Oncogene (2014) 33, 4709–4721 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc

REVIEW Oncoprotein stabilization in brain tumors

S-M Hede1, V Savov1, H Weishaupt1, O Sangfelt2 and FJ Swartling1

Proteins involved in promoting cell proliferation and viability need to be timely expressed and carefully controlled for the proper development of the brain but also efficiently degraded in order to prevent cells from becoming brain cancer cells. A major pathway for targeted degradation in cells is the –proteasome system (UPS). Oncoproteins that drive tumor development and tumor maintenance are often deregulated and stabilized in malignant cells. This can occur when oncoproteins escape degradation by the UPS because of mutations in either the oncoprotein itself or in the UPS components responsible for recognition and ubiquitylation of the oncoprotein. As the pathogenic accumulation of an oncoprotein can lead to effectively sustained cell growth, viability and tumor progression, it is an indisputable target for cancer treatment. The most common types of malignant brain tumors in children and adults are medulloblastoma and glioma, respectively. Here, we review different ways of how deregulated proteolysis of oncoproteins involved in major signaling cancer pathways contributes to medulloblastoma and glioma development. We also describe means of targeting relevant oncoproteins in brain tumors with treatments affecting their stability or therapeutic strategies directed against the UPS itself.

Oncogene (2014) 33, 4709–4721; doi:10.1038/onc.2013.445; published online 28 October 2013 Keywords: glioma; medulloblastoma; ubiquitin–proteasome system; brain tumor development; E3 ligases

INTRODUCTION system itself may be attractive candidates for brain cancer Medulloblastoma is the most common malignant brain tumor in therapy. children. Around 70% of patients are cured after surgery and radiotherapy; however, survivors often succumb from serious side effects following these treatments.1 Glioblastoma (GBM) is one of MEDULLOBLASTOMA BIOLOGY the most lethal of all cancers and also the most common type of Recent genomic efforts have divided medulloblastoma into four primary malignant brain tumors (glioma) in adults.2 GBM is a WHO main molecular subgroups: WNT, SHH (Sonic Hedgehog), Group 3 grade IV astrocytoma and can either develop de novo without and Group 4.5 For all molecular subgroups, there are key previous diagnosis (primary GBM) or derive and progress into a medulloblastoma that are characterized as target secondary GBM from an astrocytoma of a lower grade. Other proteins for the UPS. For example, the WNT (Wingless) pathway less frequent types of glioma include oligodendroglioma and substrate b-catenin is regulated by the UPS through the beta- ependymoma. transducin repeat-containing E3 Ub protein ligase (beta-TrCP).6 In recent years, great strides have been made in characterizing The beta-TrCP recognition site of the b-catenin CTNNB1 is changes in or genetic and epigenetic alterations often mutated, leading to b-catenin stabilization in WNT tumors.5,7 associated with the genesis and/or progression of various types of The SHH tumors show alterations in proteins that lead to SHH brain tumors. However, less is known about how alterations in pathway activation. For example, the beta-TrCP substrate, GLI protein level and protein activity contribute to brain tumor family zinc-finger 2 (GLI2),8 is often elevated and is a marker of development. One specific type of post-translational modification poor prognosis for SHH tumors.9 Furthermore, inactivation of is the covalent attachment of a small (8 kDa) and highly conserved Patched (PTCH)10,11 can result in stabilization of the downstream protein called ubiquitin (Ub), in a process known as ubiquityla- MYCN oncoprotein.12 Finally, Group 3 and Group 4 tumors often tion.3 Ubiquitylation typically involves (1) the covalent attachment present amplifications in MYC and MYCN, respectively,13,14 of Ub to a specific protein substrate and (2) the degradation of the although MYC protein levels can also be elevated in other polyubiquitylated substrate in a large multisubunit protease tumors without amplifications in MYC .15 complex known as the 26S proteasome.4 The ubiquitin–proteasome system (UPS) controls diverse cellular processes involving proliferation, maturation, differentia- GLIOBLASTOMA BIOLOGY tion and cell death. A wealth of recent experimental evidence GBMs have recently been divided into 3–6 molecular subgroups further demonstrates that deregulation of the Ub system has a depending on the classification scheme and clustering.16,17 The causative role in cancer development and progression. In this most commonly used scheme is provided by The Cancer Genome review, we describe important UPS substrates involved in brain Atlas currently classifying GBMs into four molecular subtypes: tumor development and the potential premise that protein Proneural, Neural, Classical and Mesenchymal groups.18 GBMs are stabilization can be targeted or that components of the Ub genetically diverse; however, generally speaking, 70–80% of GBMs

1Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden and 2Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden. Correspondence: Dr F Swartling, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden. E-mail: [email protected] Received 29 June 2013; revised 11 September 2013; accepted 12 September 2013; published online 28 October 2013 Oncoprotein stabilization S-M Hede et al 4710 are frequently characterized by loss of 10 and gain repair, among others.27 Ubiquitylation is counteracted by specific of . Several proteins driving GBM development and isopeptidases called deubiquitinating or DUBs, which progression are direct or indirect targets of the UPS. Receptor catalyze the hydrolysis of the peptide bond between the Ub tyrosine kinases (RTKs) that are commonly overexpressed or molecule and the substrate, highlighting the reversibility of the Ub amplified in GBMs including epidermal growth factor receptors system (Figure 1).28 (EGFRs), platelet-derived growth factor (PDGF) receptors and MET (encoding the hepatocyte growth factor receptor) are normally targeted for degradation by the CBL proto-oncogene and E3 Ub E3 Ub enzymes protein ligase (CBL), and these RTKs often find means to avoid CBL In the Ub system, substrate specificity is achieved through the use recognition during cancer development.19 Important downstream of a large number of different E3s that physically interact with kinases including phosphatidylinositol 3’-kinase (PI3K) and the specific target substrates, linking them to the E2 Ub-conjugating 29 mammalian target of rapamycin (mTOR) are also targets of the . E3 ligases are encoded by at least 600 human genes, 30 UPS. For instance, PI3K is regulated by the Ub ligases CBL-B20 and thus exceeding the number of protein kinase genes. The F-box and leucine-rich repeat protein 2 (FBXL2),21 whereas the majority of E3s contain a RING (Really-Interesting New Gene)- F-box and WD repeat domain containing 7 (FBW7), E3 Ub protein finger homology domain, which binds to the E2 enzyme and ligase promotes proteasomal degradation of mTOR.22 The E3 Ub directly mediates the transfer of Ub to the substrate. In contrast, ligase mouse double minute 2 () is a well-established HECT (Homologous to E6-AP C-Terminus) domain E3s form a oncoprotein and negative regulator of p53 protein expression.23 transient covalent linkage between Ub and a cysteine residue on Several other proteins regulated by the UPS that are involved in the E3 prior to transferring the Ub molecule to the bound 31 brain tumor development and progression will be further substrate. RING E3 ligases can be further categorized into single reviewed below. subunit E3s (one protein contains both a RING domain and a substrate-binding domain—for example, MDM2) or multisubunit E3s (for example, -RING ligases (CRLs)). The CRL is the largest THE UPS family of E3s and includes the well-described SCF (SKP1-Cullin1-F- box) and APC/C (anaphase-promoting complex/cyclosome) Ub Breakdown of proteins (proteolysis) occurs constantly inside cells ligase complexes that have central roles in regulation, as the production of new proteins must be balanced by an equal checkpoint control and genome stability.32 amount of protein degradation for cells to survive. The molecular The APC/C is composed of at least 13 protein subunits and machinery catalyzing the majority of intracellular protein degrada- 24,25 targets numerous cell cycle regulators for proteolysis in mitosis tion is known as the UPS. and in G1. The SCF Ub ligases consist of the following four subunits: the RING protein retinoblastoma (RB)X1, the scaffold The Ub enzymatic cascade protein Cullin1, the adapter protein SKP1 and a variable substrate- binding component known as the F-box protein.32 SCF ligases are The decoration of protein substrates with chains of Ub molecules B is the result of multiple rounds of Ub-conjugation reactions carried active throughout the cell cycle, and to date, 70 different F-box proteins have been identified in humans, each likely targeting a out by a three-step enzymatic cascade. First, an E1 enzyme 33 mediates the ATP-dependent activation of Ub. Next, Ub is unique set of substrates for degradation. Not surprisingly, translocated to an E2-conjugating enzyme, and finally, an E3 Ub deregulation of Ub ligases or their target substrates has a ligase facilitates the transfer of Ub to a specific lysine (K) residue causative role in cancer development and progression. on the target substrate (Figure 1).3 The 76-amino-acid Ub polypeptide has seven internal lysines, and Ub chains may be assembled through any of these lysine residues (in addition to the PROTEIN STABILIZATION AND REGULATION BY THE UPS IN amino group in the N-terminus of Ub). Substrates tagged with K48 BRAIN CANCER or K11-linked polyubiquitin chains are usually destined for The central nervous system arises from the neural plate, and the proteolytic destruction by the 26S proteasome,26 whereas brain and the spinal cord later develop from the neural tube of the substrates conjugated with a single Ub (monoubiquitin) or embryo. Proteins that are involved in early regulation of neuronal polyubiquitin chains linked through other lysines in Ub (for embryogenesis include bone morphogenetic proteins, SHH example, K63) have non-proteolytic functions modulating various proteins and fibroblast growth factors (FGFs).34 Other factors biological processes, such as , DNA replication and essential for normal brain development include the NOTCH family,

Ub Ub Ubiquitin Ub Conjugation DUBs Ub E3 Ub Substrate Ub Substrate E1 E2 Substrate ATP

ATP Specific E3 Degradation Ub Targeting Ub Ub Ub Ub Substrate 26S Proteasome E1 E2 Ub

Figure 1. Schematic overview of the UPS. Ubiquitylation, or Ubiquitin (Ub) conjugations of protein substrates, is an ATP-dependent reaction that involves a concerted effort of an ubiquitin-activating enzyme (E1), an ubiquitin-conjugating enzyme (E2) and an (E3). In the ubiquitylation process, E3 ligases are the primary sources of substrate specificity, resulting in ubiquitin conjugation to specific lysine (K) residue(s) on the protein substrate. Proteins with K48-linked polyubiquitin chains are degraded by the 26S proteasome complex in an ATP- dependent manner, whereas other types of ubiquitin chains (for example, K63) have non-proteolytic functions regulating protein activity. The ubiquitylation process is counteracted by DUBs enzymes that hydrolyze ubiquitin–protein peptide bonds and recycle ubiquitin molecules.

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4711 Table 1. Oncoprotein stabilization in brain tumor models and molecular targets for putative therapies

Cancer protein/s/ stabilized or altered Brain tumor model/cell system Potential destabilizing treatment strategy

Medulloblastoma CTNNB1 (exon 3) with conditional Floxed exon 3 stabilizes b-catenin generating WNT Targeting downstream TCF-dependent p53 loss medulloblastoma45 transcription MYCWT/MYCT58A with p53 loss A mutationally stabilized MYC protein drives Targeting MYC by inhibiting CDKs, Aurora Kinases, Group 3 medulloblastoma114,115 BRDs or PI3K/mTOR MYCNT58A A mutationally stabilized MYCN protein drives Targeting MYCN by inhibiting CDKs, Aurora SHH or Group 4 medulloblastoma117 Kinases, BRDs or PI3K/mTOR MYCN via PTCH1 loss Patched1 loss increases MYCN protein stability as Targeting SMO or GLI proteins a mechanism of medulloblastoma initiation12

Glioma MYCNT58A Stabilized MYCN protein drives MG-PNET or Targeting MYCN by inhibiting CDKs, Aurora pediatric glioma117 Kinases, BRDs or PI3K/mTOR PDGFB Post-translationally modified PDGFB lacking UTR Targeting PDGF ligand binding or PDGF receptors parts drives more aggressive gliomas68 MYC in PTEN À / À /Ink4a/Arf À / À NSCs FBW7 loss raises MYC levels in NSCs to generate Targeting MYC by inhibiting CDKs, Aurora Kinases, and Fbxw7 knockdown GBMs187 BRDs or PI3K/mTOR E Knockdown of PARK2 accumulates Cyclin E levels Inhibiting CDKs that bind with Cyclin E in glioma cells137 TRIM11 TRIM11 promotes accumulation of EGFR and Inhibitors of EGFR or MEK MAPK activation199 Abbreviations: BRDs, BET Bromodomain proteins; CDK, Cyclin-dependent kinase; EGFR, epidermal growth factor receptor; GLI, GLI family zinc finger; MG-PNET, malignant glioma with PNET-like morphologies; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; mTOR, mammalian target of rapamycin; NSC, neural stem cell; PARK2, Parkinson protein 2, E3 ubiquitin protein ligase (); PI3K, phosphatidylinositol 3’-kinase; PDGF, platelet- derived growth factor; PTCH1, Patched1; PTEN, phosphatase and tensin homolog; SHH, Sonic Hedgehog; SMO, Smoothened; TCF, transcription factor; TRIM11, tripartite motif-containing protein 11; UTR, untranslated region.

WNT proteins as well as various kinase receptors and their like SCFb-TrCP further regulates downstream signaling of the SHH downstream targets, including transcription factors and cell cycle pathway, as GLI2 is also targeted by SCFb-TrCP and then degraded.8 regulators. Several of these proteins are intimately involved in Gorlin’s syndrome is a rare inherited condition with germline brain tumor formation and cancer progression. Important mutations in the PTCH gene. It is characterized by an increased mechanisms that control oncoprotein stabilization during tumor incidence of tumors including basal cell carcinoma and development have further been discovered when using various medulloblastoma.10 Interestingly, step-wise loss of PTCH1 during animal models of brain tumors (Table 1). In the following sections, tumor formation and progression in mice leads to stabilization of we will review the recent literature of how the UPS regulates the MYCN protein12 (Table 1). proteins involved in brain tumor biology. The WNT pathway. There are two different WNT pathways: the Developmental pathways canonical or the WNT/b-catenin pathway controlling cell-fate determination and the non-canonical pathway regulating cell The SHH pathway. The Hedgehog pathway is a well-conserved movement and tissue polarity.41 SCFb-TrCP negatively regulates the developmental pathway. Hedgehog ligands, such as SHH, act as WNT/b-catenin signaling pathway by targeting b-catenin for morphogens during embryogenesis and regulate cell fate and 35 ubiquitylation following GSK3b-mediated phosphorylation on brain patterning. SHH signaling depends on a functional primary specific serine/threonine residues in b-catenin at a conserved cilium—a non-motile microtubule organelle that forms an 6 36 beta-TrCP-binding motif (degron). These residues are commonly antenna-like structure on the cell membrane. When SHH binds mutated on CTNNB1 in the WNT group of medulloblastoma,7,42 to the inhibitory receptor PTCH it releases smoothened, leading to 43 37 resulting in defective degradation of b-catenin because of the the activation of GLI family (GLI1-3) transcription factors. failure of beta-TrCP to interact with b-catenin.44 Similarly, Whereas GLI1 works as an activator of the SHH pathway, both mutationally stabilized CTNNB1 has been valuable in describing GLI2 and GLI3 can be processed into transcription repressor forms. how WNT-driven medulloblastoma could be derived from distinct A speckle-type POZ protein—cullin 3 Ub ligase—promotes developing brain stem regions (of the lower rhombic lip) in ubiquitylation and degradation of GLI2 and GLI3 full-length 45 38 transgenic animals (Table 1). Mutations in another member of activators. GLI1 is constitutively active and cannot convert the WNT pathway, the adenomatous polyposis coli (APC) gene, between activator and repressor forms. GLI1 interacts with itchy can also lead to stabilization of b-catenin in the development of E3 Ub protein ligase (ITCH)—a member of the HECT family of E3 medulloblastoma in Turcot’s syndrome.46 enzymes.39 ITCH binds GLI1 and promotes its polyubiqitylation in collaboration with numb homolog (Drosophila) (NUMB)—an adaptor protein that enhances ITCH activity and GLI1 The NOTCH pathway. NOTCH signaling is involved in the degradation.39 Interestingly, NUMB expression is attenuated in communication between neighboring cells during brain develop- the SHH group of medulloblastoma consistent with its role in ment.47 Interaction with ligands, DELTA or JAGGED, results in promoting GLI1 degradation. In the absence of SHH, GLI3 is sequential cleavage of NOTCH receptors at the membrane and phosphorylated at multiple sites by v-akt murine thymoma viral translocation of the NOTCH intracellular domains into the nucleus, oncogene homolog 1 (AKT), glycogen synthase kinase 3 beta where they serve as transcription factors regulating transcription (GSK3b) and casein kinase 1 (CK1). This leads to direct binding and of various target genes including HES (hairy and enhancer of split) ubiquitylation by the SCF Ub ligase, SCFb-TrCP and subsequent or HES-related (HESR/HEY) transcription factors.48 The NOTCH processing of GLI3 into its transcription-repressor form.40 It seems pathway is tightly controlled by the UPS system at several levels.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721 Oncoprotein stabilization S-M Hede et al 4712 NOTCH receptors are regulated by multiple E3s including ITCH, The tyrosine kinase receptor MET is amplified in a smaller DELTEX and NEDD4,49 and turnover of the unstable NOTCH fraction of GBMs.64 In addition, MET is overexpressed in many intracellular domains is mediated by the Ub ligase FBW7 (also GBMs75 in which expression correlates with increasing malignancy known as SEL-10, Ago and CDC4).50,51 FBW7 preferentially targets grade of the brain tumor. The degradation of activated RTKs can NOTCH intracellular domain phosphorylated by Cyclin C-CDK8 for be altered on many levels in human tumors. The activity of CBL is degradation in the nucleus.52 The activated NOTCH1 intracellular regulated by the tyrosine kinase SRC. Phosphorylation by SRC domain induces SHH group medullobastoma when overexpressed leads to enhanced autoubiquitylating activity and proteasomal in p53-deficient animals.53 Surprisingly, NOTCH2 rather than degradation of both CBL and SRC.76 An alternative degradation NOTCH1 levels are elevated in tumors generated from this mechanism was shown for EGFR in clear cell renal carcinoma, in model. As compared with NOTCH1 levels, NOTCH2 levels are which polyubiquitylation of the activated RTK by von Hippel– also elevated in primary human medulloblastoma.54 NOTCH1 Lindau tumor suppressor, E3 Ub protein ligase leads to nuclear staining further correlates with improved outcomes of proteasomal degradation.77 Cancer cells have different ways of proneural high-grade glioma.55 These data may suggest that escaping CBL recognition and avoiding RTK degradation by the NOTCH1 activity regulated by UPS is hypothetically necessary for UPS. For instance, CBL-binding sites on RTK genes can be mutated, tumor initiation but not for tumor progression. CBL recruitment could become inefficient, degradation of CBL itself could increase or mutations could generate an RTK that will form a fusion protein that escapes proteasomal degradation.19 Kinase receptors and downstream pathways Receptor tyrosine kinases. RTKs are transmembrane proteins that, The transforming growth factor beta (TGF-b) pathway upon ligand-binding, dimerize and activate intracellular signaling The TGF-b family of growth factors signals through TGFR1 and pathways controlling cell proliferation, survival, migration and TGFR2, activating downstream signaling pathways via phosphor- differentiation.56 Signaling via EGFRs (ERBB1-4) is important in ylation of the SMAD family members (SMADs) that when activated controlling cell proliferation and fate during neurogenesis. function as transcription factors together with various cofactors. Signaling via PDGFRA and PDGFRB has important roles both Activation of TGF-b signaling results in differential responses, during embryogenesis and in adulthood (reviewed in Andrae depending on the cellular context.78 Ubiquitylation of the TGF-b et al.57). PDGFRA signaling controls neural stem cell (NSC) fate, receptors and downstream SMAD effector proteins is regulated by inducing the formation of oligodendrocytic rather than neuronal different E3 ligases such as the HECT-type SMAD-specific E3 Ub lineage.58,59 protein ligase (SMURF) repressing TGF-b signaling and RING-finger RTK activity is terminated both by the action of tyrosine domain E3 ligase Arkadia (also known as RNF111) enhancing the phosphatases and by internalization and degradation of the TGF-b signal.79 Indeed, dysregulation of both SMURFs and Arkadia activated receptor. RTK signaling leads to phosphorylation and have been found in several types of cancer.79 recruitment of the RING finger E3 Ub ligase CBL60 (See also Altered TGF-b signaling is found in many diseases and TGF-b is Figure 2). CBL binds to phosphorylated tyrosine residues on the known to promote epithelial–mesenchymal transition in cancer.80 activated RTK either directly via its tyrosine kinase-binding domain Normally, TGF-b signaling blocks proliferation by inducing or indirectly through interaction with adapter proteins such as expression of p21 via SMADs and FOXO transcription factors. GRB2. CBL ubiquitylates the intracellular part of the activated RTK, This is overcome in glioma cells where FOXG1 blocks FOXO and and monoubiquitylation at multiple sites serves as a signal for SMAD, and activation of the PI3 kinase pathway leads to receptor internalization via clathrin-coated pits followed by phosphorylation of FOXO.81 TGF-b signaling has also been lysosomal degradation.61 CBL functions as a negative regulator shown to promote proliferation of glioma cells in cases where of many RTKs, including EGFR (ERBB1), ERBB2, PDGFRA and the PDGFB gene is unmethylated. The hyperactive TGFb-SMAD PDGFRB, MET and FGFR (reviewed in Marmor and Yardon60). pathway here induces PDGFB levels, which correlate to poor Activation of oncogenic RTK signaling pathways in glioma can prognosis in glioma patients.82 be caused by elevated levels of ligand or increased abundance The DUB USP15 promotes TGFBR1 stability in GBMs by binding and activity of receptors. Both PDGF ligands and receptors are to the SMAD7-SMURF2 complex and removing Ubs from TGF-b found to be overexpressed in glioma62 often in combination with receptors. Expression of USP15 correlates with high TGF-b activity. the loss of p53.63 The PDGFRA gene is found to be amplified in Amplifications of USP15 further correlate with poor prognosis in 13% of GBM.64 Animal models have indicated that GBM tumors several tumor types including GBMs.83 may form by overexpression of PDGFB alone or in combination with the loss of p53.65–67 Interestingly, excessive expression of PDGFB ligand causes the formation of brain tumors in mice in a The PI3K/PTEN/AKT pathway and mTOR proteins dose-dependent way.68 In this study, certain untranslated regions PI3Ks are activated by several different types of membrane are responsible for post-translational modifications of PDGF, and receptors, including RTKs, T-cell receptors, integrins, cytokine removing these leads to PDGFB protein stabilization (Table 1). receptors and G-protein-coupled receptors. The class I PI3K is the The EGFR is found to be overexpressed and amplified in human best characterized subclass of PI3Ks and is composed of gliomas, and amplification of the EGFR gene is found in B40% of regulatory (p85) and catalytic (p110) subunits. The phosphatase GBM.69 The most common EGFR mutant in GBM is EGFRvIII, which and tensin homolog (PTEN) acts as a phosphatase for phospha- lacks the extracellular ligand-binding domain.70 EGFRvIII is tidylinositol (3,4,5)-triphosphate (PIP3), thereby negatively regulat- constitutively activated, has a reduced capacity of recruiting CBL ing downstream AKT signaling.84 The mTOR is a serine/threonine and shows reduced receptor internalization and degradation.71 kinase that is present in two specific complexes, mTOR complexes Overexpression of ERBB2 is found in many tumors, and the ERBB2 1 and 2 (mTORC1 and mTORC2), which act downstream or gene is found to be mutated in 8% of GBM.64 Studies in breast upstream of AKT, respectively. The mTOR pathway couples tumors have shown that the ERBB2 receptor stabilizes EGFR and nutrient and energy availability to the execution of various inhibits CBL-mediated receptor ubiquitylation by blocking biological processes including cell growth and proliferation, phosphorylation of CBL- and GRB2-binding sites in EGFR.72 survival, motility, protein synthesis and transcription.85 Interestingly, v-erbB, the transforming viral homolog of EGFR, The p85 regulatory subunit of PI3K is a substrate for the Ub has been found to lack the CBL-binding site,73 and overexpressing ligase CBL-B.86 However, when Cbl-b is knocked out in mouse T v-erbB from the S100b promoter has been shown to induce cells, ubiquitylation of p85 is not abrogated. Instead, CBL-B loss led oligodendrogliomas in mice.74 to enhanced ubiquitylation of PTEN upon T-cell receptor

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4713

RTK

P P

P P P Ub P CBL CBL DUB

Ub

P P Ub CBL ERK P P AKT mTORC1 P P CBL LYSOSOME

GSK3B PROTEASOMAL DEGRADATION

Ub Ub S62 Ub P MYC Ub PP2A P MYC FBW7 Ub MDM2 Ub Ub Ub P UBE4B Ub P p53 p53 T58 MYC MDM2 PIRH2 MDM2 CDKs MDM2 MDM4

Ub Ub DUB Ub P Ub Ub MYC p53 MDM4 Ub Ub MAD P Ub MDM2 MXI1 ATM

MNT P MYC MAX p53 p53 E-box Figure 2. A schematic representation of ubiquitin-mediated protein degradation in the key signaling pathways involved in brain tumor development and progression. RTK signaling pathways modulate MYC stability via phosphorylation and dephosphorylation events required for substrate recognition and ubiquitylation by the E3 ligase FBW7. RTK internalization and lysosomal degradation are mediated by E3 ligase CBL. Alternatively, RTKs can be recirculated to the cell surface via early endosomes. In the nucleus, degradation of p53 is regulated by mono- and polyubiquitylation carried out by various ubiquitin ligases (not shown). The monoubiquitylated form of p53 is exported to the cytosol, where further polyubiquitylation targets the protein for proteasomal degradation. Phosphorylation of p53 by stress-induced kinases such as ATM disrupts ubiquitylation mediated by MDM2. The activities of both p53 and MYC factors are regulated by shuttling in and out of the nucleus, and proteasomal targeting could take place both within the nucleus and in the cytosol. In the figure, ubiquitin ligases are shown as gray ovals. stimulation.87 Recently, SCFFBXL2 has further been shown to target PTEN.22 mTOR and especially the mTORC1/2 complexes are the p85 for proteasomal degradation.21 In GBMs, mutations have been essential brain tumor pathways.92 In response to growth factor found in PIK3R1 and PIK3CA genes that encode regulatory and stimulation, SCFb-TrCP also regulates mTOR activity through the catalytic subunits of PI3K.64 targeted degradation of DEPTOR (DEP domain containing mTOR- The HECT-domain protein NEDD4-1 can regulate PTEN by interacting protein), an inhibitor of both mTORC1 and mTORC2.93 mono- and polyubiquitylation.88 Surprisingly, NEDD4-1 is not required for PTEN degradation, as PTEN stability is unaltered in Nedd4-1 knockout mice.89 Furthermore, a recent study Transcriptional regulators demonstrates that WWP2, a member of the NEDD4-like protein MYC proteins. The MYC family of proteins consists of three family, interacts with PTEN and stimulates Ub-dependent members— MYC, MYCL and MYCN—that are important regulators degradation of PTEN.90 Loss of heterozygosity on chromosome of transcription. MYC proteins are essential for normal brain 10 is a hallmark of many types of tumors including GBMs. PTEN, development and specific disruption of Mycn in nestin-positive being situated on this chromosome, is frequently altered in brain neural stem, and precursor cells (NSCs) present a severely tumor cells.91 diminished forebrain and hindbrain.94 FBW7 binds a specific phosphodegron in mTOR, promoting its MYC proteins are regulated by different post-translational ubiquitylation and proteasomal degradation.22 This targeting by modifications. At least five different E3 Ub ligases, FBW7, beta- FBW7 leads to breast cancer suppression in cooperation with TrCP, transient receptor potential cation channel, subfamily C,

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721 Oncoprotein stabilization S-M Hede et al 4714 member 4 associated protein (TRUSS), S-phase kinase-associated The MDM2 protein can either homo- or heterodimerize with the protein 2, E3 Ub protein ligase (SKP2) and HECT, UBA and WWE homologous RING finger protein MDM4 (also known as MDMX) domains containing 1, E3 Ub protein ligase (HECTH9/HUWE1) have (Figure 2). MDM4 binds p53; however, it has no intrinsic Ub ligase been shown to regulate MYC.95 Whereas FBW7 and TRUSS act as activity.121 The MDM2/p53 interaction and subsequent ubiquitylation negative regulators of MYC,96–98 Beta-TrCP positively regulates is inhibited by various post-translational modifications including MYC protein stability.99 Interestingly, several other Ub ligases, phosphorylation, acetylation, neddylation and sumoylation.122 including SKP2,100,101 HECTH9/HUWE1102,103 and F-box protein 28 MDM2 can either monoubiquitylate p53, facilitating transport to (FBXO28),104 regulate MYC protein function through non- the cytoplasm or cooperate with MDM4 and other Ub ligases to proteolytic ubiquitylation. polyubiquitylate, and thereby target p53 for degradation by the Regulation of MYC activity and stability is a tightly controlled proteasome.123 Several p53-targeting Ub ligases with process. ERK or CDKs must first phosphorylate MYC on residue complementing functions have been discovered lately, although S62.105 GSK3b needs this priming phosphorylation in order to MDM2 seems to be the main factor regulating p53 levels under phosphorylate T58,96 which can be recognized by FBW7 and normal conditions.124 For example, the E3 ligase PIRH2 (p53-induced degraded first after S62 is dephosphorylated by combined actions protein with RING-H2 domain) targets phosphorylated p53 during of PIN1 prolyl isomerase and the PP2A phosphatase106 (see also DNA damage125 (Figure 2). UBE4B is another Ub ligase that interacts Figure 2). However, FBW7-dependent MYC ubiquitylation appears with both MDM2 and p53. UBE4B is found to be amplified and to be more complex, and MYC stability is also controlled by other overexpressed in brain tumors, and its expression is inversely factors (reviewed in Thomas and Tansley95). correlated to levels of p53.126 The UPS system also controls cellular reprogramming.107 Whereas mutations and loss of heterozygosity of p53 are Depletion of FBW7 results in enhanced reprogramming commonly found in human secondary GBMs, the primary GBM frequency, whereas inactivation of the DUB PSMD14 abrogates more often displays upstream inactivation of the p53 pathway the reprogramming process. The effect of FBW7 loss is designated through amplification and overexpression of MDM2 or deletion of to the stabilization of MYC that is involved in induced pluripotent Ink4a/Arf.69 Recent results from the The Cancer Genome Atlas stem cell generation.108 showed that altered p53 signaling was found in 87% of GBM, with The MYC proteins MYC and MYCN are commonly amplified or amplification of MDM2 in 14% and inactivation of alternate reading overexpressed in brain tumors (for details see the study by frame (ARF) of the CDKN2A gene by deletion or mutation in 49% of Swartling109). Focal high-level amplifications of MYC or MYCN are the cases.64 p53 status is an important risk factor in SHH in most cases mutually exclusive110 and can occur in both GBM64 medulloblastoma subtypes despite the fact that the majority of and medulloblastoma.111 Interestingly, mutations in the histone human medulloblastoma, including Groups 3 and 4, lack p53 H3 gene, H3F3A, are commonly found in pediatric GBMs and cause mutations.127 MDM2canbeinvolvedinregulatingp53levelsin profound upregulation of MYCN.112 MYC or MYCN is further these tumors, as the MDM2 protein is often overexpressed amplified in almost half of all brain cancers categorized as predominantly in medulloblastoma cells where the p53 gene is not malignant glioma with PNET-like morphologies.113 mutated.128 MYCN is downstream of the SHH pathway and is essential for driving SHH-driven brain tumors in various mouse models Cell cycle proteins. The intricate interplay between different E3s is of medulloblastoma. MYCN overexpression can, together of major importance for the timely activation/inactivation of with p53 loss, further drive SHH-dependent medulloblastoma various CDK regulatory proteins (for example, CDK inhibitors and from GPCs following transplantation to the brain.114 By contrast, ). SCF ligases target numerous cell cycle regulatory proteins overexpressed stabilized forms of its sibling MYC for degradation. SCFSKP2 recognizes p27 phosphorylated by Cyclin lead to development and progression of SHH-independent E-CDK2, an interaction which also depends on the co-factor aggressive large cell/anaplastic Group 3 medulloblastoma114,115 CKS1.129 Degradation of p27 results in elevated Cyclin E-CDK2 (Table 1). activity, entry into the S-phase and initiation of DNA replication. As We have recently shown that MYCN can drive aggressive large CDK activity rises in G1, the RB pocket proteins (RB, p107 and cell/anaplastic medulloblastoma when overexpressed from the p130) are inactivated by phosphorylation and E2F factors are Glutamate transporter 1 promoter in an inducible transgenic released. Several different Ub ligases target for proteasomal model.116 We further showed that MYCN carrying a stabilizing degradation, including SKP2 at the end of the S-phase130 and APC/ FBW7 degron mutation (T58A) was necessary for brain tumor C in the prometaphase.131 The APC/C ligase controls both positive generation from NSCs117 (Table 1). By transducing forebrain and negative regulators of the cell cycle through association with NSCs with MYCN-T58A, we could mimic malignant glioma its co-activator subunits, CDC20 and CDH1, which determine with PNET-like morphologies113 or pediatric GBMs that are substrate specificity.132 These differences in substrate specificity shown to depend on high levels of MYCN.112 Interestingly, have important roles during neurogenesis, (reviewed by Puram depending on the timing of MYCN induction, both and Bonni133), where, for example, APC/C together with CDH1 SHH-dependent- and -independent medulloblastoma types target ID proteins, regulating cell cycle exit.134 formed from cerebellar NSCs despite using the same stabilized CDKs are often amplified or overexpressed in both glioma and MYCN oncoprotein. medulloblastoma (for a more detailed review on this, see Vlachostergios et al.135). The CDK inhibitor p27 is downregulated p53. The transcription factor p53 is a tightly regulated sensor of and associated with increased glioma grade,136 and SKP2 (that cellular stress and its activation might result in cell cycle arrest, controls p27 degradation) is found at elevated levels in high-grade apoptosis, senescence, DNA repair, altered metabolism or glioma. In addition to FBW7, the Ub ligase PARK2 also targets autophagy.118 Cyclin E for degradation and is commonly found to be mutated in Under normal conditions, protein levels of p53 are held low by GBMs with increased Cyclin E stability137 (Table 1). proteasomal degradation, promoted in part through continuous Almost 80% of GBM tumors display alterations in the RB targeting by the RING finger E3 Ub ligase MDM2.23 Specifically, pathway. The RB gene itself is found to be mutated or deleted in the transcription of MDM2 is upregulated by p53, creating B10% of the cases, whereas CDKN2A and CDKN2B are deleted or a feedback loop where MDM2 targets both p53 and itself mutated in 52 and 47% of GBMs, respectively.64 RB interacts with for proteasomal degradation.119 MDM2 also blocks the APC/C that controls genomic stability by regulating proteins transactivating activity of p53, preventing transcriptional controlling the mitotic spindle formation checkpoint. Several APC/ activation of p53 target genes.120 C targets, including CDC20, polo-like kinase 1, Aurora kinases,

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4715 SKP2, UBE2C and survivin are overexpressed in brain tumors.138 increased protein degradation.156 Surprisingly, in some cancer cells EMI1 is a transcriptional target of E2F1 that inhibits APC/CCdh1 this process is not dependent on GSK-3b phosphorylation that function. Overexpression of EMI1 accelerates S-phase entry and normally regulates b-catenin protein stability. leads to chromosomal instability.139 EMI1 is found to be overexpressed in ependymomas and high-grade gliomas. Loss PI3K, mTOR and MYC proteins. MYCN is indirectly stabilized by the of RB leads to dysregulation of EMI1 and genomic instability in activation of PI3K.157 Blockade of PI3K leads to decreased MYCN human tumors, including oligodendrogliomas.140 EMI1 is further protein levels in neuroblastoma—a childhood tumor with frequent regulated by beta-TrCP and fibroblasts from mice lacking beta- MYCN amplifications.158 mTOR inhibitors are also effective in TrCP show decreased genetic stability and mitotic progression in suppressing MYCN in neuroblastoma159 or inhibiting MYC in part owing to the accumulation of EMI1.141,142 murine medulloblastoma.115 Furthermore, a dual inhibition of PI3K and mTOR shows emergent efficacy when targeting human glioma NFkB. NFkB signaling controls diverse physiological functions cells.160 including cell growth and survival, differentiation, development, Stabilization of MYCN by Aurora A kinase is controlled by FBW7 in immunity and inflammation.143 In unstimulated cells, NFkB neuroblastoma,161 and MYCN protein stabilization is found in a large proteins are generally kept inactive through binding to proteins set of patients who lack MYCN amplifications.15 Small Aurora A known as inhibitors of NFkB (IkBs).144 kinase inhibitors can disrupt the binding of Aurora-A to MYCN and The NFkB pathway is subjected to tight post-translational allow FBW7 to recognize and promote the degradation of MYCN.162 regulation controlled by protein kinases, DUBs145 andUbligases. Therapeutic MYCN depletion totally cures tumors in transgenic Phosphorylation of IkBa by IKK targets this inhibitor for ubiquitylation mouse models of brain tumors,116 and the depletion of all MYC and proteasomal degradation by the SCFb-TrCP Ub ligase, allowing the proteins also cures tumors driven by other oncogenes,163 NFkB protein RelA to translocate to the nucleus and activate gene highlighting MYC proteins as indisputable targets for tumor expression. SCFb-TrCP also contributes to NFkB pathway activation by treatment.164 promoting the formation of specific NFkB protein complexes in the nucleus through ubiquitylation and partial proteolysis of IkBs, such as Targeting UPS components. Inhibition of proteasomal activity p105 and p100. Furthermore, we and others have recently using reversible (Bortezomib) or irreversible (Carfilzomib) protease demonstrated that the SCFFBW7 Ub ligase targets p100 for inhibitors has remarkable efficiency against multiple mye- degradationinaGSK3b-dependent manner.146–148 In this context, loma165,166 but also demonstrates potency for the treatment of NFkB pathway activity is modulated by both SCFFBW7 and SCFb-TrCP. different types of lymphomas and is currently undergoing clinical The IkBa gene NFKB1A is often heterozygously deleted in trials for the treatment of many other malignancies, including nonclassical subtypes of GBM patients,149 suggesting an active role pediatric and adult brain tumors.167,168 for the NFkB pathway in these brain tumors. NFkB activity is higher in Proteasomal inhibitors induce cell cycle arrest and apoptosis. GBMs as compared with the normal brain;150 however, the However, the molecular mechanism(s) is still not clear and various mechanism of NFkB activation in GBMs is largely undefined factors seem to be important in different cell types. Treatment perhaps because of the large heterogeneity of these tumors. with proteasomal inhibitors can shift cellular homeostasis towards However, NFkB could putatively be activated by cytokines such as apoptosis by causing an accumulation of pro-apoptotic proteins TNF-alpha or interleukins that are secreted by activated tumor- such as BID and BAX.169,170 Proteasomal inhibitors can also reduce associated macrophages.151 The regulation of NFkB by the UPS is the expression of anti-apoptotic proteins of the BCL2 or inhibitor further discussed below. of apoptosis families possibly through the inactivation of NFkB transcriptional activity following stabilization of the IkB. In medulloblastoma, Bortezomib has been reported to trigger TARGETING UPS SUBSTRATES OR THE UPS ITSELF IN FUTURE reactive oxidative stress (ROS)-associated apoptosis by stabilizing 171 CANCER THERAPIES the pro-apoptotic protein NOXA. Malignant high-grade brain tumors have a dismal prognosis, and However, as proteasomal inhibitors work non-selectively resistance-associated mechanisms to chemotherapies often pre- blocking the degradation of all proteins passing through the clude the complete eradication of cancer cells resulting in tumor 26S proteasomal machinery, more specific drugs are needed relapse. With this follows the challenge of developing novel (Figure 3). In fact, despite a strong scientific rationale for using Bortezomib as an additional therapy in patients with recurrent therapeutic strategies targeting the key proteins contributing to 167 tumor cell resistance and/or exploring new classes of drug targets. GBM, this combination is clinically ineffective. Therefore, Recently, with the discovery of the proteasome inhibitor Bortezo- targeting the enzymes regulating the ubiquitylation process is mib (PS341/Velcade), the UPS has matured as an important drug an alternative and more specific approach, possibly also with less discovery arena with Ub enzymes as the prime candidate drug unwanted side effects (Figure 3). Inhibitors of E1 and E2 enzymes targets.152 As outlined above, many proteins directly influencing have been reported; however, their potential clinical efficiency brain tumor development and progression are substrates of the remains to be demonstrated. In addition, targeting E1 or E2 UPS. We will now exemplify how such substrates or various enzymes could also result in undesirable effects on normal cells, as components of the UPS could be targeted in brain tumors. they are expected to inhibit most or many E3 ligases. Targeting the E1-activating enzyme (NAE-E1) for the Ub-like protein NEDD8 is a more specific approach because the primary targets for Targeting substrates of the UPS neddylation appears to be CRLs. Neddylation of the cullin Developmental pathways. Long-term treatment with the smooth- component is critical for CRL activity, and inhibitors of NAE-E1 ened inhibitor cyclopamine results in complete disruption of could thus preferentially block CRLs. Indeed, MLN4924—a small glioma stem cell populations. Besides inhibiting smoothened, there molecule inhibitor of the NAE-E1 enzyme—has been developed are now ways to target GLI proteins downstream of SHH and shows promise as an anticancer drug in mouse models and signaling.153 Of the GLIs, GLI2 is supposedly the most important preclinical studies.172,173 therapeutic target in medulloblastoma.154 It is not completely clear how GLI2 stabilization is regulated in tumors. Interestingly, arsenic Targeting specific E3 Ub ligases directly. Higher specificity is treatment has been effective in targeting GLI2 in SHH-dependent achieved if the activity of single E3s or DUBs is blocked. These tumors, as it destabilizes GLI2 and prevents ciliary accumulation.155 enzymes are particularly attractive drug targets, as they are Inhibition of b-catenin by methylseleninic acid is also a result of responsible for mediating ubiquitylation/deubiquitylation of

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721 Oncoprotein stabilization S-M Hede et al 4716 DUB Inhibitors E3 Ligase Inhibitors SKP1 Ub E1 Ub Inhibitors SKP2 Ub Ub p27 p53 MDM2 Ub Substrate Ub Ub E1 E2

ATP CUL1 Ub ATP

NEDD8 Substrate Interaction Neddylation ROC1 E2 Ub Inhibitors Inhibitors Ub Ub 26S Proteasome Ub Ub

E2 Inhibitors

Bortezomib Proteasome Carfilzomib Inhibitors Figure 3. Various ways to target the UPS in brain tumors. Apart from targeting E1 and E2 enzymes directly, there are ways to target the E1- activating enzyme for the ubiquitin-like protein NEDD8, which will specifically inhibit the ubiquitin–ligase activity of Cullin–Ring ubiquitin ligases (CRLs) such as SKP2 as depicted. Even more specific ways to target UPS are by targeting specific DUBs or E3 ligases, for example, by using inhibitors directed against SKP2 or by substrate interaction inhibitors, such as nutlins, that interferes with the p53–MDM2 interaction. A broader therapeutic approach involves using proteasome inhibitors that are currently evaluated in various brain tumor therapies.

protein substrates frequently dysregulated in tumor cells. In mechanism of p53 inactivation, alternative approaches are particular, many E3s/DUBs qualify as drug targets because they are warranted. Prima-1 is one example of a small molecule that overexpressed in human tumors and are often associated with the causes a conformational change in mutated p53, restoring the suppression of multiple tumor-suppressor proteins. Although the wild-type-like conformation and inducing an apoptotic development of DUB inhibitors is still in its infancy, several response.177 compounds are under development.152 Multiple strategies can be envisioned for targeting E3 Ub SKP2. Given that SKP2 is overexpressed in a large number of ligases, including development of small molecules that impair malignancies and, as outlined above, targets preferentially the binding between the E3 ligase and the E2 enzyme, or tumor-suppressor proteins important for cell division control, between the E3 and its specific target substrate(s). In principle, including p27, p21, p57 and p130, makes it an appealing drug compounds that induce a conformational change in the E3 could target.178 The oncogenic function of SKP2 is underscored by the attenuate Ub ligase stability and/or interfere with E3 activity. inverse association with p27 protein level in medulloblastoma179 Nucleic acids (for example, micro RNA, small interfering RNA) that and glioma.136 A schematic overview of SKP2 and its potential reduce the expression of the E3 ligase, or antibodies/peptides substrates in brain tumors is presented in Figure 4. SKP2 mRNA that interfere with E3 subcellular localization or activity, levels are elevated both in medulloblastoma and GBM as represent other possibilities to inhibit oncogenic E3s. Finally, it compared with the normal brain (Figure 4), indicating SKP2 as should be emphasized that there is tight interplay between a promising drug target in these types of tumors. Small molecule protein ubiquitylation and protein phosphorylation, and inhibitors of SCFSKP2 have been discovered, including a understanding how these systems cooperate in controlling the compound that prevents the binding of SKP2 to the core ligase, stability and activity of proteins deregulated in cancer cells are triggering the accumulation of p27 and cell cycle arrest and can important areas of future research. apparently even overcome Bortezomib resistance.180 Recently, inhibitors that selectively target the interaction between SKP2 MDM2. MDM2, the major regulator of p53 protein, has strong and CKS1 as well as its substrate p27 have also been identified181 clinical relevance in brain tumors with a functional p53 gene. and could thus be useful for treating tumors with deregulation of Several compounds have been developed aiming to restore p53 the SKP2–p27 axis. Another report shows how a novel SKP2- activity in tumors by interfering with the p53–MDM2 interaction. specific inhibitor directly binds the F box motif of SKP2,182 Nutlins are one example of low molecular weight compounds that leading to an effective block of SKP2–SKP1 interactions and target the p53-binding pocket of MDM2. By preventing the suppression of SKP2 ligase activity. This novel SKP2 inhibitor interaction of MDM2 with p53, the levels of p53 are increased, effectively inhibits tumor cell proliferation, survival and glycolysis leading to cell cycle arrest and apoptosis in tumor cells with wild- and is involved in triggering p53-independent senescence in type p53.174 An alternative approach involves targeting MDM4 to various animal cancer models. achieve destabilization of MDM2 and induction of a p53 response.175 Efforts have also been made to develop molecules Beta-TrCP. As outlined above, SCFb-TrCP triggers the ubiquityla- directed towards p53. The compound RITA induces apoptosis in tion of several different proteins involved in brain tumor tumor cells by binding the N-terminal domain of p53, which development and/or progression, including GLI2, b-catenin inhibits its interaction with MDM2.176 as well as the NFkB inhibitor, IkBa183 (Figure 4). Thus, targeting However, these drugs will only induce a p53 response in tumor beta-TrCP might be a promising approach in GBM suppression. cells expressing wild-type p53 protein. Depending on the Indeed, an inhibitor that suppresses IkBa ubiquitylation, but not

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4717 SCF ubiquitin ligases Substrates important for Expression of SCF ligases brain tumors in brain tumors

SKP1 p27 Ub SKP2 Ub Ub p21 ATP PP Ub CUL1 Substrate MYC ROC1 E2 Ub Cyclin D1

SKP1 Ub GLI2 bTrCP Ub Ub Ub β-catenin ATP P P

E1 CUL1 Substrate Ub ikBα

ROC1 E2 MYC Ub

MYC SKP1 Ub mTOR FBW7 Ub ATP Ub PP NFkB2 Ub Cyclin E CUL1 Substrate NOTCH1 ROC1 E2 Aurora A Ub

Figure 4. Given their critical functions in regulating the key regulatory proteins, SCF ubiquitin ligases contribute to oncogenesis. (a) SCF ligases trigger ubiquitin-mediated proteolysis of various proteins involved in brain tumorigenesis (red ovals); however, ubiquitylation of others leads to their stabilization (green ovals). Sometimes, the effect is context-dependent (yellow ovals). Both oncoproteins and tumor suppressors are targets of different SCF ligases. Phosphorylation of short conserved sequences on protein substrates (phosphodegrons) allows the F-box protein subunit (black boxes) in SCF ligases to recognize many different protein substrates in a spatial and temporal manner. (b) Comparing gene expression of SKP2, beta-TrCP (BTRC) and FBW7 (FBXW7) between human normal and tumorous fore- and hindbrain, respectively. Left panel: raw expression data for 81 GBM samples195 and 9 forebrain (cerebral cortex) samples196 were preprocessed together using the RMA algorithm197 implementation in the ExpressionFileCreator module on the GenePattern analysis platform.198 Distributions of SKP2 (top), BTRC (middle) and FBXW7 (bottom) expression values in normal and tumor sets, respectively, are depicted as box-and-whisker-plots, with the median of expression represented by horizontal lines inside each box, lower and upper box borders, indicating 25th and 75th percentiles, respectively—regions between whiskers including all non-outlier values and dots representing outliers. Right panel: raw expression data for 62 medulloblastoma samples13 and 9 hindbrain (cerebellum) samples196 were preprocessed and illustrated as above. Po0.01 for all six comparisons using two-tailed Welch’s t-test statistics. Figures were generated in MATLAB.

phosphorylation, has been identified.184 This could prove NOTCH1 could also enforce cell death by apoptosis.188,189 In fact, promising in restoring IkBa levels, as mutated alleles of NFKB1A loss of FBW7 has been shown to drive normal brain cells into were rarely found in the heterozygously NFKB1A-deleted GBMs.149 apoptosis.190,191 How FBW7-deficient tumor cells evade apoptosis Interestingly, NFKB1A and EGFR amplifications were mutually is still unclear; however, one possibility is that the accumulation of exclusive, suggesting that these events connect in a similar FBW7 substrates with pro-survival functions (for example, MCL1 oncogenic pathway.149 Judging from beta-TrCP mRNA levels, and NFkB2) protects cancer cells from cell death. Indeed, there is SCFb-TrCP is suppressed in GBM patients (Figure 4). Elevation of an intimate association between inactivation of the tumor- SCFb-TrCP expression is instead observed in medulloblastoma, suppressor FBW7 and drug resistance.192 However, whereas where NFKB1A deletions have only been found in Group 4 FBW7-deficient cells are resistant to some antitumor agents, tumors42 (Figure 4). This tumor-specific difference is interesting FBW7 inactivation might sensitize tumor cells to other drugs.193 and could suggest that SCFb-TrCP controls the stability of distinct Interestingly, in chronic myelogenous leukemia-initiating cells,194 oncoproteins in one brain tumor type as compared with another. a sudden FBW7 loss restrains tumorigenesis. There, FBW7 suppression increases the stabilization of MYC, thereby FBW7. FBW7 is an important oncoprotein suppressor and is promoting p53-dependent apoptosis.188 In principle, these frequently inactivated by mutation, deletion and promoter results suggest that specific FBW7 inhibitors could potentially be hypermethylation in multiple malignancies.185 However, the role used to force non-dividing cancer cells to proliferate, thus of FBW7 in brain tumorigenesis is not fully understood. FBW7 is increasing their sensitivity to conventional therapeutic drugs. found to be downregulated in human gliomas186 (Figure 4). Loss Overall, FBW7 levels are low in both MB and GBM (Figure 4) as of FBW7 in p53/PTEN-deficient NSCs can further promote glioma compared with the normal brain, suggesting that FBW7 expres- formation by MYC stabilization187 (Table 1). However, abrupt sion is suppressed (at least to some extent) during development accumulations of FBW7 substrates such as MYC, c-JUN and or progression of specific types of tumors. Whether targeting

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721 Oncoprotein stabilization S-M Hede et al 4718 FBW7-deficient brain tumors with drugs directed towards specific 11 Johnson RL, Rothman AL, Xie J, Goodrich LV, Bare JW, Bonifas JM et al. Human FBW7 substrates could open up new therapeutic windows homolog of patched, a candidate gene for the basal cell nevus syndrome. remains to be shown. Science 1996; 272: 1668–1671. 12 Thomas WD, Chen J, Gao YR, Cheung B, Koach J, Sekyere E et al. Patched1 deletion increases N-Myc protein stability as a mechanism of medulloblastoma CONCLUSIONS AND FUTURE PERSPECTIVES initiation and progression. Oncogene 2009; 28: 1605–1615. UPS has important functions in normal brain development 13 Kool M, Koster J, Bunt J, Hasselt NE, Lakeman A, van Sluis P et al. Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, controlling cell fate and specification. The accumulation of reports pathway signatures and clinicopathological features. PLoS One 2008; 3:e3088. describing dysregulated UPS components in cancer suggests that 14 Cho YJ, Tsherniak A, Tamayo P, Santagata S, Ligon A, Greulich H et al. Integrative increased protein stabilization of oncoproteins or destabilization genomic analysis of medulloblastoma identifies a molecular subgroup that and degradation of tumor-suppressor proteins is important for drives poor clinical outcome. J Clin Oncol 2011; 29: 1424–1430. brain tumor development. Degradation of stabilized oncoproteins 15 Valentijn LJ, Koster J, Haneveld F, Aissa RA, van Sluis P, Broekmans ME et al. or restoration of destabilized suppressors could prove to be Functional MYCN signature predicts outcome of neuroblastoma irrespective of effective in inhibiting tumor cells. Thus, the UPS regulates many MYCN amplification. Proc Natl Acad Sci USA. 2012; 109: 19190–19195. substrates that partake in brain cancer and is an unquestionable 16 Chen J, McKay RM, Parada LF. Malignant glioma: lessons from genomics, mouse therapeutic target for brain tumor treatment. Notably, brain models, and stem cells. Cell 2012; 149: 36–47. tumors have recently been divided into specific subgroups 17 Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT, Konermann C et al. depending on their genetic profiles and their molecular biology, Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 2012; 22: 425–437. implying that individual tumors depend on distinct sets of cancer 18 Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD et al. Integrated proteins. We have described that the most common types of genomic analysis identifies clinically relevant subtypes of glioblastoma char- malignant brain tumors including medulloblastoma and glioma acterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010; and their defined subgroups have distinct amplifications or 17: 98–110. deletions in well-known UPS substrates. In summary, it is clear 19 Peschard P, Park M. Escape from Cbl-mediated downregulation: a recurrent that the UPS has an important role in brain tumorigenesis, and theme for oncogenic deregulation of receptor tyrosine kinases. Cancer Cell 2003; specific targeting of this system shows promise for treating these 3: 519–523. devastating brain tumors. 20 Fang D, Wang HY, Fang N, Altman Y, Elly C, Liu YC. Cbl-b, a RING-type E3 ubiquitin ligase, targets phosphatidylinositol 3-kinase for ubiquitination in T cells. J Biol Chem 2001; 276: 4872–4878. CONFLICT OF INTEREST 21 Kuchay S, Duan S, Schenkein E, Peschiaroli A, Saraf A, Florens L et al. FBXL2- and PTPL1-mediated degradation of p110-free p85beta regulatory subunit controls The authors declare no conflict of interest. the PI(3)K signalling cascade. Nat Cell Biol 2013; 15: 472–480. 22 Mao JH, Kim IJ, Wu D, Climent J, Kang HC, DelRosario R et al. FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression. Science ACKNOWLEDGEMENTS 2008; 321: 1499–1502. This work was supported by research from the Swedish Childhood Cancer 23 Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of Foundation (FJS, OS), the Swedish Cancer Society (FJS, OS), the Swedish Research p53. Nature 1997; 387: 296–299. Council (FJS, OS), the Swedish Society of Medicine (FJS), the Swedish Brain 24 Hershko A, Ciechanover A, Heller H, Haas AL, Rose IA. Proposed role of ATP in Foundation (FJS), Åke Wibergs stiftelse (FJS), Åke Olssons Stiftelse (OS), Lions protein breakdown: conjugation of protein with multiple chains of the polypep- Cancerforskningsfond (FJS), Stiftelsen Lars Hiertas Minne (FJS), Radiumhemmets tide of ATP-dependent proteolysis. Proc Natl Acad Sci USA 1980; 77: 1783–1786. Forskningsfonder (OS), Karolinska Institute Foundations (OS) and the Association for 25 Ciehanover A, Hod Y, Hershko A. A heat-stable polypeptide component of an International Cancer Research (FJS). We apologize to authors not cited due to space ATP-dependent proteolytic system from reticulocytes. Biochem Biophys Res restrictions in this review. Commun 1978; 81: 1100–1105. 26 Xu P, Duong DM, Seyfried NT, Cheng D, Xie Y, Robert J et al. Quantitative proteomics reveals the function of unconventional ubiquitin chains in protea- REFERENCES somal degradation. Cell 2009; 137: 133–145. 1 Frange P, Alapetite C, Gaboriaud G, Bours D, Zucker JM, Zerah M et al. From 27 Chen ZJ, Sun LJ. Nonproteolytic functions of ubiquitin in cell signaling. Mol Cell childhood to adulthood: long-term outcome of medulloblastoma patients. The 2009; 33: 275–286. Institut Curie experience (1980-2000). J Neurooncol 2009; 95: 271–279. 28 Clague MJ, Coulson JM, Urbe S. Cellular functions of the DUBs. J Cell Sci 2012; 2 Huse JT, Holland EC. Targeting brain cancer: advances in the molecular 125(Pt 2): 277–286. pathology of malignant glioma and medulloblastoma. Nat Rev Cancer 2010; 10: 29 Weissman AM. Themes and variations on ubiquitylation. Nat Rev Mol Cell Biol 319–331. 2001; 2: 169–178. 3 Hershko A, Ciechanover A. The ubiquitin system. Annu Rev Biochem. 1998; 67: 30 Li W, Bengtson MH, Ulbrich A, Matsuda A, Reddy VA, Orth A et al. Genome-wide 425–479. and functional annotation of human E3 ubiquitin ligases identifies MULAN, a 4 Driscoll J, Goldberg AL. The proteasome (multicatalytic protease) is a component mitochondrial E3 that regulates the organelle’s dynamics and signaling. PLoS of the 1500-kDa proteolytic complex which degrades ubiquitin-conjugated One 2008; 3: e1487. proteins. J Biol Chem 1990; 265: 4789–4792. 31 Metzger MB, Hristova VA, Weissman AM. HECT and RING finger families of E3 5 Northcott PA, Jones DT, Kool M, Robinson GW, Gilbertson RJ, Cho YJ et al. ubiquitin ligases at a glance. J Cell Sci 2012; 125(Pt 3): 531–537. Medulloblastomics: the end of the beginning. Nat Rev Cancer. 2012; 12: 818–834. 32 Teixeira LK, Reed SI. Ubiquitin ligases and cell cycle control. Annu Rev Biochem 6 Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT. The axis-inducing 2013; 82:387. activity, stability, and subcellular distribution of beta-catenin is regulated in 33 Skaar JR, D’Angiolella V, Pagan JK, Pagano M. SnapShot: F Box Proteins II. Xenopus embryos by glycogen synthase kinase 3. Genes Dev 1996; 10: 1443–1454. Cell 2009; 137: 1358, e1. 7 Zurawel RH, Chiappa SA, Allen C, Raffel C. Sporadic medulloblastomas contain 34 Swartling FJ, Hede SM, Weiss WA. What underlies the diversity of brain tumors? oncogenic beta-catenin mutations. Cancer Res 1998; 58: 896–899. Cancer Metastasis Rev 2012; 32: 5–24. 8 Bhatia N, Thiyagarajan S, Elcheva I, Saleem M, Dlugosz A, Mukhtar H et al. Gli2 is 35 Lee J, Platt KA, Censullo P, Ruiz i Altaba A. Gli1 is a target of Sonic hedgehog that targeted for ubiquitination and degradation by beta-TrCP ubiquitin ligase. J Biol induces ventral neural tube development. Development 1997; 124: 2537–2552. Chem 2006; 281: 19320–19326. 36 Lee JH, Gleeson JG. The role of primary cilia in neuronal function. Neurobiol Dis 9 Northcott PA, Hielscher T, Dubuc A, Mack S, Shih D, Remke M et al. Pediatric and 2010; 38: 167–172. adult sonic hedgehog medulloblastomas are clinically and molecularly distinct. 37 Lai K, Kaspar BK, Gage FH, Schaffer DV. Sonic hedgehog regulates adult neural Acta Neuropathol 2011; 122: 231–240. progenitor proliferation in vitro and in vivo. Nat Neurosci 2003; 6: 21–27. 10 Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S, Chidambaram A 38 Wang C, Pan Y, Wang B. Suppressor of fused and Spop regulate the stability, et al. Mutations of the human homolog of Drosophila patched in the nevoid processing and function of Gli2 and Gli3 full-length activators but not their basal cell carcinoma syndrome. Cell 1996; 85: 841–851. repressors. Development 2010; 137: 2001–2009.

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4719 39 Di Marcotullio L, Ferretti E, Greco A, De Smaele E, Po A, Sico MA et al. Numb is a 66 Hede SM, Hansson I, Afink GB, Eriksson A, Nazarenko I, Andrae J et al. GFAP suppressor of Hedgehog signaling and targets Gli1 for Itch-dependent ubiqui- promoter driven transgenic expression of PDGFB in the mouse brain leads to tination. Nat cell Biol 2006; 8: 1415–1423. glioblastoma in a Trp53 null background. Glia 2009; 57: 1143–1153. 40 Wang B, Li Y. Evidence for the direct involvement of {beta}TrCP in Gli3 protein 67 Dai C, Celestino JC, Okada Y, Louis DN, Fuller GN, Holland EC. PDGF autocrine processing. Proc Natl Acad Sci USA 2006; 103: 33–38. stimulation dedifferentiates cultured astrocytes and induces oligoden- 41 Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer drogliomas and oligoastrocytomas from neural progenitors and astrocytes Res 2007; 13: 4042–4045. in vivo. Genes Dev 2001; 15: 1913–1925. 42 Northcott PA, Shih DJ, Peacock J, Garzia L, Morrissy AS, Zichner T et al. Subgroup- 68 Shih AH, Dai C, Hu X, Rosenblum MK, Koutcher JA, Holland EC. Dose-dependent specific structural variation across 1000 medulloblastoma genomes. Nature 2012; effects of platelet-derived growth factor-B on glial tumorigenesis. Cancer Res 488: 49–56. 2004; 64: 4783–4789. 43 Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E, Polakis P. Stabilization of 69 Ohgaki H, Kleihues P. Genetic pathways to primary and secondary glioblastoma. beta-catenin by genetic defects in melanoma cell lines. Science 1997; 275: Am J Pathol 2007; 170: 1445–1453. 1790–1792. 70 Frederick L, Wang XY, Eley G, James CD. Diversity and frequency of epidermal 44 Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H et al. The growth factor receptor mutations in human glioblastomas. Cancer Res 2000; 60: F-box protein beta-TrCP associates with phosphorylated beta-catenin and reg- 1383–1387. ulates its activity in the cell. Curr Biol 1999; 9: 207–210. 71 Schmidt MH, Furnari FB, Cavenee WK, Bogler O. Epidermal growth factor 45 Gibson P, Tong Y, Robinson G, Thompson MC, Currle DS, Eden C et al. Subtypes receptor signaling intensity determines intracellular protein interactions, ubi- of medulloblastoma have distinct developmental origins. Nature 2010; 468: quitination, and internalization. Proc Natl Acad Sci USA 2003; 100: 6505–6510. 1095–1099. 72 Hartman Z, Zhao H, Agazie YM. HER2 stabilizes EGFR and itself by altering 46 Huang H, Mahler-Araujo BM, Sankila A, Chimelli L, Yonekawa Y, Kleihues P et al. autophosphorylation patterns in a manner that overcomes regulatory APC mutations in sporadic medulloblastomas. Am J Pathol 2000; 156: 433–437. mechanisms and promotes proliferative and transformation signaling. Oncogene 47 Louvi A, Artavanis-Tsakonas S. Notch signaling in vertebrate neural develop- 2012; 32: 4169–4180. ment. Nat Rev Neurosci 2006; 7: 93–102. 73 Choi OR, Trainor C, Graf T, Beug H, Engel JD. A single amino acid substitution in 48 Koch U, Lehal R, Radtke F. Stem cells living with a Notch. Development 2013; 140: v-erbB confers a thermolabile phenotype to ts167 avian erythroblastosis virus- 689–704. transformed erythroid cells. Mol Cell Biol 1986; 6: 1751–1759. 49 Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell 74 Weiss WA, Burns MJ, Hackett C, Aldape K, Hill JR, Kuriyama H et al. Genetic Biol 2006; 7: 678–689. determinants of malignancy in a mouse model for oligodendroglioma. Cancer 50 Oberg C, Li J, Pauley A, Wolf E, Gurney M, Lendahl U. The Notch intracellular Res 2003; 63: 1589–1595. domain is ubiquitinated and negatively regulated by the mammalian Sel-10 75 Koochekpour S, Jeffers M, Rulong S, Taylor G, Klineberg E, Hudson EA et al. Met homolog. J Biol Chem 2001; 276: 35847–35853. and hepatocyte growth factor/scatter factor expression in human gliomas. 51 Gupta-Rossi N, Le Bail O, Gonen H, Brou C, Logeat F, Six E et al. Functional Cancer Res 1997; 57: 5391–5398. interaction between SEL-10, an F-box protein, and the nuclear form of activated 76 Yokouchi M, Kondo T, Sanjay A, Houghton A, Yoshimura A, Komiya S et al. Src- Notch1 receptor. J Biol Chem 2001; 276: 34371–34378. catalyzed phosphorylation of c-Cbl leads to the interdependent ubiquitination of 52 Fryer CJ, White JB, Jones KA. Mastermind recruits CycC:CDK8 to phosphorylate the both proteins. J Biol Chem 2001; 276: 35185–35193. Notch ICD and coordinate activation with turnover. Mol Cell 2004; 16: 509–520. 77 Zhou L, Yang H. The von Hippel-Lindau tumor suppressor protein promotes 53 Natarajan S, Li Y, Miller EE, Shih DJ, Taylor MD, Stearns TM et al. Notch1-induced c-Cbl-independent poly-ubiquitylation and degradation of the activated EGFR. brain tumor models the sonic hedgehog subgroup of human medulloblastoma. PloS One 2011; 6: e23936. Cancer Res 2013; 73: 5381–5390. 78 Massague J. TGFbeta signaling in context. Nat Rev Mol Cell Biol 2012; 13: 54 Fan X, Mikolaenko I, Elhassan I, Ni X, Wang Y, Ball D et al. Notch1 and notch2 616–630. have opposite effects on embryonal brain tumor growth. Cancer Res 2004; 64: 79 De Boeck M, ten Dijke P. Key role for ubiquitin protein modification in TGFbeta 7787–7793. signal transduction. Upsala J Med Sci 2012; 117: 153–165. 55 Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD et al. Molecular 80 Heldin CH, Landstrom M, Moustakas A. Mechanism of TGF-beta signaling to subclasses of high-grade glioma predict prognosis, delineate a pattern of disease growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr Opin Cell progression, and resemble stages in neurogenesis. Cancer Cell 2006; 9: 157–173. Biol 2009; 21: 166–176. 56 Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 81 Seoane J, Le HV, Shen L, Anderson SA, Massague J. Integration of Smad and 2010; 141: 1117–1134. forkhead pathways in the control of neuroepithelial and glioblastoma cell pro- 57 Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in phy- liferation. Cell 2004; 117: 211–223. siology and medicine. Genes Dev 2008; 22: 1276–1312. 82 Bruna A, Darken RS, Rojo F, Ocana A, Penuelas S, Arias A et al. High TGFbeta- 58 Jackson EL, Garcia-Verdugo JM, Gil-Perotin S, Roy M, Quinones-Hinojosa A, Smad activity confers poor prognosis in glioma patients and promotes cell VandenBerg S et al. PDGFR alpha-positive B cells are neural stem cells in the proliferation depending on the methylation of the PDGF-B gene. Cancer Cell adult SVZ that form glioma-like growths in response to increased PDGF sig- 2007; 11: 147–160. naling. Neuron 2006; 51: 187–199. 83 Eichhorn PJ, Rodon L, Gonzalez-Junca A, Dirac A, Gili M, Martinez-Saez E et al. 59 Menn B, Garcia-Verdugo JM, Yaschine C, Gonzalez-Perez O, Rowitch D, Alvarez- USP15 stabilizes TGF-beta receptor I and promotes oncogenesis through the Buylla A. Origin of oligodendrocytes in the subventricular zone of the adult activation of TGF-beta signaling in glioblastoma. Nat Med 2012; 18: 429–435. brain. J Neurosci 2006; 26: 7907–7918. 84 Yuan TL, Cantley LC. PI3K pathway alterations in cancer: variations on a theme. 60 Marmor MD, Yarden Y. Role of protein ubiquitylation in regulating endocytosis Oncogene 2008; 27: 5497–5510. of receptor tyrosine kinases. Oncogene 2004; 23: 2057–2070. 85 Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, 61 Mosesson Y, Shtiegman K, Katz M, Zwang Y, Vereb G, Szollosi J et al. Endocytosis diabetes and ageing. Nat Rev Mol Cell Biol 2011; 12: 21–35. of receptor tyrosine kinases is driven by monoubiquitylation, not poly- 86 Fang D, Liu YC. Proteolysis-independent regulation of PI3K by Cbl-b-mediated ubiquitylation. J Biol Chem 2003; 278: 21323–21326. ubiquitination in T cells. Nat Immunol 2001; 2: 870–875. 62 Lokker NA, Sullivan CM, Hollenbach SJ, Israel MA, Giese NA. Platelet-derived 87 Guo H, Qiao G, Ying H, Li Z, Zhao Y, Liang Y et al. E3 ubiquitin ligase Cbl-b growth factor (PDGF) autocrine signaling regulates survival and mitogenic regulates Pten via Nedd4 in T cells independently of its ubiquitin ligase activity. pathways in glioblastoma cells: evidence that the novel PDGF-C and PDGF-D Cell Rep 2012; 1: 472–482. ligands may play a role in the development of brain tumors. Cancer Res 2002; 62: 88 Wang X, Trotman LC, Koppie T, Alimonti A, Chen Z, Gao Z et al. NEDD4-1 is a 3729–3735. proto-oncogenic ubiquitin ligase for PTEN. Cell 2007; 128: 129–139. 63 Hermanson M, Funa K, Koopmann J, Maintz D, Waha A, Westermark B et al. 89 Fouladkou F, Landry T, Kawabe H, Neeb A, Lu C, Brose N et al. The ubiquitin Association of loss of heterozygosity on chromosome 17p with high platelet- ligase Nedd4-1 is dispensable for the regulation of PTEN stability and localiza- derived growth factor alpha receptor expression in human malignant gliomas. tion. Proc Natl Acad Sci USA. 2008; 105: 8585–8590. Cancer Res 1996; 56: 164–171. 90 Maddika S, Kavela S, Rani N, Palicharla VR, Pokorny JL, Sarkaria JN et al. WWP2 is 64 TCGA. Comprehensive genomic characterization defines human glioblastoma an E3 ubiquitin ligase for PTEN. Nat Cell Biol 2011; 13: 728–733. genes and core pathways. Nature 2008; 455: 1061–1068. 91 Wang SI, Puc J, Li J, Bruce JN, Cairns P, Sidransky D et al. Somatic mutations of 65 Uhrbom L, Hesselager G, Nister M, Westermark B. Induction of brain tumors in PTEN in glioblastoma multiforme. Cancer Res 1997; 57: 4183–4186. mice using a recombinant platelet-derived growth factor B-chain retrovirus. 92 Fan QW, Weiss WA. Inhibition of PI3K-Akt-mTOR signaling in glioblastoma by Cancer Res 1998; 58: 5275–5279. mTORC1/2 inhibitors. Methods Mol Biol 2012; 821: 349–359.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721 Oncoprotein stabilization S-M Hede et al 4720 93 Zhao Y, Sun Y. Targeting the mTOR-DEPTOR pathway by CRL E3 ubiquitin 119 Fang S, Jensen JP, Ludwig RL, Vousden KH, Weissman AM. Mdm2 is a RING ligases: therapeutic application. Neoplasia 2012; 14: 360–367. finger-dependent ubiquitin protein ligase for itself and p53. J Biol Chem 2000; 94 Knoepfler PS, Cheng PF, Eisenman RN. N-myc is essential during neurogenesis 275: 8945–8951. for the rapid expansion of progenitor cell populations and the inhibition of 120 Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm-2 oncogene neuronal differentiation. Genes Dev 2002; 16: 2699–2712. product forms a complex with the p53 protein and inhibits p53-mediated 95 Thomas LR, Tansey WP. Proteolytic control of the oncoprotein transcription transactivation. Cell 1992; 69: 1237–1245. factor Myc. Adv Cancer Res 2011; 110: 77–106. 121 Jackson MW, Berberich SJ. MdmX protects p53 from Mdm2-mediated degra- 96 Welcker M, Orian A, Jin J, Grim JE, Harper JW, Eisenman RN et al. The Fbw7 tumor dation. Mol Cell Biol 2000; 20: 1001–1007. suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent 122 Kruse JP, Gu W. Modes of p53 regulation. Cell 2009; 137: 609–622. c-Myc protein degradation. Proc Natl Acad Sci USA 2004; 101: 9085–9090. 123 Wang X, Jiang X. Mdm2 and MdmX partner to regulate p53. FEBS Lett 2012; 586: 97 Yada M, Hatakeyama S, Kamura T, Nishiyama M, Tsunematsu R, Imaki H et al. 1390–1396. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box 124 Love IM, Grossman SR. It takes 15 to Tango: making sense of the many ubiquitin protein Fbw7. EMBO J 2004; 23: 2116–2125. ligases of p53. Genes Cancer 2012; 3: 249–263. 98 Choi SH, Wright JB, Gerber SA, Cole MD. Myc protein is stabilized by suppression 125 Jung YS, Qian Y, Chen X. Pirh2 RING-finger E3 ubiquitin ligase: its role in of a novel E3 ligase complex in cancer cells. Genes Dev 2010; 24: 1236–1241. tumorigenesis and cancer therapy. FEBS Lett 2012; 586: 1397–1402. 99 Popov N, Schulein C, Jaenicke LA, Eilers M. Ubiquitylation of the amino terminus 126 Wu H, Pomeroy SL, Ferreira M, Teider N, Mariani J, Nakayama KI et al. UBE4B of Myc by SCF(beta-TrCP) antagonizes SCF(Fbw7)-mediated turnover. Nat Cell promotes Hdm2-mediated degradation of the tumor suppressor p53. Nat Med Biol 2010; 12: 973–981. 2011; 17: 347–355. 100 von der Lehr N, Johansson S, Wu S, Bahram F, Castell A, Cetinkaya C et al. The 127 Zhukova N, Ramaswamy V, Remke M, Pfaff E, Shih DJ, Martin DC et al. Subgroup- F-box protein Skp2 participates in c-Myc proteosomal degradation and acts as a specific prognostic implications of TP53 mutation in medulloblastoma. J Clin cofactor for c-Myc-regulated transcription. Mol Cell 2003; 11: 1189–1200. Oncol 2013; 31: 2927–2935. 101 Kim SY, Herbst A, Tworkowski KA, Salghetti SE, Tansey WP. Skp2 regulates Myc 128 Kunkele A, De Preter K, Heukamp L, Thor T, Pajtler KW, Hartmann W et al. protein stability and activity. Mol Cell 2003; 11: 1177–1188. Pharmacological activation of the p53 pathway by nutlin-3 exerts anti-tumoral 102 Adhikary S, Marinoni F, Hock A, Hulleman E, Popov N, Beier R et al. The ubiquitin effects in medulloblastomas. Neuro Oncol 2012; 14: 859–869. ligase HectH9 regulates transcriptional activation by Myc and is essential for 129 Carrano AC, Eytan E, Hershko A, Pagano M. SKP2 is required for ubiquitin- tumor cell proliferation. Cell 2005; 123: 409–421. mediated degradation of the CDK inhibitor p27. Nat Cell Biol 1999; 1: 193–199. 103 Zhao X, Heng JI, Guardavaccaro D, Jiang R, Pagano M, Guillemot F et al. The 130 Marti A, Wirbelauer C, Scheffner M, Krek W. Interaction between ubiquitin-pro- HECT-domain ubiquitin ligase Huwe1 controls neural differentiation and pro- tein ligase SCFSKP2 and E2F-1 underlies the regulation of E2F-1 degradation. Nat liferation by destabilizing the N-Myc oncoprotein. Nat Cell Biol 2008; 10: Cell Biol 1999; 1: 14–19. 131 Peart MJ, Poyurovsky MV, Kass EM, Urist M, Verschuren EW, Summers MK et al. 643–653. APC/C(Cdc20) targets E2F1 for degradation in prometaphase. Cell Cycle 2010; 9: 104 Cepeda D, Ng HF, Sharifi HR, Mahmoudi S, Cerrato VS, Fredlund E et al. CDK- 3956–3964. mediated activation of the SCF ubiquitin ligase promotes MYC-driven tran- 132 Visintin R, Prinz S, Amon A. CDC20 and CDH1: a family of substrate-specific scription and tumourigenesis and predicts poor survival in breast cancer. EMBO activators of APC-dependent proteolysis. Science 1997; 278: 460–463. Mol Med 2013; 5: 999–1018. 133 Puram SV, Bonni A. Novel functions for the anaphase-promoting complex in 105 Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. Multiple Ras-dependent neurobiology. Sem Cell Develop Biol 2011; 22: 586–594. phosphorylation pathways regulate Myc protein stability. Genes Dev 2000; 14: 134 Lasorella A, Stegmuller J, Guardavaccaro D, Liu G, Carro MS, Rothschild G et al. 2501–2514. Degradation of Id2 by the anaphase-promoting complex couples cell cycle exit 106 Yeh E, Cunningham M, Arnold H, Chasse D, Monteith T, Ivaldi G et al. A signalling and axonal growth. Nature 2006; 442: 471–474. pathway controlling c-Myc degradation that impacts oncogenic transformation 135 Vlachostergios PJ, Voutsadakis IA, Papandreou CN. The ubiquitin-proteasome of human cells. Nat Cell Biol 2004; 6: 308–318. system in glioma cell cycle control. Cell Div 2012; 7: 18. 107 Buckley SM, Aranda-Orgilles B, Strikoudis A, Apostolou E, Loizou E, Moran-Crusio 136 Schiffer D, Cavalla P, Fiano V, Ghimenti C, Piva R. Inverse relationship K et al. Regulation of pluripotency and cellular reprogramming by the ubiquitin- between p27/Kip.1 and the F-box protein Skp2 in human astrocytic proteasome system. Cell Stem Cell 2012; 11: 783–798. gliomas by immunohistochemistry and Western blot. Neurosci Lett 2002; 328: 108 Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse 125–128. embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126: 137 Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I et al. Somatic mutations 663–676. of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other 109 Swartling FJ. Myc proteins in brain tumor development and maintenance. Ups J human malignancies. Nat Genet 2010; 42: 77–82. Med Sci 2012; 117: 122–131. 138 Penas C, Ramachandran V, Ayad NG. The APC/C Ubiquitin Ligase: From Cell 110 Aldosari N, Bigner SH, Burger PC, Becker L, Kepner JL, Friedman HS et al. MYCC Biology to Tumorigenesis. Front Oncol 2011; 1: 60. and MYCN oncogene amplification in medulloblastoma. A fluorescence in situ 139 Hsu JY, Reimann JD, Sorensen CS, Lukas J, Jackson PK. E2F-dependent accu- hybridization study on paraffin sections from the Children’s Oncology Group. mulation of hEmi1 regulates S phase entry by inhibiting APC(Cdh1). Nat Cell Biol Arch Pathol Lab Med 2002; 126: 540–544. 2002; 4: 358–366. 111 Korshunov A, Remke M, Kool M, Hielscher T, Northcott PA, Williamson D et al. 140 Lehman NL, Verschuren EW, Hsu JY, Cherry AM, Jackson PK. Overexpression of Biological and clinical heterogeneity of MYCN-amplified medulloblastoma. Acta the anaphase promoting complex/cyclosome inhibitor Emi1 leads to tetraploidy Neuropathol 2012; 123: 515–527. and genomic instability of p53-deficient cells. Cell Cycle 2006; 5: 1569–1573. 112 Bjerke L, Mackay A, Nandhabalan M, Burford A, Jury A, Popov S et al. Histone 141 Margottin-Goguet F, Hsu JY, Loktev A, Hsieh HM, Reimann JD, Jackson PK. H3.3 mutations drive pediatric glioblastoma through upregulation of MYCN. Prophase destruction of Emi1 by the SCF(betaTrCP/Slimb) ubiquitin ligase acti- Cancer Discov 2013; 3: 512–519. vates the anaphase promoting complex to allow progression beyond prome- 113 Perry A, Miller CR, Gujrati M, Scheithauer BW, Zambrano SC, Jost SC et al. taphase. Dev Cell 2003; 4: 813–826. Malignant gliomas with primitive neuroectodermal tumor-like components: a 142 Guardavaccaro D, Kudo Y, Boulaire J, Barchi M, Busino L, Donzelli M et al. Control clinicopathologic and genetic study of 53 cases. Brain Pathol 2009; 19: 81–90. of meiotic and mitotic progression by the F box protein beta-Trcp1 in vivo. Dev 114 Kawauchi D, Robinson G, Uziel T, Gibson P, Rehg J, Gao C et al. A mouse model Cell 2003; 4: 799–812. of the most aggressive subgroup of human medulloblastoma. Cancer Cell 2012; 143 Ben-Neriah Y, Karin M. Inflammation meets cancer, with NF-kappaB as the 21: 168–180. matchmaker. Nat Immunol 2011; 12: 715–723. 115 Pei Y, Moore CE, Wang J, Tewari AK, Eroshkin A, Cho YJ et al. An animal model of 144 Gilmore TD. Introduction to NF-kappaB: players, pathways, perspectives. Onco- MYC-driven medulloblastoma. Cancer Cell 2012; 21: 155–167. gene 2006; 25: 6680–6684. 116 Swartling FJ, Grimmer MR, Hackett CS, Northcott PA, Fan QW, Goldenberg DD 145 Harhaj EW, Dixit VM. Deubiquitinases in the regulation of NF-kappaB signaling. et al. Pleiotropic role for MYCN in medulloblastoma. Genes Dev 2010; 24: Cell Res 2011; 21: 22–39. 1059–1072. 146 Arabi A, Ullah K, Branca RM, Johansson J, Bandarra D, Haneklaus M et al. Pro- 117 Swartling FJ, Savov V, Persson AI, Chen J, Hackett CS, Northcott PA et al. Distinct teomic screen reveals Fbw7 as a modulator of the NF-kappaB pathway. Nat neural stem cell populations give rise to disparate brain tumors in response to Commun 2012; 3: 976. N-MYC. Cancer Cell 2012; 21: 601–613. 147 Busino L, Millman SE, Scotto L, Kyratsous CA, Basrur V, O’Connor O et al. 118 Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell Fbxw7alpha- and GSK3-mediated degradation of p100 is a pro-survival 2009; 137: 413–431. mechanism in . Nat Cell Biol 2012; 14: 375–385.

Oncogene (2014) 4709 – 4721 & 2014 Macmillan Publishers Limited Oncoprotein stabilization S-M Hede et al 4721 148 Fukushima H, Matsumoto A, Inuzuka H, Zhai B, Lau AW, Wan L et al. SCF(Fbw7) 173 Swords RT, Kelly KR, Smith PG, Garnsey JJ, Mahalingam D, Medina E et al. Inhi- modulates the NFkB signaling pathway by targeting NFkB2 for ubiquitination bition of NEDD8-activating enzyme: a novel approach for the treatment of acute and destruction. Cell Rep 2012; 1: 434–443. myeloid leukemia. Blood 2010; 115: 3796–3800. 149 Bredel M, Scholtens DM, Yadav AK, Alvarez AA, Renfrow JJ, Chandler JP et al. 174 Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z et al. In vivo NFKBIA deletion in glioblastomas. N Engl J Med 2011; 364: 627–637. activation of the p53 pathway by small-molecule antagonists of MDM2. Science 150 Wang H, Zhang W, Huang HJ, Liao WS, Fuller GN. Analysis of the activation status 2004; 303: 844–848. of Akt, NFkappaB, and Stat3 in human diffuse gliomas. Lab Invest 2004; 84: 175 Garcia D, Warr MR, Martins CP, Brown Swigart L, Passegue E, Evan GI. Validation 941–951. of MdmX as a therapeutic target for reactivating p53 in tumors. Genes Dev 2011; 151 Hussain SF, Yang D, Suki D, Aldape K, Grimm E, Heimberger AB. The role of 25: 1746–1757. human glioma-infiltrating microglia/macrophages in mediating antitumor 176 Issaeva N, Bozko P, Enge M, Protopopova M, Verhoef LG, Masucci M et al. Small immune responses. Neuro Oncol 2006; 8: 261–279. molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 152 Cohen P, Tcherpakov M. Will the ubiquitin system furnish as many drug targets function in tumors. Nat Med 2004; 10: 1321–1328. as protein kinases? Cell 2010; 143: 686–693. 177 Lambert JM, Gorzov P, Veprintsev DB, Soderqvist M, Segerback D, Bergman J 153 Lauth M, Bergstrom A, Shimokawa T, Toftgard R. Inhibition of GLI-mediated et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. transcription and tumor cell growth by small-molecule antagonists. Proc Natl Cancer Cell 2009; 15: 376–388. Acad Sci USA. 2007; 104: 8455–8460. 178 Guardavaccaro D, Pagano M. Oncogenic aberrations of cullin-dependent 154 Buczkowicz P, Ma J, Hawkins C. GLI2 is a potential therapeutic target in pediatric ubiquitin ligases. Oncogene 2004; 23: 2037–2049. medulloblastoma. J Neuropathol Exp Neurol 2011; 70: 430–437. 179 Ayrault O, Zindy F, Rehg J, Sherr CJ, Roussel MF. Two tumor suppressors, p27Kip1 155 Kim J, Lee JJ, Gardner D, Beachy PA. Arsenic antagonizes the Hedgehog pathway and patched-1, collaborate to prevent medulloblastoma. Mol Cancer Res 2009; 7: by preventing ciliary accumulation and reducing stability of the Gli2 transcrip- 33–40. tional effector. Proc Natl Acad Sci USA 2010; 107: 13432–13437. 180 Chen Q, Xie W, Kuhn DJ, Voorhees PM, Lopez-Girona A, Mendy D et al. Targeting 156 Saifo MS, Rempinski Jr DR, Rustum YM, Azrak RG. Targeting the oncogenic the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated cell-cycle arrest protein beta-catenin to enhance chemotherapy outcome against solid human and activation of autophagy. Blood 2008; 111: 4690–4699. cancers. Mol Cancer 2010; 9: 310. 181 Wu L, Grigoryan AV, Li Y, Hao B, Pagano M, Cardozo TJ. Specific small molecule 157 Kenney AM, Widlund HR, Rowitch DH. Hedgehog and PI-3 kinase signaling inhibitors of Skp2-mediated p27 degradation. Chem Biol 2012; 19: 1515–1524. converge on Nmyc1 to promote cell cycle progression in cerebellar neuronal 182 Chan CH, Morrow JK, Li CF, Gao Y, Jin G, Moten A et al. Pharmacological inac- precursors. Development 2004; 131: 217–228. tivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer 158 Chesler L, Schlieve C, Goldenberg DD, Kenney A, Kim G, McMillan A et al. Inhi- progression. Cell 2013; 154: 556–568. 183 Karin M. Nuclear factor-kappaB in cancer development and progression. Nature bition of phosphatidylinositol 3-kinase destabilizes Mycn protein and blocks 2006; 441: 431–436. malignant progression in neuroblastoma. Cancer Res 2006; 66: 8139–8146. 184 Nakajima H, Fujiwara H, Furuichi Y, Tanaka K, Shimbara N. A novel small-mole- 159 Johnsen JI, Segerstrom L, Orrego A, Elfman L, Henriksson M, Kagedal B et al. cule inhibitor of NF-kappaB signaling. Biochem Biophys Res Commun 2008; 368: Inhibitors of mammalian target of rapamycin downregulate MYCN protein 1007–1013. expression and inhibit neuroblastoma growth in vitro and in vivo. Oncogene 185 Tan Y, Sangfelt O, Spruck C. The Fbxw7/hCdc4 tumor suppressor in human 2008; 27: 2910–2922. cancer. Cancer Lett 2008; 271: 1–12. 160 Fan QW, Knight ZA, Goldenberg DD, Yu W, Mostov KE, Stokoe D et al. A dual PI3 186 Bredel M, Bredel C, Juric D, Harsh GR, Vogel H, Recht LD et al. Functional network kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell 2006; 9: analysis reveals extended gliomagenesis pathway maps and three novel MYC- 341–349. interacting genes in human gliomas. Cancer Res 2005; 65: 8679–8689. 161 Otto T, Horn S, Brockmann M, Eilers U, Schuttrumpf L, Popov N et al. Stabilization 187 Kim HS, Woolard K, Lai C, Bauer PO, Maric D, Song H et al. Gliomagenesis arising of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell from Pten- and Ink4a/Arf-deficient neural progenitor cells is mediated by the p53- 2009; 15: 67–78. Fbxw7/Cdc4 pathway, which controls c-Myc. Cancer Res 2012; 72: 6065–6075. 162 Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L et al. Small 188 Evan GI, Wyllie AH, Gilbert CS, Littlewood TD, Land H, Brooks M et al. Induction of molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in apoptosis in fibroblasts by c-myc protein. Cell 1992; 69: 119–128. childhood neuroblastoma. Cancer Cell 2013; 24: 75–89. 189 Sanchez I, Yuan J. A convoluted way to die. Neuron 2001; 29: 563–566. 163 Soucek L, Whitfield J, Martins CP, Finch AJ, Murphy DJ, Sodir NM et al. Modelling 190 Hoeck JD, Jandke A, Blake SM, Nye E, Spencer-Dene B, Brandner S et al. Fbw7 Myc inhibition as a cancer therapy. Nature 2008; 455: 679–683. controls neural stem cell differentiation and progenitor apoptosis via Notch and 164 Larsson LG, Henriksson MA. The Yin and Yang functions of the Myc oncoprotein c-Jun. Nat Neurosci 2010; 13: 1365–1372. in cancer development and as targets for therapy. Exp Cell Res 2010; 316: 191 Jandke A, Da Costa C, Sancho R, Nye E, Spencer-Dene B, Behrens A. The F-box 1429–1437. protein Fbw7 is required for cerebellar development. Dev Biol 2011; 358: 201–212. 165 Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011; 364: 1046–1060. 192 Wang Z, Fukushima H, Gao D, Inuzuka H, Wan L, Lau AW et al. The two faces of 166 Ruschak AM, Slassi M, Kay LE, Schimmer AD. Novel proteasome inhibitors to FBW7 in cancer drug resistance. Bioessays 2011; 33: 851–859. overcome bortezomib resistance. J Natl Cancer Inst 2011; 103: 1007–1017. 193 Inuzuka H, Shaik S, Onoyama I, Gao D, Tseng A, Maser RS et al. SCF(FBW7) 167 Friday BB, Anderson SK, Buckner J, Yu C, Giannini C, Geoffroy F et al. Phase II trial regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruc- of vorinostat in combination with bortezomib in recurrent glioblastoma: a north tion. Nature 2011; 471: 104–109. central cancer treatment group study. Neuro Oncol 2012; 14: 215–221. 194 Reavie L, Buckley SM, Loizou E, Takeishi S, Aranda-Orgilles B, Ndiaye-Lobry D 168 Wolff JE, Brown RE, Buryanek J, Pfister S, Vats TS, Rytting ME. Preliminary et al. Regulation of c-Myc ubiquitination controls chronic myelogenous leuke- experience with personalized and targeted therapy for pediatric brain tumors. mia initiation and progression. Cancer Cell 2013; 23: 362–375. Pediatr Blood Cancer 2012; 59: 27–33. 195 Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S et al. Neuronal and 169 Breitschopf K, Zeiher AM, Dimmeler S. Ubiquitin-mediated degradation of the glioma-derived stem cell factor induces angiogenesis within the brain. Cancer proapoptotic active form of bid. A functional consequence on apoptosis Cell 2006; 9: 287–300. induction. J Biol Chem 2000; 275: 21648–21652. 196 Roth RB, Hevezi P, Lee J, Willhite D, Lechner SM, Foster AC et al. Gene expression 170 Chang YC, Lee YS, Tejima T, Tanaka K, Omura S, Heintz NH et al. mdm2 and bax, analyses reveal molecular relationships among 20 regions of the human CNS. downstream mediators of the p53 response, are degraded by the ubiquitin- Neurogenetics 2006; 7: 67–80. proteasome pathway. Cell Growth Differ 1998; 9: 79–84. 197 Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP. Summaries of 171 Ohshima-Hosoyama S, Davare MA, Hosoyama T, Nelon LD, Keller C. Bortezomib Affymetrix GeneChip probe level data. Nucleic Acids Res 2003; 31: e15. stabilizes NOXA and triggers ROS-associated apoptosis in medulloblastoma. J 198 Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Neurooncol 2011; 105: 475–483. Genet. 2006; 38: 500–501. 172 Soucy TA, Smith PG, Milhollen MA, Berger AJ, Gavin JM, Adhikari S et al. An 199 Di K, Linskey ME, Bota DA. TRIM11 is overexpressed in high-grade gliomas and inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature promotes proliferation, invasion, migration and glial tumor growth. Oncogene 2009; 458: 732–736. 2013; 32: 5038–5047.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4709 – 4721