<<

Molecular Pathways

Notch Signaling Lucio Miele

Background MeCP2 (12). Notch binding to CSL replaces the SMRT complex with a coactivator complex including The basics of Notch signaling. Notch and receptors SKIP, MAML1 (Mastermind in ), and histone are type I membrane that regulate cell fate during acetyltransferases PCAF, GCN5, or p300 (1–3, 9), activating cell-cell contact (1, 2). Humans have four Notch receptors transcription of target . These include ‘‘Enhancer of Split’’ and five ligands: Delta-like-1, Delta-like-3, and Delta-like-4 bHLH transcription factors in Drosophila and their mammalian and Jagged-1 and Jagged-2 (3). Notch receptors are hetero- homologues HES (1), HERP (13), and HEY (14) families, dimeric proteins that are synthesized as single-chain pre- which modulate differentiation by regulating other bHLH cursors and cleaved into an extracellular (NEC) and a proteins. HES1 also dimerizes with and activates signal trans- transmembrane (NTM) subunit by a furin-like protease in ducers and activators of transcription 3 (15). Other Notch the trans-Golgi (1–3). Notch NEC subunits include a variable targets include p21Cip/Waf (16), cyclin D1 (17), cyclin A (18), n number of epidermal –like repeats and three transcription factors of the nuclear factor- B (19), and Lin/Notch repeats (1, 2). These mediate the Ca2+-dependent poly(ADP-) polymerase families (20, 21) and interaction between NEC and NTM.NTM includes a transmem- ligase SKP2, through which Notch triggers degradation of brane region followed by a ‘‘RAM23’’ sequence, a series of p27kip1 (22). The set of directly and indirectly Notch-regulated ankyrin repeats, a nuclear localization sequence, a polyglut- genes and proteins is probably very large and context amine stretch, and a PEST sequence, which modulates dependent. IC half-life (1, 2). The RAM23 sequence and the ankyrin Phosphorylation of N by GSK-3h (23), CDK8 (24), and repeats interact with CSL transcription factors (mammalian possibly other kinases regulates its half-life. Several E3 ligases, CBF-1/Drosophila Suppressor of Hairless/ including Sel-10 (25), Itch (26), c-Cbl (27), and Deltex (28) Lag-1; refs. 4, 5). ubiquitinate Notch, triggering its degradation. Sel-10 catalyzes IC NEC glycosylation by Fringe glycosyltransferases regulates the phosphorylation-dependent ubiquitination of nuclear N relative affinity for ligands (1–3). binding dissociates (25, 29). NEC from NTM, and the NEC-ligand complex is trans- Notch and cancer. Notch activation is oncogenic in many IC endocytosed into ligand-expressing cells (6). NEC instances. N forms of all four Notch homologues act as triggers an extracellular in NTM by ADAM10 in vitro (30) and in animal models (31–34). EBV (a disintegrin and metalloprotease 10) or ADAM17 (1–3) immortalizing protein EBNA-2, which is necessary for EBV- followed by an intramembranous cleavage by g-secretase, induced transformation, mimics Notch by activating CBF-1 a multisubunit membrane aspartyl protease that includes (35, 36). Expression of Notch-1 NIC in mouse hematopoietic a catalytic subunit (Presenilin-1 or Presenilin-2), Pen-2, Aph1, precursors causes T-cell leukemia/lymphomas (31). Notch-1 and Nicastrin (7). This releases an intracellular domain (NIC) (34, 37) and Notch-4 (38) NIC cause mammary tumors in that translocates into the nucleus, where it modulates mice. transcription via CSL factors (1–3). NEC deletions of Notch-1 were discovered in T-cell acute All four mammalian Notches seem to use the same basic lymphoblastic leukemia (T-ALL; ref. 39). Subsequently, Weng signaling pathway via CSL transcription factors (8), and specific et al. (40) discovered that activating mutations of Notch-1 are pathways of individual homologues remain elusive. The present in f50% of T-ALL, making Notch-1 the most mammalian CSL, CBF-1/RBP-Jn, binds the sequence CGTGG- commonly activated in this disease. These mutations GAA and acts as a constitutive repressor by recruiting affect either the ‘‘heterodimerization domain’’ between the NEC a corepressor complex, including SMRT or N-coR, SKIP, CIR, and NTM subunits, facilitating NEC dissociation, or the COOH- and class I or II histone deacetylases (9), as well as SHARP (10), terminal PEST region, increasing the half-life of NIC. Some CtBP/CtIP (11), and, in Xenopus, methylated DNA-binding T-ALL cases contain double-mutant Notch alleles. Deregulated expression of Notch receptors, ligands, and targets is observed in solid tumors, including cervical, head and neck, endometrial, renal, lung, pancreatic, ovarian, breast and prostate carcinomas, osteosarcoma, mesothelioma, glio- Author’s Affiliation: Department of Pathology, and Breast mas, and medulloblastomas (41–55). High-level expression of Cancer Program, Cardinal Bernardin Cancer Center, Loyola University Chicago, Chicago, Illinois Notch-1 and Jagged-1 is associated with poor prognosis in Received 11/22/05; revised 12/16/05; accepted 12/16/05. breast cancer (56) and with metastasis in prostate cancer (55). Grant support: National Cancer Institute grant RO1CA 84065. Hodgkin’s lymphomas, anaplastic large-cell non-Hodgkin’s Requests for reprints: Lucio Miele, Breast Cancer Program, Cardinal Bernardin lymphomas, some acute myeloid leukemias, B-cell chronic Cancer Center, Loyola University Chicago, Room 236, Building 112, 2160 South lymphoid leukemias, and multiple myeloma also show deregu- 1st Avenue, Maywood, IL 60153. Phone: 708-327-3298; Fax: 708-327-2245; E-mail: [email protected]. lated expression of Notch receptors or ligands (57–60). Ras F 2006 American Association for Cancer Research. oncogenes activate wild-type Notch signaling, and Notch doi:10.1158/1078-0432.CCR-05-2570 activation is required for Ras-mediated transformation of

Clin Cancer Res 2006;12(4) February 15, 2006 1074 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Notch Signaling human fibroblasts (50). Transforming growth factor-a activates its maturation, be present at the membrane with Notch, and Notch through Ras in pancreatic carcinogenesis (41). Given the then cleave it after ligand binding and endocytosis. Three incidence of Ras mutations and Ras-activating mutations in recent articles support this model: endosomal accumulation of human cancers, it is perhaps not surprising that aberrant Notch Drosophila Notch caused by mutations in endosomal sorting activation is observed in numerous malignancies. regulators Vps25 or Erupted enhances Notch activity and cell The molecular basis for the oncogenic activity of Notch NICs proliferation (83–85). Endocytosis can also inhibit Notch remains unclear. Notch NIC transforms several cell types when signaling: negative regulator Numb causes Notch endocytosis expressed with oncoproteins that disable the G1-S checkpoint, with a-adaptin (86, 87) followed by -mediated such as adenovirus E1A (30), human papillomavirus E6 and E7 Notch degradation (88). Thus, depending on cellular context, (61, 62), Ras (63), (64), or SV40 large T (49). Notch may Notch may be sorted via endocytosis either to an activation contribute to tumorigenesis by inhibiting differentiation, pathway or to a degradation pathway (Fig. 1). promoting survival or accelerating proliferation. Potentially Noncanonical Notch signaling is attracting increasing atten- oncogenic targets of Notch-1 include cyclins D1 and D3 (17, 34, tion. There is evidence that ligands other than Delta/Jagged, 65), cyclin A (18), SKP2 (22), phosphatidylinositol 3-kinase such as F3/contactin (89), can activate Notch. Binding to non- (61, 66), AKT (61), ERBB2 (67), nuclear factor-nB (68), and CSL transcription factors, such as Nur77 (39), h-catenin (70), nuclear factor-nB2 (69), h-catenin (70), signal transducers or hypoxia-inducible factor-1a (71), may mediate a still and activators of transcription-3 (15), and hypoxia-inducible underappreciated subset of Notch effects. Some CSL- factor-1a (71). independent effects may be mediated by RING-finger E3 ligase An important exception is the epidermis, where Notch-1 acts Deltex (90–92). Deltex is transcriptionally up-regulated by as a tumor suppressor. Notch signaling induces growth arrest Notch, binds Notch, and may be both a mediator and an and differentiation in human (21) and murine (16) keratino- inhibitor of Notch signaling (93). Deltex inhibits Notch cytes in vitro. Tissue-specific ablation of Notch-1 in murine (34, 94), and recent data indicate that h-arrestin Kurz forms a epidermis causes hyperplasia, corneal epithelial proliferation, ternary complex with Notch and Deltex, leading to Notch and, eventually, spontaneous basal cell carcinoma–like ubiquitination and degradation (28). Deltex also catalyzes the tumors and facilitates chemical-induced carcinogenesis ubiquitination and degradation of MEKK1 (95), mediating (72). In primary keratinocytes, Notch-1 induces p21cip1/waf1 cross-talk between Notch and mitogen-activated via CBF-1 (16) and via HES-1–mediated inhibition of pathways. Deltex may have different effects on Notch depend- calcipressin, resulting in calcineurin activation (73). The ing on other binding partners. Finally, there is evidence that reasons for the differential behavior of Notch in keratinocytes Notch-1 NIC has protein kinase scaffold and anti-scaffold as opposed to other cell types are still unclear. Lathion et al., functions: Sade et al. (66) reported that NIC forms a complex however, showed that expression of Notch-1 NIC at moderate with p56Lck in T cells, promoting the phosphorylation of levels transforms keratinocytes with human papillomavirus phosphatidylinositol 3-kinase by p56Lck, thus activating the oncoproteins, whereas high-level overexpression inhibits phosphatidylinositol 3-kinase/AKT pathway. Conversely, growth (62). Like , Notch-1 may activate different targets Notch inhibits c-Jun NH2-terminal kinase activation by when expressed at low or high levels. Keratinocytes may have associating with scaffold JNP1 (96). an optimum level of Notch-1 for differentiation and In terms of tumor , although much attention has survival. High levels of Notch-1 would lead to growth arrest gone to the role of Notch in cancer cells, recent data suggest via p21cip1/waf1. Moderate levels would support survival that Notch signaling is also important in tumor microenviron- without blocking proliferation and synergize with other ment. Inside-out signaling by Notch ligands from tumor cells oncogenes. The clinical implications of these observations in activating Notch in stromal cells can promote tumorigenesis. humans are still unclear but deserve careful consideration. Myeloma cells overexpress Jagged-2, activating Notch in Notch inhibitors, which are in early clinical development, will stromal cells and inducing expression of myeloma growth have to be carefully monitored for potential dermatologic factor interleukin-6 (97). Head and neck squamous cell adverse effects, and strategies to circumvent such effects may carcinomas overexpress Jagged-1, which activates Notch in have to be devised. endothelial cells, promoting (98). Finally, an area of active investigation is the role of Notch signaling in cancer Recent Developments stem cells. Evidence suggests that many cancers, especially leukemias, breast cancers, and gliomas, contain a rare Several recent observations have highlighted the importance population of cells that are highly tumorigenic, unlike the of ubiquitin ligases and endocytic sorting in regulating Notch bulk of cancer cells that are unable to form tumors in vivo. Even signaling. In Drosophila, ubiquitination of Delta and Serrate by established cancer cell lines seem to contain this subpopula- E3 ligases Neuralized or Mindbomb is necessary for Epsin- tion. This has led to a theory that cancers arise from the mediated endocytosis (74) and Notch activation (75, 76). transformation of normal tissue stem cells or poorly differen- Monoubiquitination at a juxta membrane Lys1749 (in Notch- tiated progenitor cells. Once transformed, these generate all 1) and endocytosis of Notch are suggested to be required for other dead-end cancer cells through an aberrant process of g-secretase cleavage (77). This seems to contradict data tissue differentiation. The slowly proliferating cancer stem cells indicating that g-secretase exists as an active complex with have indefinite self-replication ability and are highly resistant to Notch at the membrane (78). There is evidence that (99, 100). According to this theory, conven- Presenilin-1 can act as an intracellular scaffold (79) and is tional chemotherapy kills the majority of progeny, nontumori- associated with newly synthesized Notch (80). Presenilin-1, genic cancer cells, but largely spares cancer stem cells, which with Nicastrin (81, 82), may form a scaffold for Notch during accumulate mutations and eventually give rise to recurrences

www.aacrjournals.org 1075 Clin Cancer Res 2006;12(4) February 15, 2006 Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Molecular Pathways

Fig. 1. Simplified diagram of a current model of canonical Notch signaling. Notch receptors exist in the membrane as heterodimers associated with g-secretase. Notch ligands bind the Notch NEC subunit and, after ubiquitination (UQ) by Neuralized or Mindbomb, dissociate it from NTM and trans-endocytose it via Epsin adaptor.This causes ADAM10 orADAM17to clip the extracellular portion of NTM, generating a cleavage intermediate. This is monoubiquitinated near the membrane by an E3 ligase (possibly Deltex), triggering endocytosis and g-secretase cleavage, which releases NIC.When endocytosis is mediated by Numb and a-adaptin, it leads to Notch degradation instead. Ubiquitination of Notch by Deltex in the presence of Kurz also leads to degradation. Cleaved NIC enters the nucleus, where it causes the dissociation of the SMRTcorepressor complex from CSL, and recruits the MAML1coactivator complex, resulting in transcription of target genes. Abbreviations: SMRT, silencing mediator of and thyroid ; CIR, CBF-1interacting corepressor; SHARP, SMRT/HDAC-1^ associated repressor protein; CtBP, COOH-terminal domain binding protein; CtIP, CtBP-interacting protein; HDAC, histone deacetylase; HAT, histone acetyltransferase (P/CAF, GCN5, or p300); MAML1,Mastermind-like 1;SKIP, ski-interacting protein. and metastases. Thus, novel therapeutic strategies that target the same (107). Consequently, Notch signaling is pathways necessary to cell fate decisions in cancer stem cells considered one of the most attractive candidate targets in cancer should improve therapeutic outcomes. Normal stem cells from stem cells. many tissues, including mammary epithelial stem cells (101), depend on Notch signaling for fate determination (102–105). Notch Inhibitors as CancerTherapeutics Cancer stem cells isolated as dye-excluding side population from numerous cancer cell lines express high levels of Numerous studies have proposed inhibition of Notch Notch-1 (106). Very recent data indicate that stem-like cells signaling as a strategy for cancer treatment (2). Selective contained in ductal carcinoma in situ clinical specimens have strategies include antisense, monoclonal , and RNA

Clin Cancer Res 2006;12(4) February 15, 2006 1076 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Notch Signaling

interference. Nonselective strategies include soluble or cell- with that block receptor activation. If intracellularly associated Notch decoys, g-secretase inhibitors, intracellular acting Notch inhibitors are shown to have unacceptable off- MAML1 decoys, and Ras signaling inhibitors. target effects, biologics, such as decoys and monoclonal g-Secretase inhibitors (GSI) have the most immediate antibodies to ligands or receptors, may be preferable. An therapeutic potential. GSI cbz-IL-CHO has Notch-1–depen- important question is whether selective inhibition of one of the dent antineoplastic activity in Ras-transformed fibroblasts four Notch homologues may be therapeutically desirable. In (50). GSI z-Leu-leu-Nle-CHO induces in melanoma this case, monoclonal antibodies would be the agent of choice, cell lines and melanoma xenografts, but not normal melano- and the best candidate may be Notch-4, given its more limited cytes, via p53-independent up-regulation of NOXA (108). GSI tissue distribution in normal mammals. Finally, it will be compound E causes growth arrest in T-ALL cells (40). Like important to establish whether cancer stem cell–targeted agents other small-molecule agents, GSIs have multiple effects. are optimally active alone or in rational combinations (e.g., a g-Secretase cleaves all Notch receptors and some ligands but Notch inhibitor with a Hedgehog inhibitor). Accumulating also ErbB4, syndecan, CD44, and other proteins (2, 7). knowledge about cross-talk between target pathways will guide Therapeutically, this may actually be advantageous because therapeutic choices. many cancers coexpress two or three different Notch The potential of cancer stem cell–targeted therapies homologues. Some GSIs may also affect proteases other than are largely unexplored. Random mutagenesis caused by DNA g-secretase. Pharmacologic studies with GSIs need to carefully damage is unlikely for most such agents, but effects on normal address target specificity, and a complementary transcriptional tissue stem cells are obviously possible. In general, it may be silencing approach is necessary in each model. Accumulating preferable to use these agents in pulsed, high-dose regimens preclinical evidence has led to the opening of phase 1 trials of rather than in chronic regimens that could eventually alter the a GSI in T-ALL and breast cancer. The pathways affected by differentiation of essential stem cells in the epidermis, intestine, Notch inhibition are likely to be context dependent, and or other vital tissues. rational combinations of GSIs with other antineoplastic drugs It is obviously far too early to evaluate the prospects of these will require mechanistic studies in individual models. Looking drugs. It is fair to say, however, that Notch inhibition in cancer beyond GSIs, ADAM inhibitors may find clinical uses to deserves a very thorough investigation because it could inhibit the first ligand-induced Notch cleavage. In light of simultaneously affect many of the most attractive therapeutic recent evidence, blocking specific E3 ligases responsible for targets recently identified, including signal transducers, and ubiquitination of Notch ligands or monoubiquitination of activators of transcription 3, nuclear factor-nB, AKT, cyclins, Notch receptors may be an alternate approach. NOXA, and others. Small molecules that interfere with Notch coactivator binding may selectively inhibit CSL signaling but not non- Acknowledgments canonical signaling. Depending on the relative importance of Notch downstream pathways in individual cancers and normal I thank Lynda Song, Paola Rizzo, and Clodia Osipo for critical reading and apolo- tissues, this may be an advantage or a disadvantage compared gize for omitting many primary references due to space limitations.

References 1. Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch 9. Lai EC. Keeping a good pathway down: transcription- transcription and CDK2activity by Notch(ic): implica- signaling: cell fate control and signal integration in al repression of Notch pathway target genes by CSL tion for disruption in transformation by development. Science 1999;284:770 ^ 6. proteins. EMBO Rep 2002;3:840 ^ 5. Notch(ic).MolCellBiol2001;21:5925^34. 2. Nickoloff BJ, Osborne BA, Miele L. Notch signaling 10. Oswald F, Kostezka U, Astrahantseff K, et al. SHARP 18. Baonza A, Freeman M. Control of cell proliferation in as a therapeutic target in cancer: a new approach to is a novel component of the Notch/RBP-Jkappa sig- the Drosophila eye by Notch signaling. Dev Cell the development of cell fate modifying agents. Onco- nalling pathway. EMBO J 2002;21:5417^ 26. 2005;8:529^ 39. 2003;22:6598 ^ 608. 11. Oswald F, Winkler M, Cao Y, et al. RBP-J{kappa}/ 19. Cheng P, Zlobin A,Volgina V, et al. Notch-1regulates 3. Lai EC. Notch signaling: control of cell communica- SHARP recruits CtIP/CtBP to silence NF-kappaB activity in hemopoietic progenitor cells. tion and cell fate. Development 2004;131:965 ^ 73. Notch target genes. Mol Cell Biol 2005;25: J Immunol 2001;167:4458 ^ 67. 4. Tamura K,Taniguchi Y, Minoguchi S, et al. Physical in- 10379 ^ 9 0. 20. Garces C, Ruiz-Hidalgo MJ, Font de Mora J, et al. teraction between a novel domain of the receptor 12. StanchevaI,CollinsAL,VandenVeyverIB,Zoghbi Notch-1controls the expression of fatty acid-activated Notch and the RBP-Jk/Su(H).Curr H, Meehan RR. A mutant form of MeCP2protein transcription factors and is required for adipogenesis. Biol 1995;5:1416 ^ 23. associated with human Rett syndrome cannot be JBiolChem1997;272:29729^34. 5. Aster JC, Robertson ES, Hasserjian RP,Turner JR, displaced from methylated DNA by notch in Xeno- 21. Nickoloff BJ, Qin JZ, Chaturvedi V, Denning MF, pus Kieff E, Sklar J. Oncogenic forms of NOTCH1 lacking . Mol Cell 2003;12:425 ^ 35. Bonish B, Miele L. Jagged-1mediated activation of either the primary for RBP-Jkappa or nu- 13. IsoT,SartorelliV,PoizatC,etal.HERP,anovelheter- notch signaling induces complete maturation of clear localization sequences retain the ability to asso- odimer partner of HES/E(spl) in Notch signaling. Mol human keratinocytes through NF-kappaB and PPAR- ciate with RBP-Jkappa and activate transcription. Cell Biol 2001;21:6080 ^ 9. gamma. Cell Death Differ 2002;9:842 ^ 55. J Biol Chem 1997;272:11336 ^ 43. 14. Maier MM, Gessler M. Comparative analysis of the 22. Sarmento LM, Huang H, Limon A, et al. Notch1 6. Parks AL, Klueg KM, Stout JR, Muskavitch MA. human and mouse hey1promoter: hey genes are new modulates timing of G1-S progression by inducing Ligand endocytosis drives receptor dissociation and notch target genes [In Process Citation]. Biochem SKP2transcription and p27Kip1degradation. J Exp activation in the Notch pathway. Development Biophys Res Commun 2000;275:652^ 60. Med 2005;202:157 ^ 68. 20 0 0;127:1373 ^ 8 5. 15. Kamakura S, Oishi K, Yoshimatsu T, Nakafuku M, 23. Foltz DR, Santiago MC, Berechid BE, Nye JS. 7. Kopan R, Ilagan MX. Gamma-secretase: proteasome Masuyama N, GotohY.Hes binding to STAT3 mediates Glycogen synthase kinase-3beta modulates notch of the membrane? Nat Rev Mol Cell Biol 2004;5: crosstalk between Notch andJAK-STATsignalling. Nat signaling and stability. Curr Biol 2002;12:1006 ^ 11. 499^504. Cell Biol 2004;6:547 ^ 54. 24. Fryer CJ, White JB, Jones KA. Mastermind recruits 8. MizutaniT,TaniguchiY,AokiT, Hashimoto N, Honjo T. 16. Rangarajan A,Talora C, Okuyama R, et al. Notch sig- CycC:CDK8 to phosphorylate the Notch ICD and co- Conservation of the biochemical mechanisms of naling is a direct determinant of keratinocyte growth ordinate activation with turnover. Mol Cell 2004;16: among mammalian Notch arrest and entry into differentiation. EMBO J 2001;20: 509^20. family members. Proc Natl Acad Sci U S A 2001;98: 3427^36. 25. Oberg C, Li J, Pauley A, Wolf E, Gurney M, Lendahl 9026^31. 17. Ronchini C, Capobianco AJ. Induction of cyclin D1 U. The notch intracellular domain is ubiquitinated and

www.aacrjournals.org 1077 Clin Cancer Res 2006;12(4) February 15, 2006 Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Molecular Pathways

negatively regulated by the mammalian sel-10 homo- 47. Suzuki T, Aoki D, Susumu N, UdagawaY, Nozawa S. lates cell death independently of differentiation in mu- log. J Biol Chem 2001;276:35847 ^ 53. Imbalanced expression of TAN-1and human notch4 in rine erythroleukemia cells through multiple apoptosis 26. Qiu L, Joazeiro C, Fang N, et al. Recognition and endometrial cancers. Int J Oncol 2000;17:1131^ 9. and cell cycle pathways. J Cell Physiol 2004;199: Ubiquitination of Notch by Itch, a Hect-type E3 Ubiq- 48. Hendrix MJ, Seftor RE, Seftor EA, et al. Transendo- 418 ^ 33. uitin Ligase. J Biol Chem 2000;275:35734 ^ 7. thelial function of human metastatic melanoma cells: 69. Oswald F, Liptay S, Adler G, Schmid RM. NF- 27. Jehn BM, Dittert I, Beyer S, von der MK, Bielke W. role of the microenvironment in cell-fate determination. kappaB2is a putative target gene of activated Notch- c-Cbl binding and ubiquitin-dependent lysosomal Cancer Res 2002;62:665 ^ 8. 1viaRBP-Jkappa.MolCellBiol1998;18:2077^88. degradation of membrane-associated Notch1. J Biol 49. Bocchetta M, Miele L, Pass HI, Carbone M. Notch-1 70. Hayward P,Brennan K, Sanders P,et al. Notch mod- Chem 2002;277:8033^40. induction, a novel activity of SV40 required for growth ulates Wnt signalling by associating with Armadillo/ 28. MukherjeeA,Veraksa A, BauerA, Rosse C, Camonis of SV40-transformed human mesothelial cells. Onco- beta-catenin and regulating its transcriptional activity. J, Artavanis-Tsakonas S. Regulation of Notch signal- gene 2003;22:81 ^ 9. Development 2005;132:1819 ^ 30. ling by non-visual beta-arrestin. Nat Cell Biol 2005;7: 50.Weijzen S, Rizzo P, Braid M, et al. Activation of 71. Gustafsson MV, Zheng X, PereiraT, et al. Hypoxia 1091 ^ 101. Notch-1signaling maintains the neoplastic phenotype requires notch signaling to maintain the undifferentiat- 29. Gupta-Rossi N, Le BO, Gonen H, et al. Functional in human Ras-transformed cells. Nat Med 2002;8: ed cell state. Dev Cell 2005;9:617^ 28. interaction between SEL-10, an F-box protein, and 979 ^ 86. 72. Nicolas M,WolferA, Raj K, et al. Notch1functions as the nuclear form of activated Notch1receptor. J Biol 51. Haruki N, Kawaguchi KS, Eichenberger S, et al. a tumor suppressor in mouse skin. Nat Genet 2003; Chem 2001;276:34371 ^ 8. Dominant-negative Notch3 receptor inhibits mitogen 33:416^21. 30. Capobianco AJ, Zagouras P, Blaumueller CM, activated protein kinase pathway and the growth of 73. Mammucari C, Tommasi di Vignano A, Sharov AA, Artavanis-Tsakonas S, BishopJM. Neoplastic transfor- human lung cancers. Cancer Res 2005;65:3555 ^ 61. et al. Integration of and calcineurin/NFAT mation by truncated alleles of human NOTCH1/TAN1 52. Purow BW, Haque RM, Noel MW, et al. Expression signaling pathways in keratinocyte growth and differ- and NOTCH2. Mol Cell Biol 1997;17:6265 ^ 73. of Notch-1and its ligands, Delta-like-1and Jagged-1, is entiation control. Dev Cell 2005;8:665 ^ 76. 31. PearWS,AsterJC,ScottML,etal.Exclusivedevel- critical for glioma cell survival and proliferation. Cancer 74. Wang W, Struhl G. Distinct roles for Mind bomb, opment of T cell neoplasms in mice transplanted with Res 2005;65:2353^63. Neuralized and Epsin in mediating DSL endocytosis bone marrow expressing activated Notch alleles. 53. FanX,MikolaenkoI,ElhassanI,etal.Notch1and and signaling in Drosophila. Development 2005;132: JExpMed1996;183:2283^91. notch2have opposite effects on embryonal brain tu- 2883^94. 32. Bellavia D, CampeseAF, Alesse E, et al. Constitutive mor growth. Cancer Res 2004;64:7787 ^93. 75. Pitsouli C, Delidakis C. The interplay between DSL activation of NF-kappaB and T-cell leukemia/lympho- 54. Hopfer O, Zwahlen D, Fey MF, Aebi S. The and ubiquitin ligases in Notch signaling. De- ma in Notch3 transgenic mice. EMBO J 2000;19: pathway in ovarian carcinomas and adenomas. Br J velopment 2005;132:4041 ^ 50. 3337^48. Cancer 2005;93:709 ^ 18. 76. Le Borgne R, Bardin A, Schweisguth F.The roles of 33. Callahan R, Raafat A. Notch signaling in mammary 55. Santagata S, Demichelis F, Riva A, et al. JAGGED1 receptor and ligand endocytosis in regulating Notch gland tumorigenesis. J Mammary Gland Biol Neopla- expression is associated with prostate cancer metas- signaling. Development 2005;132:1751 ^ 62. sia 2001;6:23 ^ 36. tasis and recurrence. Cancer Res 2004;64:6854 ^ 7. 77. Gupta-Rossi N, Six E, LeBail O, et al. Monoubiquiti- 34. Kiaris H, Politi K, Grimm LM, et al. Modulation of 56. Reedijk M, Odorcic S, Chang L, et al. High-level nation and endocytosis direct gamma-secretase notch signaling elicits signature tumors and inhibits coexpression of JAG1and NOTCH1is observed in hu- cleavage of activated Notch receptor. J Cell Biol hras1-induced oncogenesis in the mouse mammary man breast cancer and is associated with poor overall 2004;166:73 ^ 83. epithelium. Am J Pathol 2004;165:695 ^ 705. survival. Cancer Res 2005;65:8530 ^ 7. 78. Tarassishin L,Yin YI, Bassit B, Li YM. Processing of 35. Hsieh JJ, Hayward SD. Masking of the CBF1/RBPJ 57. Tohda S, Nara N. Expression of Notch1and Jagged1 Notch and amyloid precursor protein by gamma-sec- kappa transcriptional repression domain by Epstein- proteins in acute myeloid leukemia cells. Leuk Lym- retase is spatially distinct. Proc Natl Acad Sci U S A Barr virus EBNA2. Science 1995;268:560 ^ 3. phoma 2001;42:467 ^ 72. 2004;101:17050^5. 36. Hsieh JJ, Nofziger DE,Weinmaster G, Hayward SD. 58. Hubmann R, Schwarzmeier JD, Shehata M, et al. 79. Kang DE, Soriano S, Xia X, et al. Presenilin couples Epstein-Barr virus immortalization: Notch2interacts Notch2is involved in the overexpression of CD23in the paired phosphorylation of beta-catenin indepen- with CBF1 and blocks differentiation. J Virol 1997;71: B-cell chronic lymphocytic leukemia. Blood 2002;99: dent of axin: implications for beta-catenin activation 193 8 ^ 4 5. 3742^7. in tumorigenesis. Cell 2002;110:751 ^ 62. 37. Dievart A, Beaulieu N, Jolicoeur P. Involvement of 59. Jundt F, Anagnostopoulos I, Forster R, Mathas S, 80. RayWJ,Yao M, Mumm J, et al. Cell surface preseni- Notch1 in the development of mouse mammary Stein H, Dorken B. Activated Notch1 signaling pro- lin-1participates in the gamma-secretase-like proteol- tumors. Oncogene 1999;18:5973 ^ 81. motes tumor cell proliferation and survival in Hodgkin ysis of Notch. J Biol Chem1999;274:36801^ 7. 38. Gallahan D, Callahan R. The mouse mammary and anaplastic large cell lymphoma. Blood 2002;99: 81. Li T, Ma G, Cai H, Price DL, Wong PC. Nicastrin is tumor associated gene INT3 is a unique member of 3398^403. required for assembly of presenilin/gamma-secretase the NOTCH gene family (NOTCH4). Oncogene 1997; 60. Nefedova Y, Cheng P, Alsina M, Dalton WS, complexes to mediate Notch signaling and for pro- 14 :18 8 3 ^ 9 0. Gabrilovich DI. Involvement of Notch-1 signaling in cessing and trafficking of beta-amyloid precursor pro- 39. Jehn BM, Bielke W, Pear WS, Osborne BA. Protec- bone marrow stroma-mediated de novo resis- tein in mammals. J Neurosci 2003;23:3272 ^ 7. tive effects of notch-1 on TCR-induced apoptosis. tance of myeloma and other malignant lymphoid cell 82. Chen F,Yu G, Arawaka S, et al. Nicastrin binds to JImmunol1999;162:635^8. lines. Blood 2004;103:3503 ^ 10. membrane-tethered Notch. Nat Cell Biol 2001;3: 40.Weng AP, Ferrando AA, Lee W, et al. Activating 61. Rangarajan A, Syal R, Selvarajah S, Chakrabarti O, 751 ^ 4. mutations of NOTCH1 in human T cell acute lympho- A, Krishna S. Activated Notch1signaling cooper- 83.Vaccari T, Bilder D. The Drosophila tumor sup- blastic leukemia. Science 2004;306:269^71. ates with papillomavirus oncogenes in transformation pressor vps25 prevents nonautonomous overprolif- 41. Miyamoto Y, Maitra A, Ghosh B, et al. Notch medi- and generates resistance to apoptosis on matrix with- eration by regulating Notch trafficking. Dev Cell atesTGFalpha-induced changes in epithelial differenti- drawal through PKB/Akt.Virology 2001;286:23 ^ 30. 2005;9:687^98. ation during pancreatic tumorigenesis. Cancer Cell 62. Lathion S, Schaper J, Beard P, Raj K. Notch1 84.Thompson BJ, MathieuJ, Sung HH, Loeser E, Rorth 2003;3:565 ^ 76. can contribute to viral-induced transformation of pri- P, Cohen SM. Tumor suppressor properties of the 42. Zagouras P, Stifani S, Blaumueller CM, Carcangiu mary human keratinocytes. Cancer Res 2003;63: ESCRT-II complex component Vps25 in Drosophila. ML, Artavanis-Tsakonas S. Alterations in Notch signal- 8687 ^ 94. Dev Cell 2005;9:711^ 20. ing in neoplastic lesions of the human cervix. Proc Natl 63. Fitzgerald K, Harrington A, Leder P. Ras pathway 85. Moberg KH, Schelble S, Burdick SK, Hariharan IK. Acad Sci U S A1995;92:6414^ 8. signals are required for notch-mediated oncogenesis. Mutations in erupted, the Drosophila ortholog of 43. Daniel B, Rangarajan A, Mukherjee G, Vallikad E, Oncogene 2000;19:4191 ^ 8. mammalian tumor susceptibility gene 101, elicit Krishna S.The link between integration and expression 64. Girard L, Hanna Z, Beaulieu N, et al. Frequent provi- non-cell-autonomous overgrowth. Dev Cell 2005;9: of human papillomavirus type 16 genomes and cellular rus insertional mutagenesis of Notch1in thymomas of 699 ^710. changes in the evolution of cervical intraepithelial neo- MMTVD/myc transgenic mice suggests a collabora- 86. Berdnik D, Torok T, Gonzalez-Gaitan M, Knoblich plastic lesions. J GenVirol 1997;78:1095 ^ 101. tion of c-myc and Notch1for oncogenesis. Genes Dev JA. The endocytic protein alpha-Adaptin is required 44. Dang TP, GazdarAF,Virmani AK, et al. 19 9 6;10 :193 0 ^ 4 4. for numb-mediated asymmetric cell division in Dro- 19 translocation, overexpression of Notch3, and hu- 65. Sicinska E, Aifantis I, Le Cam L, et al. Requirement sophila. Dev Cell 2002;3:221 ^ 31. man lung cancer. JNatl Cancer Inst 2000;92:1355 ^ 7. for cyclin D3 in lymphocyte development and T cell 87. Santolini E, Puri C, Salcini AE, et al. Numb is an 45. Leethanakul C, PatelV, Gillespie J, et al. Distinct pat- leukemias. Cancer Cell 2003;4:451 ^ 61. endocytic protein. J Cell Biol 2000;151:1345 ^ 52. tern of expression of differentiation and growth-relat- 66. Sade H, Krishna S, Sarin A.The anti-apoptotic effect 88. McGillMA,McGladeCJ.Mammaliannumb ed genes in squamous cell carcinomas of the head and of Notch-1 requires p56lck-dependent, Akt/PKB- proteins promote Notch1receptor ubiquitination and neck revealed by the use of laser capture microdissec- mediated signaling in T cells. J Biol Chem 2004;279: degradation of the Notch1intracellular domain. J Biol tion and cDNA arrays. Oncogene 2000;19:3220 ^ 4. 2937 ^ 44. Chem 2003;278:23196 ^ 203. 46. Rae FK, Stephenson SA, Nicol DL, Clements JA. 67. Chen Y, Fischer WH, Gill GN. Regulation of the 89. Hu QD, Ang BT, Karsak M, et al. F3/contactin acts Novel association of a diverse range of genes with re- ERBB-2promoter by RBPJkappa and NOTCH. J Biol as a functional ligand for Notch during oligodendro- nal cell carcinoma as identified by differential display. Chem 1997;272:14110 ^ 4. cyte maturation. Cell 2003;115:163 ^ 75. Int J Cancer 2000;88:726 ^ 32. 68. Jang MS, Miao H, Carlesso N, et al. Notch-1 regu- 90. Matsuno K, Eastman D, Mitsiades T, et al. Human

Clin Cancer Res 2006;12(4) February 15, 2006 1078 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Notch Signaling

deltex is a conserved regulator of Notch signalling. Nat 97. Houde C, Li Y, Song L, et al. Over-expression of the 104. Luo D, Renault VM, Rando TA. The regulation of Genet 1998;19:74 ^ 8. NOTCH ligand JAG2in malignant plasma cells from Notch signaling in muscle stem cell activation and 91. Ordentlich P, Lin A, Shen CP, et al. Notch inhibition multiple myeloma patients and cell lines. Blood 2004; postnatal myogenesis. Semin Cell Dev Biol 2005;16: of E47 supports the existence of a novel signaling 104:3697^704. 612^ 22. pathway.MolCellBiol1998;18:2230^9. 98. Zeng Q, Li S, Chepeha DB, et al. Crosstalk between 105. Fre S, Huyghe M, Mourikis P, Robine S, Louvard D, 92. Yamamoto N, Yamamoto S, Inagaki F, et al. Role of tumor and endothelial cells promotes tumor angiogen- Artavanis-Tsakonas S. Notch signals control the fate of Deltex1 as a transcriptional regulator downstream esis by MAPK activation of Notch signaling. Cancer immature progenitor cells in the intestine. Nature of the Notch receptor. J Biol Chem 2001;276: Cell 2005;8:13^ 23. 2005;435:964^8. 45031^40. 99. Al Hajj M, Becker MW,Wicha M,Weissman I, Clarke 106. Patrawala L, Calhoun T, Schneider-Broussard R, 93. Matsuno K, Ito M, Hori K, et al. Involvement of a MF.Therapeutic implications of cancer stem cells. Curr Zhou J, Claypool K, Tang DG. Side population is -rich motif and RING-H2finger of Deltex in Opin Genet Dev 2004;14:43 ^ 7. enriched in tumorigenic, stem-like cancer cells, where- the regulation of Notch signaling. Development 100. Song L, Miele L. Cancer stem cells: an old idea as ABCG2+ and ABCG2À cancer cells are similarly 2002;129:1049^59. that’s new again.Womens Onc Rev 2006, in press. tumorigenic. Cancer Res 2005;65:6207 ^ 19. 94. Izon DJ, Aster JC, HeY, et al. Deltex1redirects lym- 101. Dontu G, Jackson KW, McNicholas E, Kawamura 107. Farnie G, Brennan K, Clarke RB, Bundred NJ. phoid progenitors to the B cell lineage by antagonizing MJ, Abdallah WM, Wicha MS. Role of Notch signal- Ductal Carcinoma In situ (DCIS) mammosphere Notch1. Immunity 2002;16:231 ^ 43. ing in cell-fate determination of human mammary formation effect of (EGF) 95. LiuWH, Lai MZ. Deltex regulatesT-cell activation by stem/progenitor cells. Breast Cancer Res 2004;6: and notch signaling pathways on self-renewal ca- targeted degradation of active MEKK1. Mol Cell Biol R605 ^ 15. pacity. Breast Cancer Res Treat 2005;94 Suppl 2005;25:1367 ^ 78. 102. Campos LS, Decker L,TaylorV, Skarnes W. Notch, 1:S14. 96. KimJW, Kim MJ, Kim KJ, et al. Notch interferes with EGFR and beta 1integrin pathways are coordinated in 108. Qin JZ, Stennett L, Bacon P, et al. p53-indepen- the scaffold function ofJNK-interacting protein 1to in- neural stem cells. J Biol Chem. Epub 2005 Dec 6. dent NOXA induction overcomes apoptotic resistance hibit the JNK signaling pathway. Proc Natl Acad Sci 103. SuzukiT, Chiba S. Notch signaling in hematopoiet- of malignant melanomas. Mol Cancer Ther 2004;3: U S A 2005;102:14308 ^ 13. ic stem cells. Int JHematol 2005;82:285 ^94. 895^902.

www.aacrjournals.org 1079 Clin Cancer Res 2006;12(4) February 15, 2006 Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Notch signaling.

Lucio Miele

Clin Cancer Res 2006;12:1074-1079.

Updated version Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/12/4/1074.citation

Cited articles This article cites 106 articles, 56 of which you can access for free at: http://clincancerres.aacrjournals.org/content/12/4/1074.citation.full#ref-list-1

Citing articles This article has been cited by 33 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/12/4/1074.citation.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/12/4/1074.citation. Click on "Request Permissions" which will take you to the Copyright Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research.