations in PRKAG2 are responsible

    for familial cardiac hypertrophy and Wolff-Parkinson-White

    syndrome, a distinctive ventricular electrophysiologic abnormality

    [112,113]. The knowledge that an important hallmark of cardiac

    hypertrophy (a major risk for cardiac morbidity and mortality) is

    hyperactivation of the PI3K-mTOR cascade, initiated a discussion

    whether inhibition of the mTOR pathway could be beneficial for

    therapies of these diseases [17,60,114,115].

    6. The mTOR pathway in obesity and type 2 diabetes

    p70S6K has been demonstrated to phosphorylate IRS1 on

    multiple inhibitory sites promoting its degradation (Fig. 1) [12].

    Inhibition of IRS protein function is one means by which cells

    become desensitized to insulin. Phosphorylation of IRS1, which is

    known to antagonize IRSsignalling, is elevated in animal models ofobesity and in muscle from type 2 diabetic patients. Recent studies

    have shown that insulin resistance can be regulated by mTORC1

    activation of p70S6K through the negative feedback loop. The

    inability to respond to insulin (insulin resistance) is a hallmark of

    both, obesity and type 2 diabetes [4,12,17,60,114,115].

    7. Summary

    The mTOR signalling pathway is activatedduring a wide variety

    of cellular responses, which includes T-lymphocyte activation,

    neurogenesis, muscle regeneration, insulin signalling and tumor

    formation. This has provoked intense interest in the TSC/mTOR

    cascade from virtually all major therapeutic areas related to many

    human diseases. mTOR plays an important role in Alzheimersdisease, cardiac hypertrophy, obesity and type 2 diabetes. mTOR is

    regulated by TNF-a and Wnt, both of which have been shown to

    play critical roles in many human cancers. The TSC proteins have

    been implicated in the development of several sporadic tumors

    and in the control of p27, known to be of relevance for several

    cancers. The mTOR upstream regulators Ras, PI3K, Akt and Rheb

    are known to be deregulated in a wide variety of human tumors.

    Tuberous sclerosis, Peutz-Jeghers syndrome, Cowden syndrome,

    Bannayan-Riley-Ruvalcaba syndrome, Lhermitte-Duclos disease,

    Proteus syndrome, von Hippel-Lindau disease, Neurofibromatosis

    type 1, and Polycystic kidney disease are caused by mutations in

    the mTOR pathway component genes TSC1, TSC2, LKB1, PTEN, VHL,

    NF1 and PKD1, respectively. Very recent data suggest that

    inhibition of mTOR results in clinical benefit in renal cell carcinomapatients with poor prognostic features and in the treatment of

    renal tumors and lung cysts found in tuberous sclerosis. It is the

    hope of investigators and patients alike that a more detailed

    understanding of the mTOR signalling cascade will lead to new

    therapies for many different human diseases.

    Acknowledgements

    Research in our laboratory is supported by the FWF Austrian

    Science Fund (P18894-B12), by the Marie Curie Research Network

    of the European Community (FP6 036097-2) and by the

    Herzfeldersche Familienstiftung. We apologize to those colleagues

    whose work is not cited due to space limitations or our inability to

    draw connections between elements of the primary literature.

    Conflict of interest

    None.

    References

    [1] D.A. Guertin, D.M. Sabatini, Defining the role of mTOR in cancer, Cancer Cell 12(2007) 922.

    [2] Q. Yang, K.-L. Guan, Expanding mTOR signalling, Cell Res. 17 (2007) 666681.[3] G.G. Chiang, R.T. Abraham, Targeting the mTOR signalling network in cancer,

    Trends Mol. Med. 13 (2007) 433442.[4] S.G. Dann, A. Selvaraj, G. Thomas, mTOR complex 1-S6K1 signaling: at the

    crossroadsof obesity, diabetes and cancer, Trends Mol.Med. 13 (2007) 252259.[5] P.T. Bhaskar, N. Hay, The two TORCs and Akt, Dev. Cell 12 (2007) 487502.[6] B.D. Manning,L.C. Cantley, AKT/PKB signalling: navigatingdownstream, Cell129

    (2007) 12611274.[7] The European Chromosome 16 Tuberous Sclerosis Consortium, Identification

    and characterization of the tuberous sclerosis gene on chromosome 16, Cell 75(1993) 13051315.

    [8] The TSC1 Consortium, Identification of the tuberous sclerosis gene TSC1 onchromosome 9q34, Science 277 (1997) 805808.

    [9] D.J.Kwiatkowski, Tuberous sclerosis:from tubers to mTOR,Ann. Hum.Genet. 67(2003) 8796.

    [10] A. Astrinidis, E.P. Henske, Tuberous sclerosis complex: linking growth and energysignaling pathways with human disease, Oncogene 24 (2005) 74757481.

    [11] X. Bai, D. Ma, A. Liu, X. Shen, Q.J. Wang, Y. Liu, Y. Jiang, Rheb activates mTOR by

    antagonizing its endogenous inhibitor, FKBP38, Science 318 (2007) 977980.[12] Y. Zick, Ser/thr phosphorylation of IRS proteins: a molecular basis for insulinresistance, Sci. STKE 25 (2005) pe4.

    [13] A. Jaeschke, J. Hartkamp, M. Saitoh, W. Roworth, T. Nobukuni, A. Hodges, A.J.Sampson, G. Thomas, R. Lamb, Tuberous sclerosis complex tumor suppressor-mediated S6 kinase inhibition by phosphatidylinositide-3-OH kinase is mTORindependent, J. Cell. Biol. 159 (2002) 217224.

    [14] D.J. Kwiatkowski, H. Zhang, J.L. Bandura, K.M. Heilberger, M. Glogauer, N. el-Hashemite, H. Onda, A mouse model of TSC1 reveals sex-dependent lethalityfromliver hemangiomas, andup-regulationof p70S6kinaseactivity in TSC1nullcells, Hum. Mol. Genet. 11 (2002) 525534.

    [15] Q.Yang,K. Inoki,E. Kim,K.-L.Guan, TSC1/TSC2andRhebhavedifferent effects onTORC1 and TORC2 activity, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 68116816.

    [16] J. Huang, C.C. Dibble, M. Matsuzaki, B.D. Manning, The TSC1TSC2 complex isrequired for proper activation of mTOR complex 2, Mol. Cell. Biol. (2008),doi:10.1128/MCB.00289-08 .

    [17] S. Wullschleger, R. Loewith, M. Hall, TOR signaling in growth and metabolism,Cell 124 (2006) 471484.

    [18] L.M. Neri, P. Borgatti, S. Capitani, A.M. Martelli, The nuclear phosphoinositide 3-

    kinase/AKT pathway: a new second messenger system, Biochim. Biophys. Acta1584 (2002) 7380.

    [19] C.K. Kikani, L.Q. Dong, F. Liu, New-clear functions of PDK1: beyond a masterkinase in the cytosol? J. Cell. Biochem. 96 (2005) 11571162.

    [20] Y. Pekarsky, A. Koval, C. Hallas, R. Bichi, M. Tresini, S. Malstrom, G. Russo, P.Tsichlis, C.M. Croce, Tcl1 enhances Akt kinase activity and mediates its nucleartranslocation, Proc. Natl. Acad. Sci. U.S.A. 97 (1999) 30283033.

    [21] P. Borgatti, A.M. Martelli, A. Bellacosa, R. Casto, L. Massari, S. Capitani, L.M. Neri,Translocation of Akt/PKB to the nucleus of osteoblast-like MC3T3-E1 cellsexposed to proliferative growth factors, FEBS Lett. 477 (2000) 2732.

    [22] Y. Tsujita, J. Muraski, I. Shiraishi, T. Kato, J. Kajstura, P. Anversa, M.A. Sussman,Nuclear targeting of Akt antagonizesaspects of cardiomoycte hypertrophy, Proc.Natl. Acad. Sci. U.S.A. 103 (2006) 1194611951.

    [23] J.-Y.Ahn,X. Liu, Z. Lju, L. Pereira,D. Cheng,J. Peng,P.A. Wade, A.W. Hamburger, K.Ye, Nuclear Akt associates with PKC-phosphorylated Ebp1, preventing DNAfragmentation by inhibition of caspase-activated DNase, EMBO J. 25 (2006)20832095.

    [24] R. Wang, M.G. Brattain, AKT can be activated in the nucleus, Cell. Signal. 18(2006) 17221731.

    [25] L.C. Trotman, A. Alimonti, P.P. Scaglioni, J.A. Koutcher, C. Cordon-Cardo, P.P.Pandolfi, Identification of a tumour suppressor network opposing nuclear Aktfunction, Nature 441 (2006) 523527.

    [26] Z. Lian, A. Di Christofano, Class reunion: PTEN joins the nuclear crew, Oncogene24 (2005) 73947400.

    [27] L.C. Trotman, X. Wang, A. Alimonti, Z. Chen, J. Teruya-Feldstein, H. Yang, N.P.Pavletich, B.S. Carver, C. Cordon-Cardo, H. Erdjument-Bromage, P. Tempst, S.G.Chi, H.J. Kim, T. Mistell, X. Jiang, P.P. Pandolfi, Ubiquitination regulates PTENnuclear import and tumor suppression, Cell 128 (2007) 141156.

    [28] R.Wienecke,J.C. Maize, F. Shoarinejad, W.C. Vass, J.Reed, J.S.Bonifacino, J.H.Resau,J. de Gunzburg, R.S. Yeung, J.E. DeClue,Co-localization of the TSC2 product tuberinwith its target Rap1 in the Golgi apparatus, Oncogene 13 (1996) 913923.

    [29] M. Nellist, M.A. van Slegtenhorst, M. Goedbloed, A.M. van den Ouweland, D.J.Halley, P. Van der Sluijs, Characterization of the cytosolic tuberin-hamartincomplex. Tuberin is a cytosolic chaperonefor hamartin,J. Biol. Chem. 274(1999)3564735652.

    [30] D.Lou, N.Griffith,D.J.Noonan,The tuberoussclerosis 2 gene product canlocalizeto nuclei in a phosphorylation-dependent manner, Mol. Cell. Biol. Res. Commun.

    4 (2001) 374380.

    M. Rosner et al./ Mutation Research 659 (2008) 284292290

    http://dx.doi.org/10.1128/MCB.00289-08http://dx.doi.org/10.1128/MCB.00289-08http://dx.doi.org/10.1128/MCB.00289-08http://dx.doi.org/10.1128/MCB.00289-08
  • 8/2/2019 Mtor Review

    8/9

    [31] M. Rosner, A. Freilinger, M. Hengstschlager, Akt regulates nuclear/cytoplasmiclocalization of tuberin, Oncogene 26 (2007) 521531.

    [32] S.-J. Kim, C.R. Kahn, Insulin stimulates p70SKinase in the nucleus of cells,Biochem. Biophys. Res. Commun. 234 (1997) 681685.

    [33] J.E.Kim, J. Chen, Cytoplasmic-nuclearshuttling of FKBP12-rapamycin-associatedprotein is involved in rapamycin-sensitive signaling and translation initiation,Proc. Natl. Acad. Sci. U.S.A. 97 (2000) 1434014345.

    [34] X. Zhang, L. Shu, H. Hosoi, K.G. Murit, P.J. Houghton, Predominant nuclearlocalization of mammalian target of rapamycin in normal and malignant cellsin culture, J. Biol. Chem. 277 (2002) 2812728134.

    [35] R.A. Bachmann, J.-H. Kim, A.-L. Wu, I.-H. Park, J. Chen, A nuclear transport signalin mammalian target of rapamycin is critical for its cytoplasmic signaling to S6kinase 1, J. Biol. Chem. 281 (2006) 73577363.

    [36] F. Furuya, J.A. Hanover, S.-Y. Cheng, Activation of phosphatidylinositol 3-kinasesignalling by a mutant thyroid hormone b receptor, Proc. Natl. Acad. Sci. U.S.A.103 (2006) 17801785.

    [37] I. Ruvinsky, O. Meyuhas, Ribosomal protein S6 phosphorylation: from proteinsynthesis to cell size, Trends Biochem. Sci. 31 (2006) 342346.

    [38] T. Kruger, H. Zentgraf, U. Scheer, Intranucleolar sites of ribosome biogenesisdefined by the localization of early binding ribosomal proteins, J. Cell Biol. 177(2007) 573578.

    [39] T.L. Plank, R.S. Yeung, E.P. Henske, Hamartin, the product of the tuberoussclerosis 1 (TSC1) gene interacts with tuberin and appears to be localised tocytoplasmic vesicles, Cancer Res. 58 (1998) 47664770.

    [40] M. Van Slegtenhorst, N. Nellist, B. Nagelkerken, J. Cheadle, R. Snell, A. van denOuweland,A. Reuser, J.Sampson,D. Halley, P.van derSluijs,Interaction betweenhamartin and tuberin, the TSC1 and TSC2 gene products, Hum. Mol. Genet. 7(1998) 10531057.

    [41] M. Rosner, M. Hanneder, N. Siegel, A. Valli, M. Hengstschlager, The tuberous

    sclerosis gene products hamartin and tuberin are multifunctional proteins witha wide spectrumof interacting partners,Mutat. Res.: Rev. Mutat.Res.658 (2008)234246.

    [42] A.Onda,A. Lueck,P.W.Marks, H.B. Warren, D.J. Kwiatkowski, TSC+ mice developtumors in multiple sites that express gelsolin and are influenced by geneticbackground, J. Clin. Invest. 104 (1999) 687695.

    [43] T. Kobayashi, O. Minowa, J. Kuno, H. Mitani, O. Hino, T. Noda, Renal carcinogen-esis, hepatic hemaniomatosis, and embryonic lethality caused by a germ-lineTSC2 mutation in mice, Cancer Res. 59 (1999) 12061211.

    [44] T. Kobayashi, O. Minowa, Y.Sugitani, S. Takai,H. Mitani, E. Kobayashi, T. Noda, O.Hino, A germ-line TSC1 mutation causes tumor development and embryoniclethality that are similar, but not identical to those caused by TSC2 mutation inmice, Proc. Natl. Acad. Sci. U.S.A. 98 (2001) 87628767.

    [45] M. Rosner, A. Freilinger, G. Lubec, M. Hengstschlager, The tuberous sclerosisgenes, TSC1 and TSC2, trigger different gene expression responses, Int. J. Oncol.27 (2005) 14111424.

    [46] M.Hengstschlager,M. Rosner, M. Fountoulakis,G. Lubec, Thecellularresponsetoectopic overexpression of the tuberous sclerosis genes, TSC1 and TSC2: a

    proteomic approach, Int. J. Oncol. 27 (2005) 831838.[47] H.C. Dan, M. Sun, L. Yang, R.L. Feldmann, X.-M. Sui, C. Chen Ou, M. Nellist, R.S.Yeung, D.J.J. Halley, S.V. Nicosia, W.J. Pledger, J.Q. Cheng, Phosphatidylinositol 3-kinase/Akt pathway regulates tuberous sclerosis tumor suppressor complex byphosphorylation of tuberin, J. Biol. Chem. 277 (2002) 3536435370.

    [48] E.A. Goncharova, D.A. Goncharov, A.W. Eszterhas, D.S. Hunter, M.K. Glassberg,R.S. Yeung, C.L. Walker, D. Noonan, D.J. Kwiatkowski, M.M. Chou, R.A. Panettieri,V.P. Krymskaya, Tuberin regulates p70 S6kinase activation and ribosomal pro-tein S6 phosphorylation, J. Biol. Chem. 277 (2002) 3095830967.

    [49] A.R. Tee, D.C. Fingar, B.D. Manning, D.J. Kwiatkowski, L.C. Cantley, J. Blenis,Tuberous sclerosis complex-1 and -2 gene products function together to inhibitmammalian target of rapamycin (mTOR)-mediated downstream signalling,Proc. Natl. Acad. Sci. U.S.A. 99 (2002) 1357113576.

    [50] K. Inoki, Y. Li, T. Zhu, J. Wu, K.-L. Guan, TSC2 is phosphorylated and inhibited byAkt and suppresses mTOR signalling, Nat. Cell Biol. 4 (2002) 648657.

    [51] B.D. Manning, A.R. Tee, M.N. Logsdon, J. Blenis, L.C. Cantley, Identification of thetuberoussclerosiscomplex-2tumor suppressorgene product tuberin as a targetof the phosphoinositide 3-kinase/akt pathway, Mol. Cell 10 (2002) 151162.

    [52] C.J. Potter, L.G. Pedraza, T. Xu, Akt regulates growth by directly phosphorylating

    Tsc2, Nat. Cell Biol. 4 (2002) 658665.[53] P.B.Crino,K.L. Nathanson, E.P.Henske, The tuberous sclerosiscomplex, N. Engl.J.

    Med. 355 (2006) 13451356.[54] J.J. Bissler, F.X. McCormack, L.R. Young, J.M. Elwing, G. Chuck, J.M. Leonard, V.J.

    Schmithorst, T. Laor, A.S. Brody, J. Bean, S. Salisbury, D.N. Franz, Sirolimus forangiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis,N. Engl. J. Med. 358 (2008) 140151.

    [55] D.M. Davies, S.R. Johnson, A.E. Tatterfield, J.C. Kingswood, J.A. Cox, D.L. McCart-ney, T. Doyle, F. Elmslie, A. Saggar, P.J. deVries, Sirolimus therapy in tuberoussclerosis or sporadic lymphangioleiomyomatosis, N. Engl. J. Med. 358 (2008)200203.

    [56] M.N. Corradetti, K. Inoki, N. Bardeesy, R.A. DePinho, K.-L. Guan, Regulation of theTSC pathway by LKB1: evidence of a molecular link between tuberous sclerosiscomplex and Peutz-Jeghers syndrome, Genes Dev. 18 (2004) 15331538.

    [57] R.J. Shaw, N. Bardeesy, B.D. Manning, L. Lopez, M. Kostmatka, R.A. DePinho, C.Cantley, The LKB1 tumor suppressor negatively regulates mTOR signalling,Cancer Cell 6 (2004) 9199.

    [58] K. Inoki, H. Ouyang, T. Zhu, C. Lindvall, Y. Wang, X. Zhang, Q. Yang, C. Bennet, Y.Harada, K. Stankunas, C.-Y. Wang,X. He, O.A.MacDougald, M. You,B.O. Williams,

    K.-L. Guan, TSC2 integrates Wnt and energy signals via a coordinated phosphor-ylation by AMPK and GSK3 to regulate cell growth, Cell 126 (2006) 955968.

    [59] J. Brugarolas, W.G. Kaelin, Dysregulation of HIF and VEGF is a unifying feature ofthe familial hamartoma syndromes, Cancer Cell 6 (2004) 710.

    [60] K. Inoki, M.N. Corradetti, K.-L. Guan, Dysregulation of the TSC-mTOR pathway inhuman disease, Nat. Genet. 37 (2005) 1924.

    [61] J. Brugarolas, F. Vazquez, A. Reddy, W.R. Sellers, W.G. Kaelin, TSC2 regulatesVEGF through mTOR-dependent and -independent pathways, Cancer Cell 4(2003) 147156.

    [62] N. El-Hashemite, V. Walker, H. Zhang, D.J. Kwiatkowski, Loss of Tsc1 and Tsc2

    induces vascular endothelial growth factor production through mammaliantarget of rapamycin, Cancer Res. 63 (2003) 51735177.

    [63] W.G. Kaelin, The von Hippel-Lindau tumorsuppressor gene and kidney cancer,Clin. Cancer Res. 10 (2004) 6290S6295S.

    [64] J. Brugarolas, Renal-cellcarcinomamolecular pathway and therapies,N. Engl.J.Med. 356 (2007) 185187.

    [65] C. Johannessen, E.E. Reczek, M.F. James, H. Brems, E. L egius, K. Cichowski, TheNF1 tumor suppressor critically regulates TSC2 and mTOR, Proc. Natl. Acad. Sci.U.S.A. 102 (2005) 85738578.

    [66] C. Johannessen, B.W. Johnson, S.M.G. Williams, A.W. Chan, E.E. Reczek, R.C.Lynch, M.J. Rioth, A. McClathehey, S. Ryeom, K. Cichowski, TORC1 is essentialfor NF1-associated malignancies, Curr. Biol. 18 (2008) 5662.

    [67] E. Kleymenova, O. Ibraghimov-Beskrovnaya, H. Kugoh, J. Everitt, H. Xu, K.Kiguchi, G. Landes, P. Harris, C.L. Walker, Tuberin-dependent membrane loca-lization of polycystin-1: a functional linkbetween polycystickidney disease andthe TSC2 tumor suppressor gene, Mol. Cell 7 (2001) 823832.

    [68] J.M. Shillingford, N.S. Murcia, C.H. Larson, S.H. Low, R. Hedgepeth, N. Brown, C.A.Flask, A.C. Novick, D.A. Goldfarb, A. Kramer-Zucker, G. Walz, K.B. Piontek, G.G.Germino, T. Weimbs, The mTOR pathway is regulated by polycystin-1 and its

    inhibition reverses renal cystogenesis in polycystic kidney disease, Proc. Natl.Acad. Sci. 103 (2006) 54665471.

    [69] M.J. Surpili, T.M. Delben, J. Kobarg, Identification of proteins that interact withthecentral coiled-coilregionof thehuman protein kinase NEK1, Biochemistry 42(2003) 1536915376.

    [70] T. Weimbs, Polycystic kidney disease and renal injury repair: common pathway,fluid flow, and the function of polycystin-1, Am. J. Ren. Physiol. 293 (2007)F1423F1432.

    [71] L.A. Haddad, N. Smith, M. Browser, Y. Niida, V. Murthy, C. Gonzalez-Agosti, V.Ramesh, The TSC1 tumor suppressor hamartin interacts with neurofilament-Land possibly functions as a novel integrator of the neuronal cytoskeleton, J. Biol.Chem. 277 (2002) 4418044186.

    [72] T. Soucek, G. Holzl, G. Bernaschek, M. Hengstschlager, A role of the tuberoussclerosis gene-2 product during neuronal differentiation, Oncogene 16 (1998)21972204.

    [73] R.E. Tanzl, L. Bertram, Twenty years of the Alzheimers disease amyloid hypoth-esis: a genetic perspective, Cell 120 (2005) 545555.

    [74] R. Ferrando-Miguel, M. Rosner, A. Freilinger, G. Lubec, M. Hengstschlager,

    Tuberina new molecular target in Alzheimers disease? Neurochem. Res. 30(2005) 14131419.[75] R.J. Griffin, A. Moloney, M. Kelliher, J.A. Johnston, R. Ravid, P. Dockery, R.

    OConnor, C. ONeill, Activation of Akt/PKB, increased phosphorylation of Aktsubstrates and loss and altered distribution of Akt and PTEN are features ofAlzheimers disease pathology, J. Neurochem. 93 (2005) 105117.

    [76] M. Damjanac, A. Rioux Bilan, R. Paccalin, B. Pontcharraud, J. Fauconneau, G.Hugon, Page,dissociation of Akt/PKB and ribosomal S6 kinase signalling markersin a transgenic mouse model of Alzheimers disease, Neurobiol. Dis. 29 (2008)354367.

    [77] T. Chano, H. Okabe, C.M. Hulette, RB1CC1 insufficiency causes neuronal atrophythrough mTOR signalling alteration and is involved in the pathology of Alzhei-mers disease, Brain Res. 1168 (2007) 97105.

    [78] X. Li, I. Alafuzoff, H. Soininen, B. Winblad, J.J. Pei, Levels of mTOR and itsdownstream targets 4E-BP1, eEF2, and eEF2 kinase in relationships with tauin Alzheimers disease brain, FEBS J. 272 (2005) 42114220.

    [79] W.L. An,R.F.Cowburn, H.Braak, I.Alafuzoff, K.Iqbal, I.G. Iqbal,B. Winblad,J.J.Pei,Up-regulation of phosphorylated/activated p70 S6 kinase and its relationship toneurofibrillary pathology in Alzheimers disease, Am. J. Pathol. 163 (2003) 591

    607.[80] B.-H. Jiang, L.-Z. Liu, PI3K/PTEN signalling in tumorigenesis and angiogenesis,

    Biochim. Biophys. Acta 1784 (2008) 150158.[81] A.D. Basso, A. Mirza, G. Liu, B.J. Long, W.R. Bishop, P. Kirschmeier, The FTI

    SCH66336 (Lonafarnib) inhibits Rheb farnesylation and mTOR signalling: roleof FTI enhancement of taxane and tamoxifen anti-tumor activity, J. Biol. Chem.280 (2005) 3110131108.

    [82] M. Barlund,F. Forozan, J. Kononen,L. Bubendorf, Y. Chen, M.L.Bittner,J. Torhorst,P. Haas,C. Bucher, G. Sauter, O.P.Kallioniemi,A. Kallioniemi, Detectingactivationof ribosomal protein S6 kinase by complementary DNA and tissue microarrayanalysis, J. Natl. Cancer Inst. 92 (2000) 12521259.

    [83] R.J. Shaw, L.C. Cantley, Ras, PI3K and mTOR signalling controls tumour cellgrowth, Nature 441 (2006) 424430.

    [84] L. Ma, Z. Chen, H. Erdjument-Bromage, P. Tempst, P.P. Pandolfi, Phosphorylationand functional inactivation of TSC2 by ERK: implications for tuberous sclerosisand cancer pathogenesis, Cell 121 (2005) 179193.

    [85] P.P.Roux,B.A. Ballif,R. Anjum,S.P. Gygi, J. Blenis, Tumor-promotingphorbol estersand activated Ras inactivate the tuberous sclerosistumor suppressor complex viap90 ribosomal S6 kinase, Proc. Natl. Acad. Sci. 101 (2004) 1348913494.

    M. Rosner et al./ Mutation Research 659 (2008) 284292 291

  • 8/2/2019 Mtor Review

    9/9

    [86] A. Freilinger, M. Rosner, G. Krupitza, M. Nishino, G. Lubec, S.J. Korsmeyer, M.Hengstschlager, Tuberin activates the proapoptotic molecule BAD, Oncogene 25(2006) 64676479.

    [87] A. Freilinger, M. Rosner, M. Hanneder, M. Hengstschlager, Ras mediates cellsurvival by regulating tuberin, Oncogene 27 (2008) 20722083.

    [88] D.-F. Lee, H.-P. Kuo, C.-T. Chen, J.-M. Hsu, C.-K. Chou, Y. Wie, H.-L. Sun, L.-Y. Li, B.Ping, W.-C. Huang, X. He, J.-Y. Hung, C.-C. Lai, Q. Ding, J.-L. Su, J.-Y. Yang, A.A.Sahin, G.N. Hortobagyi, F.-J. Tsai, C.-H. Tsai, M.-C. Hung, IKKb suppression ofTSC1 links inflammation and tumor angiogenesis via the mTOR pathway, Cell130 (2007) 440455.

    [89] A.Y. Choo, P.R. Roux, J. Blenis, Mind the GAP: Wnt steps onto the mTORC1 train,Cell 126 (2006) 834836.

    [90] T. Reya, H. Clevers, Wnt signalling in stem cells and cancer, Nature 434 (2005)843850.

    [91] B.C. Mak, K.-I. Takemaru, H.L. Kenerson, R.T. Moon, R.S. Yeung, The tuberin-hamartin complex negatively regulates b-catenin signaling activity, J. Biol.Chem. 278 (2003) 59475951.

    [92] B.C. Mak, H.L. Kenerson, L.D. Aicher, E.A. Barnes, R.S. Yeung, Aberrrantb-cateninsignaling in tuberous sclerosis, Am. J. Pathol. 167 (2005) 107116.

    [93] S.C. Hanna, S.A. Heathcote, W.Y. Kim, mTOR pathway in renal cell carcinoma,Expert Rev. Anticancer Ther. 8 (2008) 283292.

    [94] M.A. Knowles, N. Hornigold, E. Pitt, Tuberous sclerosis complex (TSC) geneinvolvement in sporadic tumours, Biochem. Soc. Trans. 31 (2003) 597602.

    [95] M.A. Knowles, T. Habuchi, W. Kennedy, D. Cuthbert-Heavens, Mutation spec-trum of the 9q34 tuberous sclerosis gene TSC1 in transitional cell carcinoma ofthe bladder, Cancer Res. 63 (2003) 76527656.

    [96] H. Adachi, M. Igawa, H. Shiina, S. Urakami, K. Shigeno, O. Hino, Human bladdertumors with 2-hit mutations of the tumor suppressor gene TSC1 and decreasedexpression of p27, J. Urol. 170 (2003) 601604.

    [97] L.S. Pymar, F.M. Platt, J.M. Askham, E.E. Morrison, M.A. Knowles, Bladder tumorderived somatic TSC1 missense mutations cause loss of function via distinctmechanisms, Hum. Mol. Genet. (April 2008) (Epub ahead).

    [98] K. Kataoka, K. Fujimoto, D. Ito, M. Koizumi, E. Toyoda, T. Mori, K. Kami, R. Doi,Expressionand prognosticvalue of tuberoussclerosiscomplex 2 gene product inhuman pancreatic cancer, Surgery 138 (2005) 450455.

    [99] W.G. Jiang, J. Sampson, T.A. Martin, L. Lee-Jones, G. Watkins, A. Douglas-Jones, K.Mokbel, R.E. Mansel, Tuberin and hamartin are aberrantly expressed and linkedto clinical outcome in human breast cancer: the role of promoter methylation ofTSC genes, Eur. J. Cancer 41 (2005) 16281636.

    [100] R.G. Weber, A. Hoischen, M. Ehrler, P. Zipper, K. Kaulich, B. Blaschke, A.J. Becker,S. Weber-Mangal, A. Jauch, B. Radlwimmer, J. Schramm, O.D.Wiestler,P. Lichter,G. Reifenberger, Frequent loss of chromosome 9, homozygous CDKN2A/p14/CDKN2B deletion and low TSC1 mRNA expression in pleiomorphic xanthoas-trocytomas, Oncogene 26 (2007) 10881097.

    [101] K.H.Lu, W.Wu, B.Dave,B.M.Slomovitz, T.W. Burke,M.F.Munsell, R.R. Broaddus,C.L. Walker, Loss of tuberous sclerosis complex-2 function and activation of

    mammalian target of rapamycin signalling in endometrial cancer, Clin. CancerRes. 14 (2008) 25432550.

    [102] C.J. Sherr, J.M. Roberts, CDK inhibitors: positive and negative regulators of G1-phase progression, Genes Dev. 13 (1999) 15011512.

    [103] C.J. Sherr, The Pezcoller lecture: cancer cell cycles revisited, Cancer Res. 60(2000) 36893695.

    [104] T. Soucek, O. Pusch, R. Wienecke, J.E. DeClue, M. Hengstschlager, Role of thetuberous sclerosis gene-2 product in cell cycle control, J. Biol. Chem. 272 (1997)2930129308.

    [105] T. Soucek, R.S. Yeung, M. Hengstschlager, Inactivation of the cyclin-dependent

    kinase inhibitor p27 upon loss of the tuberous sclerosis complex gene-2, Proc.Natl. Acad. Sci. U.S.A. 95 (1998) 1565315658.

    [106] A. Miloloza, M. Rosner, M. Nellist, D. Halley, G. Bernaschek, M. Hengstschlager,The TSC1 gene product, hamartin, negatively regulates cell proliferation, Hum.Mol. Genet. 9 (2000) 17211727.

    [107] T. Soucek, M. Rosner, A. Miloloza, M. Kubista, J.P. Cheadle, J.R. Sampson, M.Hengstschlager, Tuberous sclerosis causing mutants of the TSC2 gene pro-duct affect proliferation and p27 expression, Oncogene 20 (2001) 49044909.

    [108] M. Rosner, M. Hengstschlager, Tuberin binds p27 and negatively regulates itsinteraction with the SCF component Skp2, J. Biol. Chem. 279 (2004) 4870748715.

    [109] M. Rosner, A. Freilinger, M. Hanneder, N. Fujita, G. Lubec, T. Tsuruo, M. Hengsts-chlager, p27KIP1 localization depends on the tumor suppressor protein tuberin,Hum. Mol. Genet. 16 (2007) 15411556.

    [110] A. Alkarain, R. Jordan, J. Slingerland, p27 deregulation in breast cancer: prog-nostic significance and implications for therapy, J. Mammary Gland Biol. Neo-plasia 67 (2004) 6780.

    [111] M. Rosner, A. Freilinger, M. Hengstschlager, The tuberous sclerosis genes and

    regulationof the cyclin-dependent kinase inhibitorp27, Mutat. Res.:Rev. Mutat.Res. 613 (2006) 1016.

    [112] E. Blair, C. Redwood, H. Ashrafian, M. Oliveira, J. Broxholme, B. Ker, A. Salmon, I.Ostman-Smith, H. Watkins, Mutations in the gamma(2) subunit of AMP-acti-vated protein kinase cause familial hypertrophic cardiomyopathy: evidence forthecentral roleof energy compromisein disease pathogenesis, Hum.Mol. Genet.10 (2001) 12151220.

    [113] M.H. Gollob, M.S.Green, A.S.Tang, T. Gollob, A. Karibe, A.S.Ali Hassan, F. Ahmad,R. Lozado, G. Shah, L. Fananapazir, L.L. Bachinski, R. Roberts, Identification of agene responsible for familial Wolff-Parkinson-White syndrome, N. Engl. J. Med.344 (2001) 18231831.

    [114] T. Shioi, J.R. McMullen, O. Tarnavski, K. Converso, M.C. Sherwood, W.J. Manning,S. Izumo, Rapamycin attenuates load-induced cardiac hypertrophy in mice,Circulation 107 (2003) 16641670.

    [115] A.Y. Chan, C.L. Soltys, M.E. Young, C.G. Proud, J.R. Dyck, Activation of AMP-activated protein kinase inhibits protein synthesis associated with hypertrophyin the cardiac myocyte, J. Biol. Chem. 279 (2004) 3277132779.

    M. Rosner et al./ Mutation Research 659 (2008) 284292292