Skip to main content

The γ-Secretase Protease Complexes in Neurodegeneration, Cancer and Immunity

  • Chapter
  • First Online:

Abstract

The intramembrane-cleaving proteases (I-CliPs) are necessary for the proteolytic cleavage of several transmembrane proteins and initiation of divergent signalling events. The tetrameric γ-secretase protease complexes, comprised of presenilin and three other subunits, represent a major subclass of the I-CliPs. The γ-secretase protease complexes are involved in regulated intramembrane proteolysis, an evolutionary conserved and important signal transduction process encompassing the sequential proteolysis of transmembrane substrates that are central to many physiological processes, including embryonic development, haematopoiesis, and normal functioning of the nervous and immune systems. Deregulated intramembrane proteolysis of certain substrates is proposed to be associated with neurodegeneration, cancer and impaired immune function. In this chapter, we summarise the major biochemical and functional properties (structure, catalytic mechanisms, substrate specificities, and regulation) of the γ-secretase protease complexes. We also present evidence for a role of γ-secretase protease complexes in neurodegeneration, cancer and inflammatory disease and consider the use of γ-secretase inhibitors as prospective therapeutics in several diseases.

This is a preview of subscription content, log in via an institution.

References

  1. Brown MS, Ye J, Rawson RB et al (2000) Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell 100(4):391–398

    Article  CAS  PubMed  Google Scholar 

  2. Lichtenthaler SF, Haass C, Steiner H (2011) Regulated intramembrane proteolysis—lessons from amyloid precursor protein processing. J Neurochem 117(5):779–796

    Article  CAS  PubMed  Google Scholar 

  3. Schroder B, Saftig P (2016) Intramembrane proteolysis within lysosomes. Ageing Res Rev

    Google Scholar 

  4. Lemberg MK, Menendez J, Misik A et al (2005) Mechanism of intramembrane proteolysis investigated with purified rhomboid proteases. EMBO J 24(3):464–472

    Article  CAS  PubMed  Google Scholar 

  5. Jurisch-Yaksi N, Sannerud R, Annaert W (2013) A fast growing spectrum of biological functions of gamma-secretase in development and disease. Biochim Biophys Acta 1828(12):2815–2827

    Article  CAS  PubMed  Google Scholar 

  6. Langosch D, Scharnagl C, Steiner H et al (2015) Understanding intramembrane proteolysis: from protein dynamics to reaction kinetics. Trends Biochem Sci 40(6):318–327

    Article  CAS  PubMed  Google Scholar 

  7. Duggan SP, McCarthy JV (2016) Beyond gamma-secretase activity: the multifunctional nature of presenilins in cell signalling pathways. Cell Signal 28(1):1–11

    Article  CAS  PubMed  Google Scholar 

  8. Hurst TP, Coleman-Vaughan C, Patwal I et al (2016) Regulated intramembrane proteolysis, innate immunity and therapeutic targets in Alzheimer’s disease. AIMS Mol Sci 3(2):138–157

    Article  CAS  Google Scholar 

  9. Sannerud R, Annaert W (2009) Trafficking, a key player in regulated intramembrane proteolysis. Semin Cell Dev Biol 20(2):183–190

    Article  CAS  PubMed  Google Scholar 

  10. Lichtenthaler SF, Steiner H (2007) Sheddases and intramembrane-cleaving proteases: RIPpers of the membrane. Symposium on Regulated Intramembrane Proteolysis. EMBO Rep 8(6):537–541

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Edwards DR, Handsley MM, Pennington CJ (2008) The ADAM metalloproteinases. Mol Aspects Med 29(5):258–289

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. van Hinsbergh VWM, Koolwijk P (2008) Endothelial sprouting and angiogenesis: matrix metalloproteinases in the lead. Cardiovasc Res 78(2):203–212

    Article  PubMed  CAS  Google Scholar 

  13. Lin CY, Tseng IC, Chou FP et al (2008) Zymogen activation, inhibition, and ectodomain shedding of matriptase. Front Biosci 13:621–635

    Article  CAS  PubMed  Google Scholar 

  14. Morohashi Y, Tomita T (2013) Protein trafficking and maturation regulate intramembrane proteolysis. Biochimica et Biophysica Acta (BBA)—Biomembranes 1828(12):2855–2861

    Article  CAS  Google Scholar 

  15. McCarthy JV, Twomey C, Wujek P (2009) Presenilin-dependent regulated intramembrane proteolysis and gamma-secretase activity. Cell Mol Life Sci 66(9):1534–1555

    Article  CAS  PubMed  Google Scholar 

  16. De Strooper B, Saftig P, Craessaerts K et al (1998) Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 391(6665):387–390

    Article  PubMed  CAS  Google Scholar 

  17. De Strooper B, Annaert W, Cupers P et al (1999) A presenilin-1-dependent [gamma]-secretase-like protease mediates release of Notch intracellular domain. Nature 398(6727):518–522

    Article  PubMed  CAS  Google Scholar 

  18. Guruharsha KG, Kankel MW, Artavanis-Tsakonas S (2012) The Notch signalling system: recent insights into the complexity of a conserved pathway. Nat Rev Genet 13(9):654–666

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Marambaud P, Shioi J, Serban G et al (2002) A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. EMBO J 21(8):1948–1956

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fluhrer R, Grammer G, Israel L et al (2006) A [gamma]-secretase-like intramembrane cleavage of TNF[alpha] by the GxGD aspartyl protease SPPL2b. Nat Cell Biol 8(8):894–896

    Article  CAS  PubMed  Google Scholar 

  21. Elzinga BM, Twomey C, Powell JC et al (2009) Interleukin-1 receptor type 1 is a substrate for gamma-secretase-dependent regulated intramembrane proteolysis. J Biol Chem 284(3):1394–1409

    Article  CAS  PubMed  Google Scholar 

  22. Kuhn PH, Marjaux E, Imhof A et al (2007) Regulated intramembrane proteolysis of the interleukin-1 receptor II by alpha-, beta-, and gamma-secretase. J Biol Chem 282(16):11982–11995

    Article  CAS  PubMed  Google Scholar 

  23. Twomey C, Qian S, McCarthy JV (2009) TRAF6 promotes ubiquitination and regulated intramembrane proteolysis of IL-1R1. Biochem Biophys Res Commun 381(3):418–423

    Article  CAS  PubMed  Google Scholar 

  24. McElroy B, Powell JC, McCarthy JV (2007) The insulin-like growth factor 1 (IGF-1) receptor is a substrate for gamma-secretase-mediated intramembrane proteolysis. Biochem Biophys Res Commun 358(4):1136–1141

    Article  CAS  PubMed  Google Scholar 

  25. Sturtevant MA, Roark M, Bier E (1993) The Drosophila rhomboid gene mediates the localized formation of wing veins and interacts genetically with components of the EGF-R signaling pathway. Genes Dev 7(6):961–973

    Article  CAS  PubMed  Google Scholar 

  26. Ni CY, Murphy MP, Golde TE et al (2001) gamma-secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Science 294(5549):2179–2181

    Article  CAS  PubMed  Google Scholar 

  27. Powell JC, Twomey C, Jain R et al (2009) Association between Presenilin-1 and TRAF6 modulates regulated intramembrane proteolysis of the p75NTR neurotrophin receptor. J Neurochem 108(1):216–230

    Article  CAS  PubMed  Google Scholar 

  28. Berghoff J, Jaisimha AV, Duggan S et al (2015) Gamma-secretase-independent role for cadherin-11 in neurotrophin receptor p75 (p75(NTR)) mediated glioblastoma cell migration. Mol Cell Neurosci 69:41–53

    Article  CAS  PubMed  Google Scholar 

  29. Zampieri N, Xu CF, Neubert TA et al (2005) Cleavage of p75 neurotrophin receptor by alpha-secretase and gamma-secretase requires specific receptor domains. J Biol Chem 280(15):14563–14571

    Article  CAS  PubMed  Google Scholar 

  30. Lammich S, Okochi M, Takeda M et al (2002) Presenilin-dependent Intramembrane Proteolysis of CD44 Leads to the Liberation of Its Intracellular Domain and the Secretion of an Aβ-like Peptide. J Biol Chem 277(47):44754–44759

    Article  CAS  PubMed  Google Scholar 

  31. Wunderlich P, Glebov K, Kemmerling N et al (2013) Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and gamma-secretase-dependent intramembranous cleavage. J Biol Chem 288(46):33027–33036

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Maetzel D, Denzel S, Mack B et al (2009) Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol 11(2):162–171

    Article  CAS  PubMed  Google Scholar 

  33. Black RA, Rauch CT, Kozlosky CJ et al (1997) A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385(6618):729–733

    Article  CAS  PubMed  Google Scholar 

  34. Friedmann E, Hauben E, Maylandt K et al (2006) SPPL2a and SPPL2b promote intramembrane proteolysis of TNFalpha in activated dendritic cells to trigger IL-12 production. Nat Cell Biol 8(8):843–848

    Article  CAS  PubMed  Google Scholar 

  35. Chhibber-Goel J, Coleman-Vaughan C, Agrawal V et al (2016) γ-secretase activity is required for regulated intramembrane proteolysis of tumor necrosis factor (TNF) receptor 1 and TNF-mediated pro-apoptotic signaling. J Biol Chem 291(11):5971–5985

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Selkoe DJ (2001) Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev 81(2):741–766

    Article  CAS  PubMed  Google Scholar 

  37. Small SA, Gandy S (2006) Sorting through the cell biology of Alzheimer’s disease: intracellular pathways to pathogenesis. Neuron 52(1):15–31

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Tomita T, Iwatsubo T (2013) Structural biology of presenilins and signal peptide peptidases. J Biol Chem 288(21):14673–14680

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Weidemann A, Eggert S, Reinhard FB et al (2002) A novel epsilon-cleavage within the transmembrane domain of the Alzheimer amyloid precursor protein demonstrates homology with Notch processing. Biochemistry 41(8):2825–2835

    Article  CAS  PubMed  Google Scholar 

  40. Zhao G, Cui MZ, Mao G et al (2005) γ-cleavage is dependent on zeta-cleavage during the proteolytic processing of amyloid precursor protein within its transmembrane domain. J Biol Chem 280(45):37689–37697

    Article  CAS  PubMed  Google Scholar 

  41. Zhao G, Mao G, Tan J et al (2004) Identification of a new presenilin-dependent zeta-cleavage site within the transmembrane domain of amyloid precursor protein. J Biol Chem 279(49):50647–50650

    Article  CAS  PubMed  Google Scholar 

  42. Okochi M, Fukumori A, Jiang J et al (2006) Secretion of the Notch-1 Aβ-like peptide during notch signaling. J Biol Chem 281(12):7890–7898

    Article  CAS  PubMed  Google Scholar 

  43. Beel AJ, Sanders CR (2008) Substrate specificity of gamma-secretase and other intramembrane proteases. Cell Mol Life Sci 65(9):1311–1334

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sun L, Li X, Shi Y (2016) Structural biology of intramembrane proteases: mechanistic insights from rhomboid and S2P to γ-secretase. Curr Opin Struct Biol 37:97–107

    Article  CAS  PubMed  Google Scholar 

  45. Wolfe MS (2009) Intramembrane-cleaving Proteases. J Biol Chem 284(21):13969–13973

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Martoglio B (2003) Intramembrane proteolysis and post-targeting functions of signal peptides. Biochem Soc Trans 31(Pt 6):1243–1247

    Article  CAS  PubMed  Google Scholar 

  47. Strisovsky K (2013) Structural and mechanistic principles of intramembrane proteolysis—lessons from rhomboids. FEBS J 280(7):1579–1603

    Article  CAS  PubMed  Google Scholar 

  48. Laurent SA, Hoffmann FS, Kuhn PH et al (2015) γ-secretase directly sheds the survival receptor BCMA from plasma cells. Nat Commun 6:7333

    Article  CAS  PubMed  Google Scholar 

  49. Fluhrer R, Steiner H, Haass C (2009) Intramembrane proteolysis by signal peptide peptidases: a comparative discussion of GXGD-type aspartyl proteases. J Biol Chem 284(21):13975–13979

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nyborg AC, Kornilova AY, Jansen K et al (2004) Signal peptide peptidase forms a homodimer that is labeled by an active site-directed γ-secretase inhibitor. J Biol Chem 279(15):15153–15160

    Article  CAS  PubMed  Google Scholar 

  51. Nyborg AC, Herl L, Berezovska O et al (2006) Signal peptide peptidase (SPP) dimer formation as assessed by fluorescence lifetime imaging microscopy (FLIM) in intact cells. Mol Neurodegeneration 1(1):1–8

    Article  Google Scholar 

  52. Gertsik N, Chau D-M, Li Y-M (2015) γ-secretase inhibitors and modulators induce distinct conformational changes in the active sites of γ-secretase and signal peptide peptidase. ACS Chem Biol 10(8):1925–1931

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tomita T, Watabiki T, Takikawa R et al (2001) The first proline of PALP motif at the C terminus of presenilins is obligatory for stabilization, complex formation, and gamma-secretase activities of presenilins. J Biol Chem 276(35):33273–33281

    Article  CAS  PubMed  Google Scholar 

  54. Sato C, Takagi S, Tomita T et al (2008) The C-terminal PAL motif and transmembrane domain 9 of presenilin 1 are involved in the formation of the catalytic pore of the gamma-secretase. J Neurosci 28(24):6264–6271

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Haass C, Selkoe DJ (1993) Cellular processing of beta-amyloid precursor protein and the genesis of amyloid beta-peptide. Cell 75(6):1039–1042

    Article  CAS  PubMed  Google Scholar 

  56. Sherrington R, Rogaev EI, Liang Y et al (1995) Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 375(6534):754–760

    Article  CAS  PubMed  Google Scholar 

  57. Tanzi RE, Kovacs DM, Kim TW et al (1996) The gene defects responsible for familial Alzheimer’s disease. Neurobiol Dis 3(3):159–168

    Article  CAS  PubMed  Google Scholar 

  58. Levy-Lahad E, Wasco W, Poorkaj P et al (1995) Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science 269(5226):973–977

    Article  CAS  PubMed  Google Scholar 

  59. Yu G, Nishimura M, Arawaka S et al (2000) Nicastrin modulates presenilin-mediated notch/glp-1 signal transduction and betaAPP processing. Nature 407(6800):48–54

    Article  CAS  PubMed  Google Scholar 

  60. Francis R, McGrath G, Zhang J et al (2002) aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 3(1):85–97

    Article  CAS  PubMed  Google Scholar 

  61. Goutte C, Tsunozaki M, Hale VA et al (2002) APH-1 is a multipass membrane protein essential for the Notch signaling pathway in Caenorhabditis elegans embryos. Proc Natl Acad Sci USA 99(2):775–779

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kimberly WT, LaVoie MJ, Ostaszewski BL et al (2003) Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2. Proc Natl Acad Sci USA 100(11):6382–6387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. De Strooper B (2003) Aph-1, Pen-2, and nicastrin with presenilin generate an active γ-secretase complex. Neuron 38(1):9–12

    Article  PubMed  Google Scholar 

  64. Kaether C, Haass C, Steiner H (2006) Assembly, trafficking and function of gamma-secretase. Neurodegener Dis 3(4–5):275–283

    Article  CAS  PubMed  Google Scholar 

  65. Ahn K, Shelton CC, Tian Y et al (2010) Activation and intrinsic γ-secretase activity of presenilin 1. Proc Natl Acad Sci 107(50):21435–21440

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Takasugi N, Tomita T, Hayashi I et al (2003) The role of presenilin cofactors in the gamma-secretase complex. Nature 422(6930):438–441

    Article  CAS  PubMed  Google Scholar 

  67. Wolfe MS, Xia W, Ostaszewski BL et al (1999) Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature 398(6727):513–517

    Article  CAS  PubMed  Google Scholar 

  68. Herreman A, Hartmann D, Annaert W et al (1999) Presenilin 2 deficiency causes a mild pulmonary phenotype and no changes in amyloid precursor protein processing but enhances the embryonic lethal phenotype of presenilin 1 deficiency. Proc Natl Acad Sci USA 96(21):11872–11877

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Dries DR, Yu G (2008) Assembly, maturation, and trafficking of the gamma-secretase complex in Alzheimer’s disease. Curr Alzheimer Res 5(2):132–146

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yang DS, Tandon A, Chen F et al (2002) Mature glycosylation and trafficking of nicastrin modulate its binding to presenilins. J Biol Chem 277(31):28135–28142

    Article  CAS  PubMed  Google Scholar 

  71. Kimberly WT, LaVoie MJ, Ostaszewski BL et al (2002) Complex N-linked glycosylated nicastrin associates with active gamma-secretase and undergoes tight cellular regulation. J Biol Chem 277(38):35113–35117

    Article  CAS  PubMed  Google Scholar 

  72. De Strooper B (2005) Nicastrin: gatekeeper of the gamma-secretase complex. Cell 122(3):318–320

    Article  PubMed  CAS  Google Scholar 

  73. Bolduc DM, Montagna DR, Gu Y et al (2016) Nicastrin functions to sterically hinder gamma-secretase-substrate interactions driven by substrate transmembrane domain. Proc Natl Acad Sci USA 113(5):E509–E518

    Google Scholar 

  74. Shah S, Lee SF, Tabuchi K et al (2005) Nicastrin functions as a gamma-secretase-substrate receptor. Cell 122(3):435–447

    Article  CAS  PubMed  Google Scholar 

  75. Capell A, Beher D, Prokop S et al (2005) Gamma-secretase complex assembly within the early secretory pathway. J Biol Chem 280(8):6471–6478

    Article  CAS  PubMed  Google Scholar 

  76. Lee SF, Shah S, Li H et al (2002) Mammalian APH-1 interacts with presenilin and nicastrin and is required for intramembrane proteolysis of amyloid-beta precursor protein and Notch. J Biol Chem 277(47):45013–45019

    Article  CAS  PubMed  Google Scholar 

  77. Zhang X, Li Y, Xu H et al (2014) The γ-secretase complex: from structure to function. Front Cell Neurosci 8

    Google Scholar 

  78. Hébert SS, Serneels L, Dejaegere T et al (2004) Coordinated and widespread expression of γ-secretase in vivo: evidence for size and molecular heterogeneity. Neurobiol Dis 17(2):260–272

    Article  PubMed  CAS  Google Scholar 

  79. Kaether C, Scheuermann J, Fassler M et al (2007) Endoplasmic reticulum retention of the gamma-secretase complex component Pen2 by Rer1. EMBO Rep 8(8):743–748

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Spasic D, Raemaekers T, Dillen K et al (2007) Rer1p competes with APH-1 for binding to nicastrin and regulates gamma-secretase complex assembly in the early secretory pathway. J Cell Biol 176(5):629–640

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Dang S, Wu S, Wang J et al (2015) Cleavage of amyloid precursor protein by an archaeal presenilin homologue PSH. Proc Natl Acad Sci USA 112(11):3344–3349

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Lu P, Bai XC, Ma D et al (2014) Three-dimensional structure of human gamma-secretase. Nature 512(7513):166–170

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. X-c Bai, Yan C, Yang G et al (2015) An atomic structure of human γ-secretase. Nature 525(7568):212–217

    Article  CAS  Google Scholar 

  84. Ma G, Li T, Price DL et al (2005) APH-1a is the principal mammalian APH-1 isoform present in gamma-secretase complexes during embryonic development. J Neurosci 25(1):192–198

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Mastrangelo P, Mathews PM, Chishti MA et al (2005) Dissociated phenotypes in presenilin transgenic mice define functionally distinct gamma-secretases. Proc Natl Acad Sci USA 102(25):8972–8977

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Shirotani K, Tomioka M, Kremmer E et al (2007) Pathological activity of familial Alzheimer’s disease-associated mutant presenilin can be executed by six different gamma-secretase complexes. Neurobiol Dis 27(1):102–107

    Article  CAS  PubMed  Google Scholar 

  87. Shirotani K, Edbauer D, Prokop S et al (2004) Identification of distinct gamma-secretase complexes with different APH-1 variants. J Biol Chem 279(40):41340–41345

    Article  CAS  PubMed  Google Scholar 

  88. Meckler X, Checler F (2014) Visualization of specific gamma-secretase complexes using bimolecular fluorescence complementation. J Alzheimers Dis 40(1):161–176

    Article  CAS  PubMed  Google Scholar 

  89. Sannerud R, Esselens C, Ejsmont P et al (2016) Restricted location of PSEN2/gamma-Secretase determines substrate specificity and generates an intracellular aβ pool. Cell

    Google Scholar 

  90. Acx H, Chavez-Gutierrez L, Serneels L et al (2014) Signature amyloid beta profiles are produced by different gamma-secretase complexes. J Biol Chem 289(7):4346–4355

    Article  CAS  PubMed  Google Scholar 

  91. Bayer TA, Wirths O, Majtenyi K et al (2001) Key factors in Alzheimer’s disease: beta-amyloid precursor protein processing, metabolism and intraneuronal transport. Brain Pathol 11(1):1–11

    Article  CAS  PubMed  Google Scholar 

  92. Gouras GK, Tampellini D, Takahashi RH et al (2010) Intraneuronal beta-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta Neuropathol 119(5):523–541

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Pensalfini A, Albay R 3rd, Rasool S et al (2014) Intracellular amyloid and the neuronal origin of Alzheimer neuritic plaques. Neurobiol Dis 71:53–61

    Article  CAS  PubMed  Google Scholar 

  94. St George-Hyslop P, Fraser PE (2012) Assembly of the presenilin gamma-/epsilon-secretase complex. J Neurochem 120(1):84–88

    Article  PubMed  CAS  Google Scholar 

  95. Villa JC, Chiu D, Brandes AH et al (2014) Nontranscriptional role of Hif-1alpha in activation of gamma-secretase and notch signaling in breast cancer. Cell Rep 8(4):1077–1092

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wang X, Huang T, Zhao Y et al (2014) Sorting nexin 27 regulates Abeta production through modulating gamma-secretase activity. Cell Rep 9(3):1023–1033

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Yan R, McCarthy JV (2010) Presenilin and γ-secretase activity: a viable therapeutic target for Alzheimer’s disease? Curr Signal Transduct Ther 5(2):128–140

    Article  CAS  Google Scholar 

  98. Yan R, Farrelly S, McCarthy JV (2013) Presenilins are novel substrates for TRAF6-mediated ubiquitination. Cell Signal 25(9):1769–1779

    Article  CAS  PubMed  Google Scholar 

  99. Duggan SP, Yan R, McCarthy JV (2015) A ubiquitin-binding CUE domain in presenilin-1 enables interaction with K63-linked polyubiquitin chains. FEBS Lett 589(9):1001–1008

    Article  CAS  PubMed  Google Scholar 

  100. Uemura K, Kuzuya A, Shimozono Y et al (2007) GSK3beta activity modifies the localization and function of presenilin 1. J Biol Chem 282(21):15823–15832

    Article  CAS  PubMed  Google Scholar 

  101. Massey LK, Mah AL, Monteiro MJ (2005) Ubiquilin regulates presenilin endoproteolysis and modulates gamma-secretase components, Pen-2 and nicastrin. Biochem J 391(3):513–525

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Li J, Pauley AM, Myers RL et al (2002) SEL-10 interacts with presenilin 1, facilitates its ubiquitination, and alters A-beta peptide production. J Neurochem 82(6):1540–1548

    Article  CAS  PubMed  Google Scholar 

  103. Nunan J, Shearman MS, Checler F et al (2001) The C-terminal fragment of the Alzheimer’s disease amyloid protein precursor is degraded by a proteasome-dependent mechanism distinct from gamma-secretase. Eur J Biochem 268(20):5329–5336

    Article  CAS  PubMed  Google Scholar 

  104. Suzuki T, Nakaya T (2008) Regulation of amyloid beta-protein precursor by phosphorylation and protein interactions. J Biol Chem 283(44):29633–29637

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Rebelo S, Domingues SC, Santos M et al (2013) Identification of a novel complex AbetaPP:Fe65:PP1 that regulates AbetaPP Thr668 phosphorylation levels. J Alzheimers Dis 35(4):761–775

    Article  PubMed  CAS  Google Scholar 

  106. Lee M-S, Kao S-C, Lemere CA et al (2003) APP processing is regulated by cytoplasmic phosphorylation. The Journal of Cell Biology 163(1):83–95

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Iijima K, Ando K, Takeda S et al (2000) Neuron-specific phosphorylation of Alzheimer’s beta-amyloid precursor protein by cyclin-dependent kinase 5. J Neurochem 75(3):1085–1091

    Article  CAS  PubMed  Google Scholar 

  108. Suzuki N, Cheung TT, Cai XD et al (1994) An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science 264(5163):1336–1340

    Article  CAS  PubMed  Google Scholar 

  109. Aplin AE, Gibb GM, Jacobsen JS et al (1996) In vitro phosphorylation of the cytoplasmic domain of the amyloid precursor protein by glycogen synthase kinase-3beta. J Neurochem 67(2):699–707

    Article  CAS  PubMed  Google Scholar 

  110. Standen CL, Brownlees J, Grierson AJ et al (2001) Phosphorylation of thr668 in the cytoplasmic domain of the Alzheimer’s disease amyloid precursor protein by stress-activated protein kinase 1b (Jun N-terminal kinase-3). J Neurochem 76(1):316–320

    Article  CAS  PubMed  Google Scholar 

  111. Taru H, Iijima K, Hase M et al (2002) Interaction of Alzheimer’s beta-amyloid precursor family proteins with scaffold proteins of the JNK signaling cascade. J Biol Chem 277(22):20070–20078

    Article  CAS  PubMed  Google Scholar 

  112. Vingtdeux V, Hamdane M, Gompel M et al (2005) Phosphorylation of amyloid precursor carboxy-terminal fragments enhances their processing by a gamma-secretase-dependent mechanism. Neurobiol Dis 20(2):625–637

    Article  CAS  PubMed  Google Scholar 

  113. Chang K-A, Kim H-S, Ha T-Y et al (2006) Phosphorylation of amyloid precursor protein (APP) at Thr668 regulates the nuclear translocation of the APP intracellular domain and induces neurodegeneration. Mol Cell Biol 26(11):4327–4338

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Parra LM, Hartmann M, Schubach S et al (2015) Distinct intracellular domain substrate modifications selectively regulate ectodomain cleavage of NRG1 or CD44. Mol Cell Biol 35(19):3381–3395

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Annaert W, De Strooper B (2002) A cell biological perspective on Alzheimer’s disease. Annu Rev Cell Dev Biol 18:25–51

    Article  CAS  PubMed  Google Scholar 

  116. Traub LM, Bonifacino JS (2013) Cargo recognition in clathrin-mediated endocytosis. Cold Spring Harb Perspect Biol 5(11):a016790

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Wakabayashi T, Craessaerts K, Bammens L et al (2009) Analysis of the [gamma]-secretase interactome and validation of its association with tetraspanin-enriched microdomains. Nat Cell Biol 11(11):1340–1346

    Article  CAS  PubMed  Google Scholar 

  118. Osenkowski P, Ye W, Wang R et al (2008) Direct and potent regulation of gamma-secretase by its lipid microenvironment. J Biol Chem 283(33):22529–22540

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Urano Y, Hayashi I, Isoo N et al (2005) Association of active gamma-secretase complex with lipid rafts. J Lipid Res 46(5):904–912

    Article  CAS  PubMed  Google Scholar 

  120. Vetrivel KS, Cheng H, Lin W et al (2004) Association of gamma-secretase with lipid rafts in post-Golgi and endosome membranes. J Biol Chem 279(43):44945–44954

    Article  CAS  PubMed  Google Scholar 

  121. Hur JY, Welander H, Behbahani H et al (2008) Active gamma-secretase is localized to detergent-resistant membranes in human brain. FEBS J 275(6):1174–1187

    Article  CAS  PubMed  Google Scholar 

  122. Vaccari T, Lu H, Kanwar R et al (2008) Endosomal entry regulates Notch receptor activation in Drosophila melanogaster. J Cell Biol 180(4):755–762

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Gupta-Rossi N, Six E, LeBail O et al (2004) Monoubiquitination and endocytosis direct gamma-secretase cleavage of activated Notch receptor. J Cell Biol 166(1):73–83

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Urra S, Escudero CA, Ramos P et al (2007) TrkA receptor activation by nerve growth factor induces shedding of the p75 neurotrophin receptor followed by endosomal gamma-secretase-mediated release of the p75 intracellular domain. J Biol Chem 282(10):7606–7615

    Article  CAS  PubMed  Google Scholar 

  125. Chhibber-Goel J, Coleman-Vaughan C, Agrawal V et al (2016) γ-secretase activity is required for regulated intramembrane proteolysis of tumor necrosis factor (TNF) receptor 1 and TNF-mediated pro-apoptotic signaling. J Biol Chem

    Article  CAS  Google Scholar 

  126. Peric A, Annaert W (2015) Early etiology of Alzheimer’s disease: tipping the balance toward autophagy or endosomal dysfunction? Acta Neuropathol 129(3):363–381

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ueda N, Tomita T, Yanagisawa K et al (2016) Retromer and Rab2-dependent trafficking mediate PS1 degradation by proteasomes in endocytic disturbance. J Neurochem 137(4):647–658

    Article  CAS  PubMed  Google Scholar 

  128. St George-Hyslop PH (2000) Genetic factors in the genesis of Alzheimer’s disease. Ann NY Acad Sci 924:1–7

    Article  CAS  PubMed  Google Scholar 

  129. Godyń J, Jończyk J, Panek D et al (2016) Therapeutic strategies for Alzheimer’s disease in clinical trials. Pharmacol Rep 68(1):127–138

    Article  PubMed  CAS  Google Scholar 

  130. De Strooper B, Chavez Gutierrez L (2015) Learning by failing: ideas and concepts to tackle gamma-secretases in Alzheimer’s disease and beyond. Annu Rev Pharmacol Toxicol 55:419–437

    Article  PubMed  CAS  Google Scholar 

  131. De Strooper B (2014) Lessons from a failed γ-secretase Alzheimer trial. Cell 159(4):721–726

    Article  PubMed  CAS  Google Scholar 

  132. Vassar R, Bennett BD, Babu-Khan S et al (1999) β-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286(5440):735–741

    Article  CAS  PubMed  Google Scholar 

  133. Crump CJ, Johnson DS, Li Y-M (2013) Development and mechanism of γ-secretase modulators for Alzheimer’s disease. Biochemistry 52(19):3197–3216

    Article  CAS  PubMed  Google Scholar 

  134. Golde TE, Koo EH, Felsenstein KM et al (1828) γ-secretase inhibitors and modulators. Biochimica et Biophysica Acta (BBA)—Biomembranes 12:2898–2907 (2013)

    Google Scholar 

  135. Josien H (2002) Recent advances in the development of gamma-secretase inhibitors. Curr Opin Drug Discov Devel 5(4):513–525

    CAS  PubMed  Google Scholar 

  136. Kreft AF, Martone R, Porte A (2009) Recent advances in the identification of gamma-secretase inhibitors to clinically test the Abeta oligomer hypothesis of Alzheimer’s disease. J Med Chem 52(20):6169–6188

    Article  CAS  PubMed  Google Scholar 

  137. Karran E, Hardy J (2014) Antiamyloid therapy for Alzheimer’s disease—are we on the right road? N Engl J Med 370(4):377–378

    Article  CAS  PubMed  Google Scholar 

  138. Doody RS, Raman R, Farlow M et al (2013) A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med 369(4):341–350

    Article  CAS  PubMed  Google Scholar 

  139. Henley DB, Sundell KL, Sethuraman G et al (2014) Safety profile of semagacestat, a gamma-secretase inhibitor: identity trial findings. Curr Med Res Opin 30(10):2021–2032

    Article  CAS  PubMed  Google Scholar 

  140. Coric V, van Dyck CH, Salloway S et al (2012) Safety and tolerability of the γ-secretase inhibitor avagacestat in a phase 2 study of mild to moderate alzheimer disease. Arch Neurol 69(11):1430–1440

    Article  PubMed  Google Scholar 

  141. Albright CF, Dockens RC, Meredith JE et al (2013) Pharmacodynamics of selective inhibition of γ-secretase by avagacestat. J Pharmacol Exp Ther 344(3):686–695

    Article  CAS  PubMed  Google Scholar 

  142. Probst G, Aubele DL, Bowers S et al (2013) Discovery of (R)-4-Cyclopropyl-7,8-difluoro-5-(4-(trifluoromethyl)phenylsulfonyl)-4,5-dihydro-1H-pyrazolo[4,3-c]quinoline (ELND006) and (R)-4-Cyclopropyl-8-fluoro-5-(6-(trifluoromethyl)pyridin-3-ylsulfonyl)-4,5-dihydro-2H-pyrazolo[4,3-c]quinoline (ELND007): metabolically stable γ-secretase inhibitors that selectively inhibit the production of amyloid-β over Notch. J Med Chem 56(13):5261–5274

    Article  CAS  PubMed  Google Scholar 

  143. Weggen S, Eriksen JL, Das P et al (2001) A subset of NSAIDs lower amyloidogenic A[beta]42 independently of cyclooxygenase activity. Nature 414(6860):212–216

    Article  CAS  PubMed  Google Scholar 

  144. Lim GP, Yang F, Chu T et al (2000) Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 20(15):5709–5714

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Das C, Berezovska O, Diehl TS et al (2003) Designed helical peptides inhibit an intramembrane protease. J Am Chem Soc 125(39):11794–11795

    Article  CAS  PubMed  Google Scholar 

  146. Mohr OL (1919) character changes caused by mutation of an entire region of a chromosome in Drosophila. Genetics 4(3):275–282

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Groeneweg JW, Foster R, Growdon WB et al (2014) Notch signaling in serous ovarian cancer. J Ovarian Res 7:95

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Hales EC, Taub JW, Matherly LH (2014) New insights into Notch1 regulation of the PI3 K-AKT-mTOR1 signaling axis: targeted therapy of gamma-secretase inhibitor resistant T-cell acute lymphoblastic leukemia. Cell Signal 26(1):149–161

    Article  CAS  PubMed  Google Scholar 

  149. Takebe N, Nguyen D, Yang SX (2014) Targeting notch signaling pathway in cancer: clinical development advances and challenges. Pharmacol Ther 141(2):140–149

    Article  CAS  PubMed  Google Scholar 

  150. Reynolds TC, Smith SD, Sklar J (1987) Analysis of DNA surrounding the breakpoints of chromosomal translocations involving the beta T cell receptor gene in human lymphoblastic neoplasms. Cell 50(1):107–117

    Article  CAS  PubMed  Google Scholar 

  151. Ellisen LW, Bird J, West DC et al (1991) TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell 66(4):649–661

    Article  CAS  PubMed  Google Scholar 

  152. Mao L (2015) NOTCH mutations: multiple faces in human malignancies. Cancer Prev Res (Phila) 8(4):259–261

    Article  CAS  Google Scholar 

  153. Mutvei AP, Fredlund E, Lendahl U (2015) Frequency and distribution of Notch mutations in tumor cell lines. BMC Cancer 15:311

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Weng AP, Ferrando AA, Lee W et al (2004) Activating mutations of Notch1 in human T cell acute lymphoblastic leukemia. Science 306(5694):269–271

    Article  CAS  PubMed  Google Scholar 

  155. Liu J, Shen JX, Wen XF et al (2016) Targeting Notch degradation system provides promise for breast cancer therapeutics. Crit Rev Oncol Hematol

    Google Scholar 

  156. Guilmeau S (2012) Notch signaling and intestinal cancer. Adv Exp Med Biol 727:272–288

    Article  CAS  PubMed  Google Scholar 

  157. Bertrand FE, Angus CW, Partis WJ et al (2012) Developmental pathways in colon cancer: crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle 11(23):4344–4351

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Avila JL, Kissil JL (2013) Notch signaling in pancreatic cancer: oncogene or tumor suppressor? Trends Mol Med 19(5):320–327

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Su Q, Xin L (2016) Notch signaling in prostate cancer: refining a therapeutic opportunity. Histol Histopathol 31(2):149–157

    CAS  PubMed  Google Scholar 

  160. Lino MM, Merlo A, Boulay JL (2010) Notch signaling in glioblastoma: a developmental drug target? BMC Med 8:72

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Weijzen S, Rizzo P, Braid M et al (2002) Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat Med 8(9):979–986

    Article  CAS  PubMed  Google Scholar 

  162. Stylianou S, Clarke RB, Brennan K (2006) Aberrant activation of notch signaling in human breast cancer. Cancer Res 66(3):1517–1525

    Article  CAS  PubMed  Google Scholar 

  163. Ranganathan P, Weaver KL, Capobianco AJ (2011) Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 11(5):338–351

    Article  CAS  PubMed  Google Scholar 

  164. Koch U, Radtke F (2007) Notch and cancer: a double-edged sword. Cell Mol Life Sci 64(21):2746–2762

    Article  CAS  PubMed  Google Scholar 

  165. Andersson ER, Lendahl U (2014) Therapeutic modulation of Notch signalling—are we there yet? Nat Rev Drug Discov 13(5):357–378

    Article  CAS  PubMed  Google Scholar 

  166. Gottlinger H, Johnson J, Riethmuller G (1986) Biochemical and epitope analysis of the 17-1A membrane antigen. Hybridoma 5(1):S29–S37

    Google Scholar 

  167. Herlyn M, Steplewski Z, Herlyn D et al (1979) Colorectal carcinoma-specific antigen: detection by means of monoclonal antibodies. Proc Natl Acad Sci USA 76(3):1438–1442

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Schnell U, Cirulli V (1828) Giepmans BN (2013) EpCAM: structure and function in health and disease. Biochim Biophys Acta 8:1989–2001

    Google Scholar 

  169. Osta WA, Chen Y, Mikhitarian K et al (2004) EpCAM is overexpressed in breast cancer and is a potential target for breast cancer gene therapy. Cancer Res 64(16):5818–5824

    Article  CAS  PubMed  Google Scholar 

  170. Al-Hajj M, Wicha MS, Benito-Hernandez A et al (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100(7):3983–3988

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Spizzo G, Went P, Dirnhofer S et al (2004) High Ep-CAM expression is associated with poor prognosis in node-positive breast cancer. Breast Cancer Res Treat 86(3):207–213

    Article  CAS  PubMed  Google Scholar 

  172. Hachmeister M, Bobowski KD, Hogl S et al (2013) Regulated intramembrane proteolysis and degradation of murine epithelial cell adhesion molecule mEpCAM. PLoS ONE 8(8):e71836

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Denzel S, Maetzel D, Mack B et al (2009) Initial activation of EpCAM cleavage via cell-to-cell contact. BMC Cancer 9:402

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Nubel T, Preobraschenski J, Tuncay H et al (2009) Claudin-7 regulates EpCAM-mediated functions in tumor progression. Mol Cancer Res 7(3):285–299

    Article  PubMed  CAS  Google Scholar 

  175. Kuhn S, Koch M, Nubel T et al (2007) A complex of EpCAM, claudin-7, CD44 variant isoforms, and tetraspanins promotes colorectal cancer progression. Mol Cancer Res 5(6):553–567

    Article  CAS  PubMed  Google Scholar 

  176. van der Gun BT, Melchers LJ, Ruiters MH et al (2010) EpCAM in carcinogenesis: the good, the bad or the ugly. Carcinogenesis 31(11):1913–1921

    Article  PubMed  CAS  Google Scholar 

  177. Patriarca C, Macchi RM, Marschner AK et al (2012) Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev 38(1):68–75

    Article  CAS  PubMed  Google Scholar 

  178. Gao J, Liu X, Yang F et al (2015) By inhibiting Ras/Raf/ERK and MMP-9, knockdown of EpCAM inhibits breast cancer cell growth and metastasis. Oncotarget 6(29):27187–27198

    PubMed  PubMed Central  Google Scholar 

  179. Gilboa-Geffen A, Hamar P, Le MT et al (2015) Gene knockdown by EpCAM Aptamer-siRNA chimeras suppresses epithelial breast cancers and their tumor-initiating cells. Mol Cancer Ther 14(10):2279–2291

    Article  CAS  PubMed  Google Scholar 

  180. Sankpal NV, Mayfield JD, Willman MW et al (2011) Activator protein 1 (AP-1) contributes to EpCAM-dependent breast cancer invasion. Breast Cancer Res 13(6):R124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Gao J, Yan Q, Liu S et al (2014) Knockdown of EpCAM enhances the chemosensitivity of breast cancer cells to 5-fluorouracil by downregulating the antiapoptotic factor Bcl-2. PLoS ONE 9(7):e102590

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Ralhan R, Cao J, Lim T et al (2010) EpCAM nuclear localization identifies aggressive thyroid cancer and is a marker for poor prognosis. BMC Cancer 10:331

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. He HC, Kashat L, Kak I et al (2012) An Ep-ICD based index is a marker of aggressiveness and poor prognosis in thyroid carcinoma. PLoS ONE 7(9):e42893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Kunavisarut T, Kak I, Macmillan C et al (2012) Immunohistochemical analysis based Ep-ICD subcellular localization index (ESLI) is a novel marker for metastatic papillary thyroid microcarcinoma. BMC Cancer 12:523

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Chopra A (2004) 64Cu-Labeled 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated IRDye 800CW (a near-infrared fluorescence dye) coupled to mAb7, an anti-epithelial cell adhesion molecule monoclonal antibody. In: Molecular imaging and contrast agent database (MICAD). Bethesda (MD)

    Google Scholar 

  186. Leung K (2004) 68 Ga-Labeled anti-EpCAM diabody against epithelial cell adhesion molecule. In: Molecular Imaging and Contrast Agent Database (MICAD). Bethesda (MD)

    Google Scholar 

  187. Leung K (2004) DiD-Labeled anti-EpCAM-directed NK-92-scFv(MOC31) zeta cells. In: Molecular imaging and contrast agent database (MICAD). Bethesda (MD)

    Google Scholar 

  188. Rybalov M, Ananias HJ, Hoving HD et al (2014) PSMA, EpCAM, VEGF and GRPR as imaging targets in locally recurrent prostate cancer after radiotherapy. Int J Mol Sci 15(4):6046–6061

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Kanwar JR, Roy K, Kanwar RK (2011) Chimeric aptamers in cancer cell-targeted drug delivery. Crit Rev Biochem Mol Biol 46(6):459–477

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Flatmark K, Guldvik IJ, Svensson H et al (2013) Immunotoxin targeting EpCAM effectively inhibits peritoneal tumor growth in experimental models of mucinous peritoneal surface malignancies. Int J Cancer 133(6):1497–1506

    Article  CAS  PubMed  Google Scholar 

  191. Zhu B, Wu G, Robinson H et al (2013) Tumor margin detection using quantitative NIRF molecular imaging targeting EpCAM validated by far red gene reporter iRFP. Mol Imaging Biol 15(5):560–568

    Article  PubMed  Google Scholar 

  192. Reichardt L (2006) Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci 361(1473):1545–1564

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Roux P, Barker P (2002) Neurotrophin signaling through the p75 neurotrophin receptor. Prog Neurobiol 67(3):203–233

    Article  CAS  PubMed  Google Scholar 

  194. Hamanoue M, Middleton G, Wyatt S et al (1999) p75-mediated NF-kappaB activation enhances the survival response of developing sensory neurons to nerve growth factor. Mol Cell Neurosci 14(1):28–40

    Article  CAS  PubMed  Google Scholar 

  195. Roux P, Bhakar A, Kennedy T et al (2001) The p75 neurotrophin receptor activates Akt (protein kinase B) through a phosphatidylinositol 3-kinase-dependent pathway. J Biol Chem 276(25):23097–23104

    Article  CAS  PubMed  Google Scholar 

  196. Weskamp G, Schlöndorff J, Lum L et al (2004) Evidence for a critical role of the tumor necrosis factor alpha convertase (TACE) in ectodomain shedding of the p75 neurotrophin receptor (p75NTR). J Biol Chem 279(6):4241–4249

    Article  CAS  PubMed  Google Scholar 

  197. Kanning K, Hudson M, Amieux P et al (2003) Proteolytic processing of the p75 neurotrophin receptor and two homologs generates C-terminal fragments with signaling capability. J Neurosci: Official J Soc Neurosci 23(13):5425–5436

    Article  CAS  Google Scholar 

  198. Jung K-M, Tan S, Landman N et al (2003) Regulated intramembrane proteolysis of the p75 neurotrophin receptor modulates its association with the TrkA receptor. J Biol Chem 278(43):42161–42169

    Article  CAS  PubMed  Google Scholar 

  199. Parkhurst C, Zampieri N, Chao M (2010) Nuclear localization of the p75 neurotrophin receptor intracellular domain. J Biol Chem 285(8):5361–5368

    Article  CAS  PubMed  Google Scholar 

  200. Podlesniy P, Kichev A, Pedraza C et al (2006) Pro-NGF from Alzheimer’s disease and normal human brain displays distinctive abilities to induce processing and nuclear translocation of intracellular domain of p75NTR and apoptosis. Am J Pathol 169(1):119–131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Kenchappa R, Zampieri N, Chao M et al (2006) Ligand-dependent cleavage of the P75 neurotrophin receptor is necessary for NRIF nuclear translocation and apoptosis in sympathetic neurons. Neuron 50(2):219–232

    Article  CAS  PubMed  Google Scholar 

  202. Domeniconi M, Zampieri N, Spencer T et al (2005) MAG induces regulated intramembrane proteolysis of the p75 neurotrophin receptor to inhibit neurite outgrowth. Neuron 46(6):849–855

    Article  CAS  PubMed  Google Scholar 

  203. Krygier S, Djakiew D (2002) Neurotrophin receptor p75(NTR) suppresses growth and nerve growth factor-mediated metastasis of human prostate cancer cells. Int J Cancer 98(1):1–7

    Article  CAS  PubMed  Google Scholar 

  204. Arrighi N, Bodei S, Zani D et al (2010) Nerve growth factor signaling in prostate health and disease. Growth Factors 28(3):191–201

    Article  CAS  PubMed  Google Scholar 

  205. Marchetti D, Aucoin R, Blust J et al (2004) p75 neurotrophin receptor functions as a survival receptor in brain-metastatic melanoma cells. J Cell Biochem 91(1):206–215

    Article  CAS  PubMed  Google Scholar 

  206. Marchetti D, Mrak R, Paulsen D et al (2007) Neurotrophin receptors and heparanase: a functional axis in human medulloblastoma invasion. J Exp Clin Cancer Res CR 26(1):5–23

    CAS  PubMed  Google Scholar 

  207. Boiko A, Razorenova O, van de Rijn M et al (2010) Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 466(7302):133–137

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Denkins Y, Reiland J, Roy M et al (2004) Brain metastases in melanoma: roles of neurotrophins. Neuro-oncology 6(2):154–165

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Johnston A, Lun X, Rahn J et al (2007) The p 75 neurotrophin receptor is a central regulator of glioma invasion. PLoS Bio l5(8)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Wang X, Cui M, Wang L et al (2010) Inhibition of neurotrophin receptor p75 intramembran proteolysis by gamma-secretase inhibitor reduces medulloblastoma spinal metastasis. Biochem Biophys Res Commun 403(3–4):264–269

    Article  CAS  PubMed  Google Scholar 

  211. Forsyth PA, Krishna N, Lawn S et al (2014) p75 neurotrophin receptor cleavage by alpha- and gamma-secretases is required for neurotrophin-mediated proliferation of brain tumor-initiating cells. J Biol Chem 289(12):8067–8085

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Descamps S, Toillon R, Adriaenssens E et al (2001) Nerve growth factor stimulates proliferation and survival of human breast cancer cells through two distinct signaling pathways. J Biol Chem 276(21):17864–17870

    Article  CAS  PubMed  Google Scholar 

  213. Verbeke S, Meignan S, Lagadec C et al (2010) Overexpression of p75(NTR) increases survival of breast cancer cells through p21(waf1). Cell Signal 22(12):1864–1873

    Article  CAS  PubMed  Google Scholar 

  214. Wang L, Rahn J, Lun X et al (2008) Gamma-secretase represents a therapeutic target for the treatment of invasive glioma mediated by the p 75 neurotrophin receptor. PLoS Biol 6(11)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  215. Zhou J, Jain S, Azad AK et al (2016) Notch and TGFbeta form a positive regulatory loop and regulate EMT in epithelial ovarian cancer cells. Cell Signal 28(8):838–849

    Article  CAS  PubMed  Google Scholar 

  216. Dinicola S, Pasqualato A, Proietti S et al (2016) Paradoxical E-cadherin increase in 5FU-resistant colon cancer is unaffected during mesenchymal-epithelial reversion induced by gamma-secretase inhibition. Life Sci 145:174–183

    Article  CAS  PubMed  Google Scholar 

  217. Rosati E, Sabatini R, De Falco F et al (2013) gamma-Secretase inhibitor I induces apoptosis in chronic lymphocytic leukemia cells by proteasome inhibition, endoplasmic reticulum stress increase and notch down-regulation. Int J Cancer 132(8):1940–1953

    Article  CAS  PubMed  Google Scholar 

  218. Zou YH, Cao YQ, Wang LX et al (2011) γ-secretase inhibitor up-regulates vascular endothelial growth factor receptor-2 and endothelial nitric oxide synthase. Exp Ther Med 2(4):725–729

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Kalantari E, Saeidi H, Kia NS et al (2013) Effect of DAPT, a gamma secretase inhibitor, on tumor angiogenesis in control mice. Adv Biomed Res 2:83

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  220. Maraver A, Fernandez-Marcos PJ, Herranz D et al (2012) Therapeutic effect of gamma-secretase inhibition in KrasG12V-driven non-small cell lung carcinoma by derepression of DUSP1 and inhibition of ERK. Cancer Cell 22(2):222–234

    Article  CAS  PubMed  Google Scholar 

  221. Cullion K, Draheim KM, Hermance N et al (2009) Targeting the Notch1 and mTOR pathways in a mouse T-ALL model. Blood 113(24):6172–6181

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Engin F, Bertin T, Ma O et al (2009) Notch signaling contributes to the pathogenesis of human osteosarcomas. Hum Mol Genet 18(8):1464–1470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Meng RD, Shelton CC, Li Y-M et al (2009) γ-secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Can Res 69(2):573–582

    Article  CAS  Google Scholar 

  224. Yuan X, Wu H, Xu H et al (2015) Notch signaling: an emerging therapeutic target for cancer treatment. Cancer Lett 369(1):20–27

    Article  CAS  PubMed  Google Scholar 

  225. Kumar R, Juillerat-Jeanneret L, Golshayan D (2016) Notch antagonists: potential modulators of cancer and inflammatory diseases. J Med Chem

    Article  CAS  PubMed  Google Scholar 

  226. De Jesus-Acosta A, Laheru D, Maitra A et al (2014) A phase II study of the gamma secretase inhibitor RO4929097 in patients with previously treated metastatic pancreatic adenocarcinoma. Invest New Drugs 32(4):739–745

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Lee SM, Moon J, Redman BG et al (2015) Phase 2 study of RO4929097, a gamma-secretase inhibitor, in metastatic melanoma: SWOG 0933. Cancer 121(3):432–440

    Article  CAS  PubMed  Google Scholar 

  228. Papayannidis C, DeAngelo DJ, Stock W et al (2015) A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma. Blood Cancer J 5:e350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Messersmith WA, Shapiro GI, Cleary JM et al (2015) A Phase I, dose-finding study in patients with advanced solid malignancies of the oral gamma-secretase inhibitor PF-03084014. Clin Cancer Res 21(1):60–67

    Article  CAS  PubMed  Google Scholar 

  230. Fouladi M, Stewart CF, Olson J et al (2011) Phase I trial of MK-0752 in children with refractory CNS malignancies: a pediatric brain tumor consortium study. J Clin Oncol 29(26):3529–3534

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Krop I, Demuth T, Guthrie T et al (2012) Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors. J Clin Oncol 30(19):2307–2313

    Article  CAS  PubMed  Google Scholar 

  232. Piha-Paul SA, Munster PN, Hollebecque A et al (2015) Results of a phase 1 trial combining ridaforolimus and MK-0752 in patients with advanced solid tumours. Eur J Cancer 51(14):1865–1873

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Hoffman LM, Fouladi M, Olson J et al (2015) Phase I trial of weekly MK-0752 in children with refractory central nervous system malignancies: a pediatric brain tumor consortium study. Childs Nerv Syst 31(8):1283–1289

    Article  PubMed  PubMed Central  Google Scholar 

  234. LoConte NK, Razak AR, Ivy P et al (2015) A multicenter phase 1 study of gamma -secretase inhibitor RO4929097 in combination with capecitabine in refractory solid tumors. Invest New Drugs 33(1):169–176

    Article  CAS  PubMed  Google Scholar 

  235. Diaz-Padilla I, Hirte H, Oza AM et al (2013) A phase Ib combination study of RO4929097, a gamma-secretase inhibitor, and temsirolimus in patients with advanced solid tumors. Invest New Drugs 31(5):1182–1191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Richter S, Bedard PL, Chen EX et al (2014) A phase I study of the oral gamma secretase inhibitor R04929097 in combination with gemcitabine in patients with advanced solid tumors (PHL-078/CTEP 8575). Invest New Drugs 32(2):243–249

    Article  CAS  PubMed  Google Scholar 

  237. Tolcher AW, Messersmith WA, Mikulski SM et al (2012) Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol 30(19):2348–2353

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. McAuliffe SM, Morgan SL, Wyant GA et al (2012) Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy. Proc Natl Acad Sci USA 109(43):E2939–E2948

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Singh A, Zapata MC, Choi YS et al (2014) GSI promotes vincristine-induced apoptosis by enhancing multi-polar spindle formation. Cell Cycle 13(1):157–166

    Article  CAS  PubMed  Google Scholar 

  240. Woorons X, Mollard P, Pichon A et al (2008) Effects of a 4-week training with voluntary hypoventilation carried out at low pulmonary volumes. Respir Physiol Neurobiol 160(2):123–130

    Article  PubMed  Google Scholar 

  241. Arasada RR, Amann JM, Rahman MA et al (2014) EGFR blockade enriches for lung cancer stem-like cells through Notch3-dependent signaling. Cancer Res 74(19):5572–5584

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Li LC, Wang DL, Wu YZ et al (2015) Gastric tumor-initiating CD44+ cells and epithelial-mesenchymal transition are inhibited by gamma-secretase inhibitor DAPT. Oncol Lett 10(5):3293–3299

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Ni J, Cozzi P, Hao J et al (2013) Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway. Int J Biochem Cell Biol 45(12):2736–2748

    Article  CAS  PubMed  Google Scholar 

  244. Yahyanejad S, Theys J, Vooijs M (2016) Targeting Notch to overcome radiation resistance. Oncotarget 7(7):7610–7628

    Article  PubMed  Google Scholar 

  245. Huang X, Qian Y, Wu H et al (2015) Aberrant expression of osteopontin and E-cadherin indicates radiation resistance and poor prognosis for patients with cervical carcinoma. J Histochem Cytochem 63(2):88–98

    Article  PubMed  CAS  Google Scholar 

  246. Taylor IC, Hutt-Cabezas M, Brandt WD et al (2015) Disrupting Notch slows diffuse intrinsic pontine glioma growth, enhances radiation sensitivity, and shows combinatorial efficacy with bromodomain inhibition. J Neuropathol Exp Neurol 74(8):778–790

    Article  CAS  PubMed  Google Scholar 

  247. Mizugaki H, Sakakibara-Konishi J, Ikezawa Y et al (2012) γ-secretase inhibitor enhances antitumour effect of radiation in Notch-expressing lung cancer. Br J Cancer 106(12):1953–1959

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Vermezovic J, Adamowicz M, Santarpia L et al (2015) Notch is a direct negative regulator of the DNA-damage response. Nat Struct Mol Biol 22(5):417–424

    Article  CAS  PubMed  Google Scholar 

  249. Panaccione A, Chang MT, Carbone BE et al (2016) NOTCH1 and SOX10 are essential for proliferation and radiation resistance of cancer stem-like cells in adenoid cystic carcinoma. Clin Cancer Res 22(8):2083–2095

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Debeb BG, Cohen EN, Boley K et al (2012) Pre-clinical studies of Notch signaling inhibitor RO4929097 in inflammatory breast cancer cells. Breast Cancer Res Treat 134(2):495–510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Jowett PL, Nicohlson SS, Gamble GA (1986) Tissue levels of atrazine in a case of bovine poisoning. Vet Hum Toxicol 28(6):539–540

    CAS  PubMed  Google Scholar 

  252. Kanu OO, Hughes B, Di C et al (2009) Glioblastoma multiforme oncogenomics and signaling pathways. Clin Med Oncol 3:39–52

    PubMed  PubMed Central  Google Scholar 

  253. Kanu OO, Mehta A, Di C et al (2009) Glioblastoma multiforme: a review of therapeutic targets. Expert Opin Ther Targets 13(6):701–718

    Article  CAS  PubMed  Google Scholar 

  254. Yahyanejad S, King H, Iglesias VS et al (2016) NOTCH blockade combined with radiation therapy and temozolomide prolongs survival of orthotopic glioblastoma. Oncotarget

    Google Scholar 

  255. Natsumeda M, Maitani K, Liu Y et al (2015) Targeting Notch signaling and autophagy increases cytotoxicity in glioblastoma neurospheres. Brain Pathol

    Google Scholar 

  256. Lundy EG, Sorokin CF, Meltz SK et al (1977) Reversible inhibition of human peripheral lymphocyte DNA synthesis by an extract of breast cancer cell line SKBR-3. J Surg Res 22(6):654–659

    Article  CAS  PubMed  Google Scholar 

  257. Han J, Shen Q (2012) Targeting gamma-secretase in breast cancer. Breast Cancer 4:83–90

    CAS  PubMed  PubMed Central  Google Scholar 

  258. Demehri S, Turkoz A, Kopan R (2009) Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell 16(1):55–66

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Cheng YL, Choi Y, Sobey CG et al (2015) Emerging roles of the gamma-secretase-notch axis in inflammation. Pharmacol Ther 147:80–90

    Article  CAS  PubMed  Google Scholar 

  260. Saura CA (2010) Presenilin/γ-secretase and inflammation. Front Aging Neurosci 2:16

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Tournoy J, Bossuyt X, Snellinx A et al (2004) Partial loss of presenilins causes seborrheic keratosis and autoimmune disease in mice. Hum Mol Genet 13(13):1321–1331

    Article  CAS  PubMed  Google Scholar 

  262. Li T, Wen H, Brayton C et al (2007) Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. J Neurosci 27(40):10849–10859

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Maraver A, Tadokoro CE, Badura ML et al (2007) Effect of presenilins in the apoptosis of thymocytes and homeostasis of CD8+T cells. Blood 110(9):3218–3225

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Yagi T, Giallourakis C, Mohanty S et al (2008) Defective signal transduction in B lymphocytes lacking presenilin proteins. Proc Natl Acad Sci USA 105(3):979–984

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Beglopoulos V, Sun X, Saura CA et al (2004) Reduced β-amyloid production and increased inflammatory responses in presenilin conditional knock-out mice. J Biol Chem 279(45):46907–46914

    Article  CAS  PubMed  Google Scholar 

  266. Dong S, Li C, Wu P et al (2007) Environment enrichment rescues the neurodegenerative phenotypes in presenilins-deficient mice. Eur J Neurosci 26(1):101–112

    Article  PubMed  Google Scholar 

  267. Jiang X, Zhang D, Shi J et al (2009) Increased inflammatory response both in brain and in periphery in presenilin 1 and presenilin 2 conditional double knock-out mice. J Alzheimers Dis 18(3):515–523

    Article  CAS  PubMed  Google Scholar 

  268. Jayadev S, Case A, Eastman AJ et al (2010) Presenilin 2 is the predominant γ-secretase in microglia and modulates cytokine release. PLoS ONE 5(12):e15743

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Parks WC, Wilson CL, Lopez-Boado YS (2004) Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 4(8):617–629

    Article  CAS  PubMed  Google Scholar 

  270. Garton KJ, Gough PJ, Raines EW (2006) Emerging roles for ectodomain shedding in the regulation of inflammatory responses. J Leukoc Biol 79(6):1105–1116

    Article  CAS  PubMed  Google Scholar 

  271. Murphy G, Murthy A, Khokha R (2008) Clipping, shedding and RIPping keep immunity on cue. Trends Immunol 29(2):75–82

    Article  CAS  PubMed  Google Scholar 

  272. Lleo A, Saura CA (2011) gamma-secretase substrates and their implications for drug development in Alzheimer’s disease. Curr Top Med Chem 11(12):1513–1527

    Article  CAS  PubMed  Google Scholar 

  273. Carter C (2011) Alzheimer’s disease: APP, Gamma Secretase, APOE, CLU, CR1, PICALM, ABCA7, BIN1, CD2AP, CD33, EPHA1, and MS4A2, and their relationships with herpes simplex, C. Pneumoniae, Other suspect pathogens, and the immune system. Int J Alzheimers Dis 2011:501862

    PubMed  PubMed Central  Google Scholar 

  274. Garlind A, Brauner A, Hojeberg B et al (1999) Soluble interleukin-1 receptor type II levels are elevated in cerebrospinal fluid in Alzheimer’s disease patients. Brain Res 826(1):112–116

    Article  CAS  PubMed  Google Scholar 

  275. Chalaris A, Gewiese J, Paliga K et al (1803) ADAM17-mediated shedding of the IL6R induces cleavage of the membrane stub by gamma-secretase. Biochim Biophys Acta 2:234–245 (2010)

    Google Scholar 

  276. Glenn G, van der Geer P (2008) Toll-like receptors stimulate regulated intramembrane proteolysis of the CSF-1 receptor through Erk activation. FEBS Lett 582(6):911–915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Agrawal V, Sawhney N, Hickey E et al (2015) Loss of presenilin 2 function is associated with defective LPS-mediated innate immune responsiveness. Mol Neurobiol 53(5):3428–3438

    Article  PubMed  CAS  Google Scholar 

  278. Hickman SE, El Khoury J (2014) TREM2 and the neuroimmunology of Alzheimer’s disease. Biochem Pharmacol 88(4):495–498

    Article  CAS  PubMed  Google Scholar 

  279. Wang Y, Ulland TK, Ulrich JD et al (2016) TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med 213(5):667–675

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  280. Yuan P, Condello C, Keene CD et al (2016) TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90(4):724–739

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Glebov K, Wunderlich P, Karaca I et al (2016) Functional involvement of γ-secretase in signaling of the triggering receptor expressed on myeloid cells-2 (TREM2). J Neuroinflammation 13(1):1–7

    Article  CAS  Google Scholar 

  282. Schulte A, Schulz B, Andrzejewski MG et al (2007) Sequential processing of the transmembrane chemokines CX3CL1 and CXCL16 by alpha- and gamma-secretases. Biochem Biophys Res Commun 358(1):233–240

    Article  CAS  PubMed  Google Scholar 

  283. Mambole A, Baruch D, Nusbaum P et al (2008) The cleavage of neutrophil leukosialin (CD43) by cathepsin G releases its extracellular domain and triggers its intramembrane proteolysis by presenilin/gamma-secretase. J Biol Chem 283(35):23627–23635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Pelletier L, Guillaumot P, Freche B et al (2006) Gamma-secretase-dependent proteolysis of CD44 promotes neoplastic transformation of rat fibroblastic cells. Cancer Res 66(7):3681–3687

    Article  CAS  PubMed  Google Scholar 

  285. Fukumoto N, Shimaoka T, Fujimura H et al (2004) Critical roles of CXC chemokine ligand 16/scavenger receptor that binds phosphatidylserine and oxidized lipoprotein in the pathogenesis of both acute and adoptive transfer experimental autoimmune encephalomyelitis. J Immunol 173(3):1620–1627

    Article  CAS  PubMed  Google Scholar 

  286. Jong A, Wu CH, Shackleford GM et al (2008) Involvement of human CD44 during Cryptococcus neoformans infection of brain microvascular endothelial cells. Cell Microbiol 10(6):1313–1326

    Article  CAS  PubMed  Google Scholar 

  287. Bacsa S, Karasneh G, Dosa S et al (2011) Syndecan-1 and syndecan-2 play key roles in herpes simplex virus type-1 infection. J Gen Virol 92(Pt 4):733–743

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. de Witte L, Bobardt M, Chatterji U et al (2007) Syndecan-3 is a dendritic cell-specific attachment receptor for HIV-1. Proc Natl Acad Sci 104(49):19464–19469

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  289. Schulz JG, Annaert W, Vandekerckhove J et al (2003) Syndecan 3 intramembrane proteolysis is presenilin/gamma-secretase-dependent and modulates cytosolic signaling. J Biol Chem 278(49):48651–48657

    Article  CAS  PubMed  Google Scholar 

  290. Persson BD, Schmitz NB, Santiago C et al (2010) Structure of the extracellular portion of CD46 provides insights into its interactions with complement proteins and pathogens. PLoS Pathog 6(9):e1001122

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  291. Weyand NJ, Calton CM, Higashi DL et al (2010) Presenilin/gamma-secretase cleaves CD46 in response to Neisseria infection. J Immunol 184(2):694–701

    Article  CAS  PubMed  Google Scholar 

  292. Tsao PN, Wei SC, Huang MT et al (2011) Lipopolysaccharide-induced Notch signaling activation through JNK-dependent pathway regulates inflammatory response. J Biomed Sci 18:56

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Zhang W, Zhang X, Sheng A, Weng C, Zhu T, Zhao W, Li C (2015) γ-secretase inhibitor alleviates acute airway inflammation of allergic asthma in mice by downregulating Th17 cell differentiation. Mediators of Inflammation 2015 (2015):258168

    Google Scholar 

  294. Varfolomeev E, Goncharov T, Vucic D (2015) Roles of c-IAP proteins in TNF receptor family activation of NF-kappaB signaling. Methods Mol Biol 1280:269–282

    Article  CAS  PubMed  Google Scholar 

  295. Muppidi JR, Tschopp J, Siegel RM (2004) Life and death decisions: secondary complexes and lipid rafts in TNF receptor family signal transduction. Immunity 21(4):461–465

    Article  CAS  PubMed  Google Scholar 

  296. Tchikov V, Bertsch U, Fritsch J et al (2011) Subcellular compartmentalization of TNF receptor-1 and CD95 signaling pathways. Eur J Cell Biol 90(6–7):467–475

    Article  CAS  PubMed  Google Scholar 

  297. Schneider-Brachert W, Heigl U, Ehrenschwender M (2013) Membrane trafficking of death receptors: implications on signalling. Int J Mol Sci 14(7):14475–14503

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  298. Cabal-Hierro L, Lazo PS (2012) Signal transduction by tumor necrosis factor receptors. Cell Signal 24(6):1297–1305

    Article  CAS  PubMed  Google Scholar 

  299. Irannejad R, von Zastrow M (2014) GPCR signaling along the endocytic pathway. Curr Opin Cell Biol 27:109–116

    Article  CAS  PubMed  Google Scholar 

  300. Tsvetanova NG, Irannejad R, von Zastrow M (2015) G protein-coupled receptor (GPCR) signaling via heterotrimeric G proteins from endosomes. J Biol Chem 290(11):6689–6696

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Saleh AZ, Fang AT, Arch AE et al (2004) Regulated proteolysis of the IFNaR2 subunit of the interferon-alpha receptor. Oncogene 23(42):7076–7086

    Article  CAS  PubMed  Google Scholar 

  302. Hemming ML, Elias JE, Gygi SP et al (2008) Proteomic profiling of ?-secretase substrates and mapping of substrate requirements. PLoS Biol 6(10):e257

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  303. Carey BW, Kim DY, Kovacs DM (2007) Presenilin/gamma-secretase and alpha-secretase-like peptidases cleave human MHC Class I proteins. Biochem J 401(1):121–127

    Article  CAS  PubMed  Google Scholar 

  304. Saxena MT, Schroeter EH, Mumm JS et al (2001) Murine notch homologs (N1-4) undergo presenilin-dependent proteolysis. J Biol Chem 276(43):40268–40273

    Article  CAS  PubMed  Google Scholar 

  305. Okochi M, Steiner H, Fukumori A et al (2002) Presenilins mediate a dual intramembranous γ-secretase cleavage of Notch-1. EMBO J 21(20):5408–5416

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Shimizu K, Chiba S, Hosoya N et al (2000) Binding of Delta1, Jagged1, and Jagged2 to Notch2 rapidly induces cleavage, nuclear translocation, and hyperphosphorylation of Notch2. Mol Cell Biol 20(18):6913–6922

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  307. Wang H, Li ZY, Liu Y et al (2011) Desmoglein 2 is a receptor for adenovirus serotypes 3, 7, 11 and 14. Nat Med 17(1):96–104

    Article  PubMed  CAS  Google Scholar 

  308. May P, Reddy YK, Herz J (2002) Proteolytic processing of low density lipoprotein receptor-related protein mediates regulated release of its intracellular domain. J Biol Chem 277(21):18736–18743

    Article  CAS  PubMed  Google Scholar 

  309. Hoe HS, Rebeck GW (2005) Regulation of ApoE receptor proteolysis by ligand binding. Brain Res Mol Brain Res 137(1–2):31–39

    Article  CAS  PubMed  Google Scholar 

  310. Lee HJ, Jung KM, Huang YZ et al (2002) Presenilin-dependent gamma-secretase-like intramembrane cleavage of ErbB4. J Biol Chem 277(8):6318–6323

    Article  CAS  PubMed  Google Scholar 

  311. Tuffereau C, Bénéjean J, Blondel D et al (1998) Low-affinity nerve-growth factor receptor (P75NTR) can serve as a receptor for rabies virus. The EMBO J 17(24):7250–7259

    Article  CAS  PubMed  Google Scholar 

  312. Kim DY, Ingano LA, Kovacs DM (2002) Nectin-1alpha, an immunoglobulin-like receptor involved in the formation of synapses, is a substrate for presenilin/gamma-secretase-like cleavage. J Biol Chem 277(51):49976–49981

    Article  CAS  PubMed  Google Scholar 

  313. Tousseyn T, Thathiah A, Jorissen E et al (2009) ADAM10, the rate-limiting protease of regulated intramembrane proteolysis of Notch and other proteins, is processed by ADAMS-9, ADAMS-15, and the γ-secretase. J Biol Chem 284(17):11738–11747

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Wilke GA, Bubeck Wardenburg J (2010) Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus alpha-hemolysin-mediated cellular injury. Proc Natl Acad Sci USA 107(30):13473–13478

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We apologise to all colleagues whose work has not been discussed or cited owing to space limitations. This work was supported and funded by grants from Science Foundation Ireland (02/IN1/B218 and 09/IN.1/B2624) and a student bursary to CCV from the College of Science Engineering & Food Science, University College Cork.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Justin V. McCarthy .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2017 Springer Nature Singapore Pte Ltd.

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Coleman-Vaughan, C., Mal, A., De, A., McCarthy, J.V. (2017). The γ-Secretase Protease Complexes in Neurodegeneration, Cancer and Immunity. In: Chakraborti, S., Dhalla, N. (eds) Pathophysiological Aspects of Proteases. Springer, Singapore. https://doi.org/10.1007/978-981-10-6141-7_3

Download citation

Publish with us

Policies and ethics