Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Feb 17.
Published in final edited form as: J Alzheimers Dis. 2011;25(1):3–28. doi: 10.3233/JAD-2011-101065

The Many Substrates of Presenilin/γ-Secretase

Annakaisa Haapasalo a,*, Dora M Kovacs b
PMCID: PMC3281584  NIHMSID: NIHMS351966  PMID: 21335653

Abstract

The Alzheimer’s disease (AD)-associated amyloid-β protein precursor (AβPP) is cleaved by α-, β-, and presenilin (PS)/γ-secretases through sequential regulated proteolysis. These proteolytic events control the generation of the pathogenic amyloid-β (Aβ) peptide, which excessively accumulates in the brains of individuals afflicted by AD. A growing number of additional proteins cleaved by PS/γ-secretase continue to be discovered. Similarly to AβPP, most of these proteins are type-I transmembrane proteins involved in vital signaling functions regulating cell fate, adhesion, migration, neurite outgrowth, or synaptogenesis. All the identified proteins share common structural features, which are typical for their proteolysis. The consequences of the PS/γ-secretase-mediated cleavage on the function of many of these proteins are largely unknown. Here, we review the current literature on the proteolytic processing mediated by the versatile PS/γ-secretase complex. We begin by discussing the steps of AβPP processing and PS/γ-secretase complex composition and localization, which give clues to how and where the processing of other PS/γ-secretase substrates may take place. Then we summarize the typical features of PS/γ-secretase-mediated protein processing. Finally, we recapitulate the current knowledge on the possible physiological function of PS/γ-secretase-mediated cleavage of specific substrate proteins.

Keywords: Alzheimer’s disease, amyloid-β protein precursor, γ-secretase, presenilin, regulated intramembrane processing

INTRODUCTION

Since the original discovery that amyloid-β protein precursor (AβPP) is a substrate for presenilin (PS)-dependent γ-secretase (PS/γ-secretase), at least 90 additional proteins have been found to undergo similar proteolysis by this enzyme complex [1, 2]. PS/γ-secretase is a multiprotein complex consisting of four essential components: presenilin 1 (PS1) or presenilin 2 (PS2), nicastrin, Aph-1 (anterior pharynx defective-1), and Pen-2 (presenilin enhancer-2) [3, 4]. These four components are required for the formation of a functional PS/γ-secretase complex, even though several other proteins have been shown to also associate with the complex [3, 5]. Together with PS/γ-secretase, site-2 protease (S2P), signal peptide peptidases (SPPs), and rhomboids constitute a novel family of intramembrane cleaving proteases, I-CLiPs [6]. PS/γ-secretase typically cleaves type-I transmembrane proteins, S2P cleaves the sterol regulatory element binding protein in the Golgi membrane, and SPPs are GxGD-type aspartyl proteases cleaving type-II transmembrane proteins in membranes along the secretory pathway, endosomes, and lysosomes. Rhomboids regulate different processes, such as epidermal growth factor receptor signaling, mitochondrial dynamics, or apoptotic stimuli, even though the identities of their substrates are unknown. All the I-CLiPs enzymatically cleave their substrate proteins within the plane of a membrane in a process termed regulated intramembrane proteolysis (RIP) [68].

Amyloid-β (Aβ) peptides form plaques in the cortical brain areas of patients with Alzheimer’s disease (AD). The proteolytic processing steps involved in the cleavage of AβPP and generation of Aβ have been characterized (Fig. 1). AβPP first undergoes ectodomain shedding at two alternative sites: one cleavage is mediated by α-secretase activity of ADAM (a disintegrin and metalloproteinase) family metalloproteases, and the other by β-secretase or BACE1 (β-site AβPP cleaving enzyme 1). These cleavages result in the release of secreted AβPP ectodomains (sAβPP) from the membrane and formation of two major membrane-associated C-terminal fragments (CTFs) termed C83 and C99. The subsequent PS/γ-secretase-mediated cleavage of C83 or C99 at the ε-site releases the AβPP intracellular domain (AICD). PS/γ-secretase also cleaves at the γ-site(s), releasing either a small soluble p3 peptide in the case of C83, or alternatively, Aβ peptides of different lengths after cleavage of C99 [9].

Fig. 1.

Fig. 1

AβPP processing and Aβ degradation. AβPP is a type-I transmembrane protein undergoing sequential proteolytic processing by α- or β- and γ-secretases. The non-amyloidogenic processing (1a) is initiated by α-secretase (ADAM10 or ADAM17), which cuts AβPP within the Aβ domain and thus excludes Aβ generation. α-secretase cleavage sheds AβPP ectodomain from the membrane, releasing the soluble AβPPα (sAβPPα) and yielding the membrane-bound C83 AβPP C-terminal fragment (CTF). C83 is cleaved by γ-secretase at the ε- and γ-sites, which result in the release of AβPP intracellular domain (AICD) and a p3 peptide, respectively (2a). Alternatively, the ectodomain of AβPP may be cleaved at the N-terminus of the Aβ region by β-secretase (BACE1). This initiates the amyloidogenic processing (1b) of AβPP by releasing the soluble sAβPPβ and producing the membrane-associated C99 AβPP-CTF. C99 is further cleaved by γ-secretase at ε- and γ-sites to release AICD and Aβ (2b). Aβ peptides may be degraded (3) e.g., by Aβ-degrading enzymes IDE (insulin degrading enzyme) or NEP (neprilysin).

Over the past decade, a growing number of additional proteins have been identified as substrates for the three secretases. Similarly to AβPP, the ectodomain shedding is followed by RIP, which releases the soluble C-terminal domains of the proteins from the membrane [7]. It is intriguing that these PS/γ-secretase substrate proteins play a vast variety of functions in both developing and adult tissues. Therefore, PS/γ-secretase-mediated cleavage may regulate a large range of cellular events. Despite the diversity of the thus far characterized PS/γ-secretase substrates in their structure and physiological functions, the majority of these proteins share several common features (see below) [2, 10, 11].

The wide variety of cellular processes and signaling events regulated by PS/γ-secretase via the cleavage of its multiple substrate proteins creates challenges for the development of therapeutic γ-secretase inhibitors for AD. Thus, it will be of utmost importance to design inhibitors that would specifically target only Aβ generation and not interfere with normal processing of other PS/γ-secretase substrates. Moreover, specific targeting of PS/γ-secretase, and not the other I-CLiPs, will be essential in drug development for AD. However, this may be complicated by the fact that the substrates of the other I-CLiPs remain thus far largely unidentified. In this review, we focus on PS/γ-secretase substrates and the functional consequences of their PS/γ-secretase-mediated cleavage. The currently characterized PS/γ-secretase substrates and the suggested role of their PS/γ-secretase-mediated cleavage are summarized in Tables 1 and 2.

Table 1.

Type-I PS/γ-secretase substrate proteins and suggested roles of their PS/γ-secretase-mediated cleavage

Number Substrate Function PS/γ-secretase cleavage product Localization of cleavage product Suggested function of PS/γ-secretase cleavage References
1. Alcadein α Regulation of AβPP signaling and processing, regulation of postsynaptic Ca2+ signaling? 1. β-Alc
2. Alc-ICD
1. n.d.
2. N
1. n.d.
2. Suppression of AICD-Fe65-mediated transcription
[109, 184]
2. Alcadein γ (calsyntenin) Regulation of AβPP signaling and processing, regulation of postsynaptic Ca2+ signaling? 1. β-Alc
2. Alc-ICD
1. n.d.
2. N
1. n.d.
2. Suppression of AICD-Fe65-mediated transcription
[109, 184]
3. APLP1 Synaptogenesis 1. ALID1
2. p3-like fragment
1. C, N
2. CM
1. Transcriptional regulation with Fe65 and Tip60
2. n.d.
[29, 111, 117, 185]
4. APLP2 Neurite outgrowth 1. ALID2
2. p3-like fragment
3. Aβ-like fragment
1. C, N
2. CM
3. CM
1. Transcriptional regulation with Fe65 and Tip60
2. n.d.
3. n.d.
[29, 111, 117, 185]
5. ApoER2 Lipid metabolism ApoER2-ICD n.d. Transcriptional regulation? [186, 187]
6. AβPP Cell adhesion, neurite outgrowth, protein transport? 1. Aβ
2. AICD
3. p3
1. EC, IC
2. C, N
3. CM
1. AD pathogenesis
2. Transcriptional regulation with Fe65 and Tip60; neurodegeneration?
3. n.d.
[2426, 2830, 33, 37, 47]
7. Betacellulin (BTC) EGF-like growth factor; ErbB1 and ErbB4 ligand BTC-ICD N (nuclear membrane) Cell growth inhibition [188]
8. Betaglycan Type III TGF-β receptor Betaglycan cytoplasmic fragment n.d. Regulation of TGF-β2 signaling [189]
9. CD43 Cell-cell interaction CD43-ICD N? Regulation of CD43 signaling? [190192]
10. CD44 Cell adhesion, hyaluronan receptor 1.CD44-ICD
2. CD44-β
1. C, N
2. CM
1. Transcriptional regulation, cell transformation, macrophage fusion
2. n.d.
[119, 172, 193195]
11. CSF1R RPTK; signaling CSF1R cytoplasmic domain C, N n.d. [196]
12. CXCL16 Transmembrane chemokine; cell adhesion Smaller MW CTF n.d. n.d. [197]
13. CX3CL1 (fractalkine) Transmembrane chemokine; cell adhesion Smaller MW CTF n.d. n.d. [197]
14. DCC Netrin-1 receptor; axon guidance DCC-ICD C, N? Regulation of transcription, cAMP signaling and glutamatergic transmission and neurite outgrowth [120, 162]
15. Delta1 Notch ligand; cell fate determination DICD C, N Transcriptional regulation, antagonizing of Notch signaling [126129, 198]
16. Desmoglein-2 Structural component of desmosomes; formation of intercellular junctions, regulation of tissue morphogenesis DSG2-ICD n.d. n.d. [112]
17. DNER Delta/Notch-like EGF-related receptor; cerebellar development and function DNER-ICD n.d. n.d. [112]
18. Dystroglycan Member of dystrophin-glycoprotein complex; connects ECM with cytoskeleton DG-ICD n.d. n.d. [112]
19. E-cadherin Cell adhesion E-Cad/CTF2 C, N Disassembly of adherens junctions, regulation of p120 catenin-mediated transcription, suppression of apoptosis [130, 131, 168]
20. EpCAM Transmembrane glycoprotein expressed in human malignancies; cell adhesion EpICD C,N Regulation of β-catenin/Lef-1-mediated transcription; oncogenic [75]
21. EphA4 RPTK; regulation of dendritic spines EphA4-ICD C, N Enhancement of dendritic spine formation [167]
22. EphB2 RPTK; axon guidance, cell morphogenesis, tissue patterning, angiogenesis, synapse formation, LTP EphB2/CTF2 C Termination of EphB2 signaling [166]
23. EphrinB1 Cell-cell interaction eB1ICD C, N Regulation of ephrinB1 signaling related to process outgrowth, transcriptional regulation? [163]
24. EphrinB2 Axon guidance ephrinB2/CTF2 n.d. Regulation of Src-mediated signaling, negative regulation of ephrinB2-cleavage by PS/γ-secretase, formation of focal adhesions [164, 165]
25. ErbB4 RPTK; cell proliferation, differentiation, apoptosis, oligodendrocyte maturation and myelination s80 or E4ICD C, N Apoptosis, regulation of transcription and p53 levels, oligodendrocyte maturation, regulation of astrogenesis [137144, 173, 199]
26. GHR Growth hormone receptor GHR-ICD N Transcriptional regulation? [200]
27. HLA Immune response, T-cell development, synaptic plasticity and refinement n.d. n.d. n.d. [112]
28. HLA-A2 Immune response, T-cell development, synaptic plasticity and refinement HLA-A2 ICD n.d. n.d. [201]
29. IFNaR2 Type I interferon receptor IFNaR2-ICD N Regulation of STAT-mediated transcription [202, 203]
30. IGF-1R RPTK; insulin-like growth factor-1 receptor IGF-1R-ICD n.d. n.d. [204]
31. IL-1R1 Interleukin-1 receptor-1; NFκB signaling IL-1R1-ICD C Possible regulation of IL1β signaling by JNK [205]
32. IL-1R2 Interleukin-1 receptor-2; prevents IL-1R1 activation IL-1R2-ICD n.d. n.d. [206]
33. IL6R Interleukin-6 receptor n.d. n.d. n.d. [207]
34. IR Insulin receptor, RPTK IR-ICD C, N n.d. [208]
35. Ire1α Serine/threonine kinase, endoribonuclease, induction of UPR 60-kDa C-terminal fragment N Induction of UPR [209]
36. Ire1 β Serine/threonine kinase, endoribonuclease, induction of UPR 60-kDa C-terminal fragment N Induction of UPR [209]
37. Jagged2 Notch ligand; cell fate determination JICD C, N Antagonizing of Notch signaling; regulation of AP-1-mediated transcription [126, 127, 198]
38. KCNE1 Voltage-gated potassium (Kv) channel β-subunit KCNE1-ICD n.d. n.d. [175]
39. KCNE2 Voltage-gated potassium (Kv) channel β-subunit KCNE2-ICD n.d. n.d. [175]
40. KCNE3 Voltage-gated potassium (Kv) channel β-subunit KCNE3-ICD n.d. n.d. [175]
41. KCNE4 Voltage-gated potassium (Kv) channel β-subunit KCNE4-ICD n.d. n.d. [175]
42. Klotho Anti-aging protein Kl-ICD n.d. n.d. [210]
43. L1 Cell adhesion, neuronal migration, neurite outgrowth L1-CTF2 n.d. n.d. [211]
44. LAR RPTP; cell adhesion, neurite outgrowth, synapse formation and function, learning and memory LICD C, N Regulation of β-catenin/TCF/LEF-mediated transcription [135]
45. LRP1 (LDLR) Low-density lipoprotein receptor; endocytic receptor LRP1-ICD C, N Transcriptional regulation, negative regulation of AICD-Fe65-Tip60-mediated transcription, ischemic cell death [112, 146, 186, 212]
46. LRP1 b Endocytic receptor; tumor suppression LRP1b-ICD N Transcriptional regulation, tumor suppression [213]
47. LRP2 (megalin) Scavenging receptor; protein absorption in kidney n.d. n.d. n.d. [214]
48. LRP6 Endocytosis, activator of Wnt pathway LRP6-ICD n.d. Activation of Wnt pathway by inhibiting GSK3? [215]
49. MUC1 Oncogenic protein n.d. n.d. n.d. [216]
50. N-cadherin Cell adhesion, synapse formation and maintenance N-cad/CTF2 C, N Transcriptional regulation (CBP degradation) [133, 217]
51. Nav-β1 Voltage-gated sodium channel subunit; cell adhesion, synaptic transmission n.d. n.d. n.d. [218]
52. Nav-β2 Voltage-gated sodium channel subunit; cell adhesion, synaptic transmission β2-ICD C, N Regulation of cell adhesion and migration. Regulation of Nav function and levels of Nav1.1α subunit [108, 170]
53. Nav-β3 Voltage-gated sodium channel subunit; cell adhesion, synaptic transmission n.d. n.d. n.d. [218]
54. Nav-β4 Voltage-gated sodium channel subunit; cell adhesion, synaptic transmission n.d. n.d. n.d. [218]
55. Nectin-1α Formation of adherens junctions and synapses NE-ICD M (peripheral association) Remodeling of cell junctions [169]
56. Neuregulin-1 ErbB receptor ligand; regulation of Schwann cell proliferation and differentiation NRG1-ICD N Transcriptional regulation of PSD-95 [219, 220]
57. Neuregulin-2 ErbB receptor ligand n.d. n.d. n.d. [221]
58. Notch 1 Signaling receptor; cell fate determination, maintenance of stem cells 1. NICD
2. Nβ
1. C, N
2. CM
1. Transcriptional regulation (CSL-mediated)
2. n.d.
[1, 110, 114, 122]
59. Notch-2 Signaling receptor; cell fate determination, maintenance of stem cells NICD N Transcriptional regulation (CSL-mediated) [122, 123]
60. Notch-3 Signaling receptor; cell fate determination, maintenance of stem cells NICD N Transcriptional regulation (CSL-mediated) [122]
61. Notch-4 Signaling receptor; cell fate determination, maintenance of stem cells NICD N Transcriptional regulation (CSL-mediated) [122]
62. NPR-C Natriuretic peptide receptor C; blood pressure regulation n.d. n.d. n.d. [112]
63. NRADD Death receptor, p75NTR homolog NRICD N? n.d. [222]
64. p75NTR Low-affinity neurotrophin receptor; cell survival/death, cell migration, axon guidance p75-ICD C, N Regulation of trk-p75NTR complex formation, transcriptional regulation, nuclear entry of NRIF and apoptosis; potentiation of trk receptor signaling [148, 223227]
65. PAM Peptidylglycine α-amidating monooxygenase sf-CD (soluble fragment of the cytosolic domain) N Regulation of secretory granule biogenesis? [228]
66. PLXDC2 Plexin domain-containing protein 2; n.d. n.d. n.d. n.d. [112]
67. Polyductin (PKHD1) n.d.; mutations at PKDH1 locus cause autosomal recessive polycystic kidney disease polyductin-ICD N n.d. [229, 230]
68. Protocadherin-α4 (Pcdh-α4) Cell adhesion α4-CTF2 C, N n.d. [231]
69. Protocadherin-γ-C3 (Pcdh-γC3) Cell adhesion Pcdhγ-CTF2 (γ-ICD) N Induction of Pcdh-γ locus expression [232, 233]
70. Ptprz RPTP; learning and memory, neuronal migration, gliogenesis, myelin stability? Z-ICF C, N n.d. [234]
71. RAGE Receptor for advanced glycation end products and e.g. Aβ; transport of Aβ through BBB, neurite outgrowth, cell survival RICD C, N Induction of apoptosis? [160]
72. RPTPκ RPTP; cell adhesion, synapse formation, learning and memory RPTPκ PIC N Activation of β-catenin/TCF/LEF-mediated transcription oppositely to the FL-RPTPκ [134]
73. RPTPμ RPTP; cell adhesion, synapse formation, learning and memory RPTPμ PIC n.d. n.d. [134]
74. ROBO1 Axon guidance (repulsion) ROBO1-CTF2 N n.d. [235]
75. SorC3 Vps10 p family protein; protein sorting SorC3-ICD n.d. n.d. [236, 237]
76. SorCS1b Vps10 p family protein; protein sorting SorCS1b-ICD n.d. n.d. [236, 237]
77. SorLA (LR11) Vps10 p family protein; protein sorting, AβPP trafficking and processing, AD pathogenesis 1. SorLA-ICD
2. SorLA β
1. N
2. CM
1. Transcriptional regulation
2. n.d.
[236238]
78. Sortilin Vps10 p family protein; neurotensin receptor, protein sorting Sortilin-ICD n.d. n.d. [236, 237]
79. Syndecan-1 HSPG; neurite outgrowth, cell migration, learning and memory n.d. n.d. n.d. [112]
80. Syndecan-2 HSPG; neurite outgrowth, cell migration, learning and memory n.d. n.d. n.d. [112]
81. Syndecan-3 HSPG; neurite outgrowth, cell migration, learning and memory SICD C, N Transcriptional regulation, regulation of CASK subcellular localization [239]
82. Tie1 RPTK; blood vessel formation and maintenance Tie-ICD n.d. n.d. [240]
83. Tyrosinase Melanin synthesis n.d. n.d. Regulation of tyrosinase trafficking, melanin production and pigmentation [178]
84. TYRP1 Melanin synthesis n.d. n.d. Regulation of melanin production and pigmentation [178]
85. TYRP2 Melanin synthesis n.d. n.d. Regulation of melanin production and pigmentation [178]
86. Vasorin Binds to and attenuatesTGF-β signaling, vascular remodeling? Vasn-ICD n.d. n.d. [112]
87. VE-cadherin Cell adhesion n.d. n.d. n.d. [130]
88. VEGF-R1 Angiogenesis VEGF-R1-ICD C Inhibition of angiogenesis [171]
89. VLDLR Lipoprotein receptor VLDLR-ICD n.d. n.d. [187]

Abbreviations: Aβ, amyloid-β; AβPP, amyloid-β protein precursor; AD, Alzheimer’s disease; AICD, AβPP ICD; AP1, activator protein 1; APLP, amyloid-β protein precursor-like protein; ApoER2, apolipoprotein E receptor 2; BBB, blood brain barrier; C, cytoplasm; cAMP, cyclic adenosine monophosphate; CASK, Ca2+/calmodulin-dependent serine kinase; CBP, CREB (cAMP-responsive element binding protein) binding protein; CM, conditioned medium; CSF1R, colony-stimulating factor 1 receptor; CSL, C-promoter-binding factor/recombination signal-sequence binding protein Jκ/Suppressor-of-Hairless/Lag1; DCC, deleted in colorectal cancer; EC, extracellular; ECM, extracellular matrix; EGF, epidermal growth factor; FL, full-length; GHR, growth hormone receptor; GSK3, glycogen synthase kinase 3; HLA, human leukocyte antigen; HSPG, heparan sulfate proteoglycan; IC, intracellular; ICD, intracellular domain; ICF, intracellular fragment; IFN, interferon; IGF, insulin-like growth factor; IL, interleukin; IR, insulin receptor; Ire1, inositol requirement 1; JNK, c-jun N-terminal kinase; KCNE, voltage-gated potassium channel β-subunit; LAR; leukocyte-common antigen related; LEF, lymphoid enhancer factor; LRP, low-density lipoprotein receptor; M, membrane; N, nucleus; Nav, voltage-gated sodium channel; n.d., not determined; NPR-C, natriuretic peptide receptor C; NRADD, neurotrophin receptor alike death domain protein; NRIF, neurotrophin receptor interacting factor; NTR, neurotrophin receptor; PAM, peptidylglycine α-amidating monooxygenase; PIC, phosphatase intracellular portion; PLXDC2, plexin domain-containing protein 2; PSD-95, postsynaptic density protein-95; R, receptor; RAGE, receptor for advanced glycation end products; RPTK, receptor-like protein tyrosine kinase; RPTP, receptor-like protein tyrosine phosphatase; SorCS, sortilin-related VPS10 domain containing receptor; SorLA, sorting protein-related receptor with A-type repeats; STAT, signal transducer and activator of transcription; TCF, T-cell factor; TGF-β, transforming growth factor-β; TMD, transmembrane domain; TRE, TPA (12-O-tetradecanoyl phorbol-13-acetate) responsive element; trk, tropomyosin-related kinase; TYRP, tyrosinase-related protein; UPR, unfolded protein response; VEGF, vascular-endothelial growth factor; VLDLR, very low density lipoprotein receptor; Vps10p, vacuolar protein sorting 10 protein.

Table 2.

Atypical PS/γ-secretase substrate proteins and suggested roles of their PS/γ-secretase-mediated cleavage

Number Substrate Function PS/γ-secretase cleavage product Localization of cleavage product Suggested function of PS/γ-secretase cleavage References
1. GluR3 Glutamate receptor subunit; synaptic transmission GluR3sβ n.d. n.d. [116]
2. GnT-V Glucosaminyltransferase; tumor metastasis, angiogenesis, endothelial cell proliferation Soluble GnT-V CM Regulation of secretion of soluble GnT-V; tumor metastasis, angiogenesis? [115]

Abbreviations: CM, conditioned medium; GluR3, glutamate receptor 3; n.d., not determined.

PROTEOLYTIC PROCESSING OF AβPP, THE PIONEERING PS/γ-SECRETASE SUBSTRATE PROTEIN

AβPP is a type-I transmembrane protein suggested to play a role in cell adhesion, protein transport, synapse formation, neurite extension, and neuroprotection [1218]. Understanding the sequential proteolytic processing of AβPP by secretases is of high importance given that one of its products is the pathogenic Aβ peptide. These peptides excessively accumulate in the brains of individuals affected by AD and majorly contribute to dysfunction and degeneration of neurons that result in progressive decline of cognitive functions. The proteolytic processing of AβPP is well characterized, but factors regulating its proteolysis by the different secretases are a subject of intense study. AβPP can undergo proteolysis by two alternative routes. The non-amyloidogenic pathway is more prevalent and excludes Aβ production, whereas activation of the amyloidogenic route generates the detrimental Aβ peptides (Fig. 1).

The non-amyloidogenic proteolytic pathway takes place at or near the cell surface and is initiated by α-secretases such as the ADAM-family metalloproteases ADAM10 or ADAM17 (or TACE; tumor necrosis factor-α converting enzyme) [19, 20]. This first cleavage step releases sAβPPα, the large soluble AβPP ectodomain, with a possible neuroprotective function [21]. It also produces the membrane-anchored AβPP-CTF, C83. C83 is further cleaved by PS/γ-secretase at the AβPP ε-site to release AICD, and at the γ-site to generate a small p3 peptide [1, 2226] (Fig. 1). Whether p3 plays a physiological function is thus far unknown. AICD assembles in a complex with the adaptor proteins Fe65 and Tip60, translocates to the nucleus, and regulates the transcription of genes such as KAI1, glycogen synthase kinase-3β, neprilysin, p53, and AβPP, and apoptosis [2432]. However, AICD-mediated transcriptional regulation has been challenged by recent reports. Using pharmacological inhibition of PS/γ-secretase and PS/γ-secretase- or AβPP-deficient models, these studies have shown that AICD may not be directly required for the transcriptional control of its suggested target genes and that its interaction with Fe65 may not lead to nuclear translocation [33, 34]. Moreover, the expression of AβPP N-terminal domains alone rescued the physiological and learning deficits in AβPP knock-out mice, whereas the AβPP C-terminus was not required [35]. This further suggests that AICD-mediated gene transcription may not be physiologically essential, even though this matter remains controversial. Furthermore, it was recently suggested that AICD may account for some of the deleterious effects previously associated with Aβ as nuclear localization of AICD is perhaps enhanced during the amyloidogenic processing of AβPP [36, 37].

The amyloidogenic pathway is initiated by β-secretase or BACE1, cleaving AβPP N-terminally from the α-secretase cleavage site [3841]. This clip may take place at or near the cell surface or after AβPP internalization along the endocytic pathway. BACE1-mediated cleavage releases the soluble sAβPPβ and produces the membrane-bound C99 AβPP-CTF. Subsequent PS/γ-secretase-mediated cleavage of C99 at the γ-site(s) produces Aγ peptides of different lengths along with AICD released by cleavage at the ε-site (Fig. 1).

Aβ levels in the brain are regulated by its rate of generation, but also by its clearance or degradation by different Aβ-degrading enzymes, such as neprilysin and insulin-degrading enzyme (IDE). Their function has been recently reviewed elsewhere [42, 43]. An increase in BACE1 expression in specific AD patients results in elevated BACE1-mediated cleavage of AβPP [44]. Familial AD-associated (FAD) mutations in the PSEN genes encoding for PS1 and PS2 enhance the generation of the more fibrillogenic 42-amino acid long Aβ (Aβ42), therefore increasing the ratio of Aβ42 to the shorter, more soluble Aβ40 [22, 45]. These mutations also affect the exact cleavage site in the Aβ-region of AβPP, determining the C-terminus and length of the Aβ-peptide generated. Complete loss of PS function or lack of PS/γ-secretase activity prevents production of Aβ and results in increased levels of AβPP-CTFs [4648].

PS/γ-SECRETASE

PS/γ-secretase complex composition

PS/γ-secretase is a high molecular weight enzymatic complex, whose estimated size has been reported to vary between 250 and 2000 kDa. The components of the active γ-secretase complex were revealed by a genetic screen in Caenorhabditis elegans (C. elegans) and search for PS-binding proteins in mammalian cells. Three proteins have been identified, in addition to PS: Aph-1, a multipass transmembrane protein essential for C. elegans embryogenesis; the type-I transmembrane protein nicastrin; and Pen-2, a hairpin-structured membrane protein enhancing sel-12/PS function in C. elegans (Fig. 2). Aph-1 and Pen-2 were later found to be required for Notch signaling, PS1 accumulation, and γ-secretase activity [4956]. Reconstitution of an active PS/γ-secretase complex in Saccharomyces cerevisiae yeast, which does not endogenously express any of the four complex components, elegantly confirmed that PS, nicastrin, Aph-1, and Pen-2 were necessary and sufficient for creating a functional PS/γ-secretase complex [5].

Fig. 2.

Fig. 2

PS/γ-secretase complex. The four components of the functional PS/γ-secretase complex are shown. PS consists of nine transmembrane domains (marked 1–9) and exerts the catalytic activity of the complex. PS topology is somewhat controversial. Here it is presented according to Kornilova et al. [241]. During maturation of the γ-secretase complex, PS undergoes endoproteolysis yielding the N-terminal (NTF) and the C-terminal fragment (CTF). The active aspartate residues in transmembrane domains 6 and 7 are indicated with sparkles. Nicastrin is a type-I transmembrane glycoprotein. Aph-1 consists of seven transmembrane domains (marked 1–7), and its membrane topology is presented according to Fortna et al. [242]. Nicastrin and Aph-1 facilitate the assembly and trafficking of γ-secretase. Pen-2, which promotes PS endoproteolysis, is a hairpin-structured protein with two transmembrane domains. According to Crystal et al. [243], both its N- and C-termini are on the luminal side of the membrane.

PS contains the catalytic site of the PS/γ-secretase complex. This site consists of the critical aspartate residues at amino acid positions 257 and 385 in the 6th and 7th transmembrane domains, respectively [57, 58]. Nicastrin was initially suggested to function as a receptor using its Glu-333 to bind short N-termini of substrates generated by metalloprotease-mediated ectodomain shedding. As a second step, nicastrin was supposed to recruit the substrate proteins for PS/γ-secretase-mediated cleavage [59]. However, a more recent report indicated that mutagenesis of Glu-332 in the mouse nicastrin ectodomain (corresponding to human nicastrin Glu-333) affected PS/γ-secretase complex maturation, but did not impede substrate recognition or PS/γ-secretase activity. This refuted the idea of nicastrin functioning as a specific receptor for PS/γ-secretase substrates [60]. Furthermore, the PS1/Pen-2/Aph-1a trimeric complex lacking nicastrin can function as an active PS/γ-secretase with a substantially reduced activity and stability [61]. Recent data have implied that nicastrin directly participates in PS/γ-secretase activity through its Glu-333 [62]. Thus, nicastrin appears to stabilize PS/γ-secretase and may participate in its activity, but may not be indispensable for substrate recognition.

Increasing evidence implies that assembly of the active PS/γ-secretase complex takes place in a chronologic order. First, Aph-1 and nicastrin would form an intermediate complex. Aph-1 and Pen-2, in turn, have been suggested to function as chaperones assisting in PS/γ-secretase complex assembly, trafficking, and PS endoproteolysis [3]. In addition, Aph-1 was recently suggested to be involved in PS/γ-secretase activity and to directly associate with PS/γ-secretase substrates [63, 64]. Following the intermediate Aph-1 and nicastrin complex formation, PS1 holoprotein via its C-terminus binds to the transmembrane domain of nicastrin. Finally, Pen-2 binds to the PS transmembrane domain inducing simultaneous PS1 endoproteolysis [65]. Levels of any of the complex components affect the levels and stability of the other components, indicating that assembly of the active PS/γ-secretase complex is tightly regulated in an interdependent manner. Moreover, the four core components have been shown to exist in the complex in 1 : 1:1 : 1 stoichiometry [3, 66, 67].

The PS/γ-secretase complex composition and function are further complicated by the existence of different isoforms of Aph-1 and PS. In humans, two Aph-1 genes, APH-1a and APH-1b have been reported. Of these, the former one is also alternatively spliced to generate two isoforms [68]. In addition, PS has two forms, namely PS1 and PS2, encoded by two individual genes, PSEN1 and PSEN2, respectively [69, 70]. Different combinations of these proteins may create from four to six alternative PS/γ-secretase complexes with possibly diverse activities or subcellular localizations. PS/γ-secretase complexes containing Aph-1b were recently shown to contribute to the generation of Aβ in human brain, but not to be essential for Notch cleavage [63]. In addition, it has been suggested that PS1-containing γ-secretase complexes have a significantly higher specific activity than PS2-containing ones [71, 72]. Indeed, PS1 knock-out mice show severe developmental abnormalities and embryonic lethality, while PS2-deficient mice are viable, have a milder phenotype, and display unaltered AβPP processing [73, 74]. Interestingly though, two recent reports suggest that there are PS/γ-secretase functions that are specifically dependent on PS2, but not PS1. A novel PS/γ-secretase substrate, EpCAM, was shown as cleaved specifically by PS2-dependent γ-secretase activity [75]. Furthermore, PS2-dependent γ-secretase activity released AICD regulating aquaporin 1 expression, while PS1-dependent activity was dispensable for this function [76].

Even though PS, nicastrin, Aph-1, and Pen-2 suffice to create the functional, fully active PS/γ-secretase complex, several other proteins have also been suggested to associate with the complex. The transmembrane glycoprotein CD147 was reported to associate with the PS/γ-secretase complex in several different cell lines. CD147 decreased Aβ production and was proven dispensable for the PS/γ-secretase activity [77]. Another protein suggested to associate with and modulate PS/γ-secretase function is TMP21, a member of the p24 cargo protein family. TMP21 selectively modulated γ-, but not ε-cleavage of AβPP and suppressed Aβ generation [78]. Additional binding partners keep emerging, some of which affect Aβ production [79]. However, characterization of the PS/γ-secretase complex-associating proteins is hampered by artificial interactions during the purification process. Also, association of the interacting proteins with PS/γ-secretase complex in vivo lacks compelling evidence. It is possible that these proteins do not function as essential core components for the PS/γ-secretase activity. Instead, they may act as modulators of the PS/γ-secretase complex assembly, activity, or substrate selectivity, perhaps depending on the tissue, cell type, or subcellular compartment. The presence of loosely interacting proteins may explain the variations in the reported size of the PS/γ-secretase complex.

PS/γ-secretase complex localization and the site of PS/γ-secretase-mediated cleavage

PS/γ-secretase substrate proteins first undergo ectodomain shedding at their N-termini. As seen above, this cleavage is mediated by either α- or β-secretase. The α-secretase-mediated ectodomain shedding is thought to take place mostly at the cell surface, even though there is evidence for α-secretase-mediated cleavage of AβPP in the trans-Golgi network (TGN) [8083]. β-cleavage is suggested to occur in the Golgi apparatus or TGN, at the plasma membrane, and in endocytic compartments [40, 8488]. PS/γ-secretase complex components localize to different cellular compartments, such as plasma membrane, early and late endosomes, autophagic vacuoles, lysosomes, mitochondria, Golgi, and endoplasmic reticulum (ER), creating a number of possible cellular locations for Aβ generation [8995]. Blockade of endocytosis markedly decreases Aβ generation, implying that endocytosis of AβPP is critical for its processing [96]. Interestingly, altered neuronal endocytosis is one of the earliest neuropathological changes occurring in AD [97, 98]. In the amyloidogenic pathway, AβPP is most likely endocytosed to allow for β-cleavage at the optimal acidic pH for BACE1 function in the endocytic compartments [40, 85, 88, 99]. AβPP-CTFs may then recycle back to the plasma membrane for PS/γ-secretase cleavage or alternatively, this cleavage step may also take place in the endocytic compartments [93, 100, 101]. We have shown evidence that PS/γ-secretase activity regulates AβPP clearance specifically from the endocytic recycling compartment [100]. In summary, the current understanding denotes that amyloidogenic processing of AβPP mainly occurs in the TGN, plasma membrane, and endocytic compartments as AβPP traffics through the secretory and endocytic pathways. AβPP reaching the plasma membrane, however, is preferentially cleaved by α-secretase activity and undergoes non-amyloidogenic processing.

Little is known about the localization of PS/γ-secretase-mediated cleavage of substrate proteins other than AβPP. Given that ADAM family metalloproteases are generally responsible for ectodomain cleavage of the additional PS/γ-secretase substrates, their α-cleavage most likely takes place at the cell surface. The remaining CTFs may then be processed by PS/γ-secretase on the plasma membrane or alternatively after endocytosis [101103]. Surface expression of many substrate proteins is decreased after ectodomain shedding and their CTFs are found in intracellular compartments, suggesting that endocytosis occurred prior to PS/γ-secretase-mediated cleavage. Indeed, Notch cleavage by PS/γ-secretase requires monoubiquitination of a lysine present in the transmembrane domain at the PS/γ-secretase recognition site and endocytosis [104]. Nevertheless, further studies are warranted to fully understand the role of subcellular compartments in PS/γ-secretase-mediated cleavage of its different substrate proteins.

COMMON FEATURES OF THE CURRENTLY KNOWN PS/γ-SECRETASE SUBSTRATES

Even though PS/γ-secretase substrates are diverse in their structure, localization, and physiological functions, the majority of these proteins share several common features [2, 10, 11] (Table 3). First, most substrate proteins are type-I transmembrane proteins. Typically, they harbor a large ectodomain often containing cell adhesion molecule-like domains, a single-pass transmembrane domain, and a cytoplasmic C-terminus frequently capable of initiating or mediating intracellular signaling. Second, the characterized proteins appear to function as signaling proteins and regulate cellular events such as cell fate determination, adhesion, migration, neurite outgrowth, axon guidance, or formation and maintenance of synapses. Interestingly, many of these same events are disrupted during neurodegeneration in AD [105, 106]. Third, PS/γ-secretase preferentially cleaves membrane-bound protein stubs after ectodomain shedding of the full-length substrate [10]. α-secretase mediated ectodomain shedding can be constitutive, but it may also be induced by several stimuli, such as ligand binding, protein kinase C (PKC) activation by phorbol esters, or Ca2+ influx. Some of the currently characterized PS/γ-secretase substrate proteins, such as lipoprotein-related protein-1 (LRP) or voltage-gated sodium channel β2 subunit (Navβ2), have been shown to undergo an alternative N-terminal cleavage by BACE1 in a similar manner to AβPP [107, 108]. Fourth, the PS/γ-secretase-mediated γ-like cleavage (corresponding to the ε-cleavage in AβPP, which releases AICD) takes place at or near the boundary of the transmembrane and cytoplasmic domains. The ε-like cleavage site flanks a stretch of hydrophobic amino acid sequence rich in lysine and/or arginine residues. It appears that PS/γ-secretase cleavage is not dependent on a specific amino acid target sequence at or adjacent to the cleavage site, but rather perhaps on the conformational state of the transmembrane domain [6]. Fifth, PS/γ-secretase-mediated cleavage releases the intracellular domain (ICD) of each substrate protein to the cytosol. Several substrate ICDs translocate to the nucleus and have been suggested to play a role in transcriptional regulation in a similar manner to AICD. In addition to the released ICDs, some substrate proteins, such as alcadeins or Notch, release Aβ-like peptides as a consequence of PS/γ-secretase cleavage, even though it is not clear whether these peptides exhibit any pathogenic functions [109, 110]. Sixth, generation of the ICDs is prevented upon inhibition of PS/γ-secretase activity by pharmacological γ-secretase inhibitors or lack of functional PSs. Concomitantly, γ-secretase inhibition results in CTF accumulation in an analogous manner to the AβPP-CTFs. Seventh, the ICDs are often labile and accumulate in the presence of proteasomal inhibitors, suggesting that the proteasome at least partially contributes to their degradation. Together with the proteasome, PS/γ-secretase may therefore function as a “proteasome of the membrane” that removes the C-terminal stubs generated after ectodomain shedding. This function may also implicate a role for PS/γ-secretase as a terminator of signaling for particular substrate proteins [11]. On the other hand, some substrate ICDs, such as those of amyloid β-protein precursor-like proteins (APLP1 and APLP2), have been shown to undergo degradation by IDE in a similar manner to AICD [111].

Table 3.

Typical features of the majority of PS/γ-secretase substrate proteins

1. Type-I transmembrane proteins*
2. Undergo ectodomain shedding by α- and/or β-secretase-like activity prior to PS/γ-secretase-mediated cleavage
3. PS/γ-secretase-mediated RIP at or near the border of transmembrane and cytoplasmic domains
4. PS/γ-secretase cleavage releases substrate ICD from the membrane, some ICDs localize in the nucleus
5. Lack of functional PS/γ-secretase prevents ICD generation and accumulation of substrate CTFs
6. Many ICDs are labile and degraded by the proteasome
*

Exceptions are the glutamate receptor subunit 3 (polytopic membrane protein) and glucosaminyltransferase GnT-V (type-II transmembrane protein).

Abbreviations: ICD, intracellular domain; CTF, C-terminal fragment; RIP, regulated intramembrane proteolysis.

PHYSIOLOGICAL EFFECTS OF PS/γ-SECRETASE-MEDIATED CLEAVAGE OF ADDITIONAL SUBSTRATE PROTEINS

To date, 91 proteins, incuding AβPP, have been identified as PS/γ-secretase substrates [112, 113]. The majority of these proteins are type-I transmembrane proteins, such as Notch (Table 1) [1, 114]. In addition, one type-II membrane protein, β1,6-acetylglucosaminyltranferease (GnT-V), residing in the Golgi apparatus [115] and one polytopic transmembrane protein, the glutamate receptor subunit 3 [116], have also been reported to undergo PS/γ-secretase-dependent proteolysis (Table 2). However, understanding the functional significance and regulation of PS/γ-secretase-mediated cleavage of these atypical substrate proteins requires further studies. In terms of function, it appears that some of the ICDs released by PS/γ-secretase-mediated cleavage perform independent signaling functions apart from the function of the full-length form of the protein, such as regulation of gene transcription. On the other hand, generation of some substrate ICDs may operate as means to terminate or antagonize full-length substrate protein signaling, such as in cell adhesion or neurite outgrowth.

The thus far characterized PS/γ-secretase substrates and the suggested functional effects of their PS/γ-secretase mediated cleavage are listed in Tables 1 and 2 and discussed in some detail below.

Transcriptional regulation

The intracellular domain of AβPP forms a complex with adaptor proteins Fe65 and Tip60. Many studies have suggested that upon PS/γ-secretase cleavage this complex translocates to the nucleus and regulates gene transcription [2432]. However, recent research has contradicted these results and suggested that AICD may not be involved in transcriptional control and that it may not translocate to the nucleus, but rather it remains associated with membranes or free in the cytoplasm [33, 34]. Moreover, studies in knock-in mice suggested that secreted sAβPPα, but not the AβPP C-terminus, was required to mediate the physiological effects of AβPP [35]. Therefore, the relevance of AICD-mediated gene transcription still remains controversial. In spite of this, APLP1 and APLP2 have also been implicated in Fe65-dependent transcriptional regulation [29, 111, 117]. Together, it still appears plausible that the full-length forms of all AβPP family members may restrain Fe65 from entering the nucleus, while their ICDs interact with Fe65 and enter the nucleus to regulate transcription [25, 117, 118].

Several additional PS/γ-secretase substrate ICDs have also been implicated in transcriptional regulation, often suggested by their nuclear localization and ability to affect transcription in reporter gene assays. In some cases, the substrate ICDs have been shown to control transcription of endogenous target genes by associating with different transcription factors. Among others, specific ICDs that have been reported to affect gene transcription include the following: the ICDs of alcadeins [109]; CD44 [119]; DCC (deleted in colorectal cancer) [120]; Notch [121125] and its ligands Delta and Jagged [126129]; E- and N-cadherin [130133]; and receptor-like protein tyrosine phoshatases (RPTP) RPTPκ [134] and LAR (leukocyte-common antigen related) [135]. The confirmed or putative function of these ICDs in transcriptional regulation constitutes an independent signaling function, which may be different from the full-length forms of the proteins. Furthermore, ICD generation may serve as a regulatory switch in the signaling of these particular substrates. For example, the full-length forms of cadherins or RPTPs are intimately involved in cell adhesion, whereas their respective ICDs have been shown to translocate to the nucleus and regulate transcription [130135].

Regulation of cell fate

Notch family proteins mediate short-range signaling controlling cell-fate decisions, patterning of gene expression, and maintenance of stem cell populations during development. Notch signaling in these events requires Notch activation and subsequent PS/γ-secretase-mediated release of Notch-ICD (NICD), which regulates gene transcription [125, 136].

In addition to Notch, some other substrates have been implicated in the regulation of cell fate as well. ErbB4 cleavage by PS/γ-secretase results in the release of ErbB4-ICD (E4ICD) [137]. E4ICD has been suggested to regulate gene transcription through several transcription factors and to confer pro-apoptotic activity of ErbB4 [138142]. In addition, ErbB4 proteolysis is involved in cell fate determination in the brain. Inhibition of PS/γ-secretase was shown to block E4ICD nuclear translocation and subsequent expression of proteins associated with myelination. One report indicated that maturation of oligodendrocytes, but not neurons or astrocytes, was thereby prevented and oligodendrocytes maintained their juvenile state [143]. Sardi and colleagues [144] have shown that PS/γ-secretase-dependent nuclear signaling of ErbB4 regulates the timing of astrogenesis in vivo in the developing mouse brain. Their data suggested that PS/γ-secretase-mediated ErbB4 cleavage is important for maintaining the neuronal precursor cells in a neuro-genic state by blocking astrocytic differentiation [144].

Regulation of cell death

Another event involving PS/γ-secretase-mediated cleavage of several substrate proteins is regulation of cell death signaling. LRP1 has been previously shown to facilitate AβPP endocytosis and enhance Aβ production and its cellular uptake [145]. On the other hand, PS/γ-secretase-mediated processing of LRP1 is suggested to mediate ischemic cell death [146]. Middle cerebral artery occlusion (MCAO) in mice was shown to induce activation of PS/γ-secretase in the ischemic hemisphere and nuclear translocation of LRP1-ICD in neurons within the ischemic penumbra. This was precluded by intracortical injection of a γ-secretase inhibitor. Concomitantly, PS/γ-secretase inhibition significantly decreased MCAO-induced caspase-3 activation, diminished the volume of the ischemic lesion by 50%, and improved the neurological outcome of the mice. This led the authors to propose that PS/γ-secretase-mediated proteolysis of LRP1 potentiates cell death during cerebral ischemia [146].

Neurotrophin signaling is usually considered to promote cell survival. However, p75 neurotrophin receptor (p75NTR), another PS/γ-secretase substrate, has been widely implicated in neurotrophin-mediated death signaling in neurons [147]. It was shown that pro and mature forms of brain-derived neurotrophic factor induce PS/γ-secretase-mediated generation of p75NTR-ICD and apoptosis by causing nuclear translocation of neurotrophin receptor interacting factor (NRIF), a DNA-binding protein essential for p75NTR-mediated apoptosis. p75NTR proteolysis and NRIF nuclear translocation were also shown to take place in vivo during naturally occurring cell death of superior cervical ganglion neurons [148]. Moreover, Aβ has been shown to induce p75NTR-mediated cell death signaling [149, 150]. An early event in AD is the loss of basal forebrain cholinergic neurons, which express high levels of p75NTR. The levels of p75NTR-CTF, p75NTR-ICD, and pro-nerve growth factor (pro-NGF), another pro-neurotrophin capable of inducing p75NTR-mediated death signaling, are increased in AD brain [151156]. Furthermore, pro-NGF isolated from AD brain, but not from control brain, was shown to induce apoptosis in different cells in a PS/γ-secretase-dependent manner [156]. Together, these findings strongly point to a central role for p75NTR and its proteolysis in neuronal death.

A recently identified PS/γ-secretase substrate protein is the receptor for advanced glycation end products (RAGE). RAGE is a cell surface receptor shown to mediate Aβ transport to the brain across the blood brain barrier and potentiate Aβ-induced disturbancies in neuronal function in transgenic mice [157159]. Overexpression of RAGE-ICD (RICD) was shown to decrease cell viability, increase the number of apoptotic cells, and activate the stress-related kinases, p38 and JNK (c-Jun N-terminal kinase), implying that RICD may be involved in apoptosis [160]. Interestingly, all PS/γ-secretase substrate proteins involved in cell death signaling appear to interact with Aβ, suggesting that PS/γ-secretase-mediated cleavage may be an important factor controlling their interaction with Aβ and mediating subsequent adverse effects.

Regulation of neurite outgrowth or cell adhesion

Neurite outgrowth, axonal pathfinding, and cell adhesion are key events during development. These same events may also take place in the adult brain during neuroplasticity when new connections are established between neurons, for example during learning or in response to injury. Large body of evidence suggests that PS/γ-secretase plays a fundamental role in controlling these events by proteolytic cleavage of its substrate proteins.

DCC (deleted in colorectal cancer) functions as a cell surface receptor for netrin-1, a protein regulating cell and axonal migration during development [161]. While full-length DCC was observed to extensively induce neurite outgrowth in mouse neuroblastoma cells, DCC-ICD, released by PS/γ-secretase activity, inhibited neurite outgrowth and associated with down-regulation of DCC-mediated intracellular signaling [162].

Eph receptors and their ephrin ligands are important players in synapse formation and plasticity in the central nervous system. Recent data have demonstrated that Eph receptors and ephrin signaling are regulated by PS/γ-secretase-mediated cleavage. Similarly to DCC, PS/γ-secretase-mediated processing of ephrinB1 prevented outgrowth of actin-enriched cellular protrusions in COS cells [163]. EphB-receptor has been shown to induce proteolytic processing of another EphB ligand, ephrinB2. EphrinB2-ICD, generated as a result of ephrinB2 processing by PS/γ-secretase, was found to activate Src-mediated endothelial cell sprouting. Interestingly, ephrinB2-ICD generation was demonstrated to inhibit PS/γ-secretase-mediated processing of its own precursor, ephrinB2. This finding suggests that ephrinB2-ICD generation functions as an inhibitory feedback mechanism to limit Eph-ephrinB2 signaling [164]. Recently, PS/γ-secretase was also shown to have an integral role in the formation of cell-matrix focal adhesions by transcriptional regulation of c-Src via ephrinB2 cleavage [165]. Furthermore, termination of the ephrinB2 receptor-EphB2 signaling has been proposed to convert the initial cell-cell adhesion to cell dissociation [166]. Finally, activation of EphA4 receptor by ephrin normally results in the retraction of dendritic spines. However, when PS/γ-secretase in hippocampal synapses released EphA4-ICD, this led to enhanced spine formation through activation of the Rac signaling pathway. Moreover, synaptic activity enhanced EphA4 cleavage by PS/γ-secretase, indicating that processing of EphA4 plays a role in the synaptic activity-dependent morphogenesis of dendritic spines [167]. These reports collectively imply that the bidirectional ephrin-Eph receptor signaling is regulated by PS/γ-secretase activity and as a result, the initial signaling of the full-length proteins is converted to opposite signaling or even terminated after receptor proteolysis to generate the cognate ICDs.

PS/γ-secretase-mediated proteolytic processing of several substrates also plays a role in the regulation of cell adhesion. PS/γ-secretase-mediated cleavage of E-cadherin has been shown to result in the disassembly of adherens junctions in epithelial cells by inducing disruption of the E-cadherin-β-catenin adhesion complex [130, 168]. On the contrary, our data suggested that in the case of nectin-1α, PS/γ-secretase inhibition results in the disassembly of adherens junctions and release of β-catenin to the cytoplasmic pool [169]. Whether PS/γ-secretase functions to stabilize or disassemble the junctions appears to depend on the substrate protein to be cleaved. We also found that whereas the full-length voltage-gated sodium channel β2 subunit (Navβ2) effectively induced cell adhesion and migration, γ-secretase inhibition completely blocked these events [170]. These results support the notion that PS/γ-secretase plays a role in the regulation of cell adhesion.

Regulation of angiogenesis and tumorigenesis

PS/γ-secretase activity is also implicated in the regulation of angiogenesis and tumorigenesis. Vascular endothelial growth factor receptor-1 (VEGFR-1) regulates angiogenesis stimulated by its ligand VEGF. Recently, VEGFR-1 was shown to undergo RIP by PS/γ-secretase, which resulted in the inhibition of angiogenesis [171]. Pigment epithelium-derived factor (PEDF), a potent inhibitor of formation of new vessels from endothelial cells, was found to enhance PS/γ-secretase-mediated cleavage of VEGFR-1. Furthermore, PEDF treatment of endothelial cells significantly increased PS/γ-secretase activity, PS1 endoproteolysis, and PS1 translocation to the cell membrane. Whereas full-length VEGFR-1 localized in the cytoskeletal and nuclear fractions, PEDF and VEGF induced the release of VEGFR-1-ICD to the cytoplasm [171]. Inhibition of PS/γ-secretase activity completely abolished the PEDF-mediated inhibitory effect on VEGF-induced angiogenesis and prevented VEGFR-1-ICD release to the cytosol [171]. These results suggest that PEDF functions as an inhibitor of VEGF-induced angiogenesis by inducing PS/γ-secretase-mediated proteolysis of VEGFR-1.

PS/γ-secretase has also been suggested to participate in tumorigenesis through the release of CD44-ICD, which may promote cell transformation [172]. As already discussed above, ErbB4-ICD (E4ICD) released by PS/γ-secretase activity may regulate gene transcription through several transcription factors, but also confer pro-apoptotic activity of ErbB4 [138142]. On the other hand, E4ICD (but not the membrane-associated forms of ErbB4) has been shown to increase the levels of the anti-apoptotic protein p53 and its transcriptional target p21 in response to neuregulin [173]. These events may be important in tumorigenesis, as nuclear ErbB4 has been reported in several human tumors and it predicts poor diagnosis in breast cancer [174].

Additional functions

Recent evidence indicates that PS/γ-secretase not only regulates Navβ2-mediated cell adhesion, but also voltage-gated sodium channel levels and activity [108]. We have found that BACE1 and PS/γ-secretase cleavages of Navβ2 release a Navβ2-ICD, which modulates the levels of the pore-forming Nav1.1 α-subunit intra-cellularly and on the cell surface [108]. Since four auxiliary subunits of the voltage-gated potassium channel are also cleaved by BACE1 and PS/γ-secretase [175], it appears that these two enzymes are involved in the regulation of ion channel function. Interestingly, voltage-gated sodium and potassium channels together are the key components of action potential generation and propagation.

Yet another cellular function involving PS/γ-secretase mediated cleavage is pigmentation. Tyrosinase (and its related proteins Tyrp1 and Tyrp2) produces eumelanin in melanosomes (endosomal/lysosomal-related organelles) of vertebrate pigment cells during melanin synthesis [176]. Tyrosinase is transported in vesicles from TGN to melanosomes and its correct processing and trafficking is essential for pigment synthesis [177]. However, impaired PS/γ-secretase function in mice resulted in defective pigmentation of the eyes and subcellular mislocalization of tyrosinase. Accordingly, treatment of mouse primary melanocytes with γ-secretase inhibitors blocked pigmentation [178]. Together, these data suggest that PS/γ-secretase is required for the correct targeting of tyrosinase to melanocytes to catalyze the production of melanin [178].

CONCLUDING REMARKS

To date, 91 PS/γ-secretase substrate proteins have been identified. Whether processing of any of these proteins results in the generation of toxic or pathogenic cleavage products similar to Aβ is thus far unknown. However, it is apparent that PS/γ-secretase-mediated processing affects the normal biological signaling of its substrate proteins. Therefore, an imbalance or potential impairment of PS/γ-secretase function might result in aberrant signaling and contribute to neurodegeneration during the course of AD pathogenesis. Presently, two different γ-secretase inhibitors are in phase I and two others in phase II and III clinical trials (http://www.alzforum.org; http://www.clinicaltrials.gov; [179]). However, the latest drawback for the potential γ-secretase inhibitor therapy in AD occurred recently when the Semagacestat (LY450139) γ-secretase inhibitor program by Eli Lilly and Company was halted in Phase III trials. The drug did not slow the cognitive decline in patients with mild to moderate AD, but in fact made them worse. Most of the commonly used experimental γ-secretase inhibitors block PS/γ-secretase-mediated processing of the substrate proteins in general (even though some substrate specificity or preference may take place) and augment substrate CTF levels while preventing ICD release [180]. Consequently, PS/γ-secretase inhibition may distort substrate signaling. For these reasons, wide-range γ-secretase inhibitors may be limited in their therapeutic potential. This has become evident in drug trials, as many of the γ-secretase inhibitors have caused side effects. Therefore, development of specific and safe γ-secretase inhibitors for clinical use faces great challenges. Encouraging evidence shows that substrate-specific γ-secretase inhibitors or modulators can inhibit Aβ production without affecting Notch cleavage [180182]. In addition, partial inhibition of PS/γ-secretase activity might provide sufficient therapeutic outcome and be considered as a form of therapy targeting PS/γ-secretase [183]. Nonetheless, even though PS/γ secretase is an appealing drug target, more preclinical and clinical studies are required before therapeutic γ-secretase inhibitors or modulators reach the market.

Acknowledgments

This work is supported by the Health Research Council of the Academy of Finland (AH) and two grants from the NIH/NIA (DMK).

Footnotes

Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=731).

References

  • 1.De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS, Ray WJ, Goate A, Kopan R. A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature. 1999;398:518–522. doi: 10.1038/19083. [DOI] [PubMed] [Google Scholar]
  • 2.Beel AJ, Sanders CR. Substrate specificity of gamma-secretase and other intramembrane proteases. Cell Mol Life Sci. 2008;65:1311–1334. doi: 10.1007/s00018-008-7462-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Spasic D, Annaert W. Building gamma-secretase: the bits and pieces. J Cell Sci. 2008;121:413–420. doi: 10.1242/jcs.015255. [DOI] [PubMed] [Google Scholar]
  • 4.Selkoe DJ, Wolfe MS. Presenilin: running with scissors in the membrane. Cell. 2007;131:215–221. doi: 10.1016/j.cell.2007.10.012. [DOI] [PubMed] [Google Scholar]
  • 5.Edbauer D, Winkler E, Regula JT, Pesold B, Steiner H, Haass C. Reconstitution of gamma-secretase activity. Nat Cell Biol. 2003;5:486–488. doi: 10.1038/ncb960. [DOI] [PubMed] [Google Scholar]
  • 6.Weihofen A, Martoglio B. Intramembrane-cleaving proteases: controlled liberation of proteins and bioactive peptides. Trends Cell Biol. 2003;13:71–78. doi: 10.1016/s0962-8924(02)00041-7. [DOI] [PubMed] [Google Scholar]
  • 7.Brown MS, Ye J, Rawson RB, Goldstein JL. Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell. 2000;100:391–398. doi: 10.1016/s0092-8674(00)80675-3. [DOI] [PubMed] [Google Scholar]
  • 8.Strisovsky K, Sharpe HJ, Freeman M. Sequence-specific intramembrane proteolysis: identification of a recognition motif in rhomboid substrates. Mol Cell. 2009;36:1048–1059. doi: 10.1016/j.molcel.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vetrivel KS, Thinakaran G. Amyloidogenic processing of beta-amyloid precursor protein in intracellular compartments. Neurology. 2006;66:S69–S73. doi: 10.1212/01.wnl.0000192107.17175.39. [DOI] [PubMed] [Google Scholar]
  • 10.Struhl G, Adachi A. Requirements for presenilin-dependent cleavage of notch and other transmembrane proteins. Mol Cell. 2000;6:625–636. doi: 10.1016/s1097-2765(00)00061-7. [DOI] [PubMed] [Google Scholar]
  • 11.Kopan R, Ilagan MX. Gamma-secretase: proteasome of the membrane? Nat Rev Mol Cell Biol. 2004;5:499–504. doi: 10.1038/nrm1406. [DOI] [PubMed] [Google Scholar]
  • 12.Masliah E, Westland CE, Rockenstein EM, Abraham CR, Mallory M, Veinberg I, Sheldon E, Mucke L. Amyloid precursor proteins protect neurons of transgenic mice against acute and chronic excitotoxic injuries in vivo. Neuroscience. 1997;78:135–146. doi: 10.1016/s0306-4522(96)00553-2. [DOI] [PubMed] [Google Scholar]
  • 13.Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci. 2006;26:7212–7221. doi: 10.1523/JNEUROSCI.1450-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lazarov O, Morfini GA, Lee EB, Farah MH, Szodorai A, DeBoer SR, Koliatsos VE, Kins S, Lee VM, Wong PC, Price DL, Brady ST, Sisodia SS. Axonal transport, amyloid precursor protein, kinesin-1, and the processing apparatus: revisited. J Neurosci. 2005;25:2386–2395. doi: 10.1523/JNEUROSCI.3089-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.LeBlanc AC, Kovacs DM, Chen HY, Villare F, Tykocinski M, Autilio-Gambetti L, Gambetti P. Role of amyloid precursor protein (APP): study with antisense transfection of human neuroblastoma cells. J Neurosci Res. 1992;31:635–645. doi: 10.1002/jnr.490310407. [DOI] [PubMed] [Google Scholar]
  • 16.Coulson EJ, Barrett GL, Storey E, Bartlett PF, Beyreuther K, Masters CL. Down-regulation of the amyloid protein precursor of Alzheimer’s disease by antisense oligonucleotides reduces neuronal adhesion to specific substrata. Brain Res. 1997;770:72–80. doi: 10.1016/s0006-8993(97)00757-9. [DOI] [PubMed] [Google Scholar]
  • 17.Soba P, Eggert S, Wagner K, Zentgraf H, Siehl K, Kreger S, Lower A, Langer A, Merdes G, Paro R, Masters CL, Muller U, Kins S, Beyreuther K. Homo- and heterodimerization of APP family members promotes intercellular adhesion. EMBO J. 2005;24:3624–3634. doi: 10.1038/sj.emboj.7600824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hoe HS, Lee KJ, Carney RS, Lee J, Markova A, Lee JY, Howell BW, Hyman BT, Pak DT, Bu G, Rebeck GW. Interaction of reelin with amyloid precursor protein promotes neurite outgrowth. J Neurosci. 2009;29:7459–7473. doi: 10.1523/JNEUROSCI.4872-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Buxbaum JD, Liu KN, Luo Y, Slack JL, Stocking KL, Peschon JJ, Johnson RS, Castner BJ, Cerretti DP, Black RA. Evidence that tumor necrosis factor alpha converting enzyme is involved in regulated alpha-secretase cleavage of the Alzheimer amyloid protein precursor. J Biol Chem. 1998;273:27765–27767. doi: 10.1074/jbc.273.43.27765. [DOI] [PubMed] [Google Scholar]
  • 20.Lopez-Perez E, Zhang Y, Frank SJ, Creemers J, Seidah N, Checler F. Constitutive alpha-secretase cleavage of the beta-amyloid precursor protein in the furin-deficient LoVo cell line: involvement of the pro-hormone convertase 7 and the disintegrin metalloprotease ADAM10. J Neurochem. 2001;76:1532–1539. doi: 10.1046/j.1471-4159.2001.00180.x. [DOI] [PubMed] [Google Scholar]
  • 21.Hiltunen M, van Groen T, Jolkkonen J. Functional roles of amyloid-beta protein precursor and amyloid-beta peptides: evidence from experimental studies. J Alzheimers Dis. 2009;18:401–412. doi: 10.3233/JAD-2009-1154. [DOI] [PubMed] [Google Scholar]
  • 22.Selkoe DJ. Cell biology of protein misfolding: the examples of Alzheimer’s and Parkinson’s diseases. Nat Cell Biol. 2004;6:1054–1061. doi: 10.1038/ncb1104-1054. [DOI] [PubMed] [Google Scholar]
  • 23.Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430:631–639. doi: 10.1038/nature02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kimberly WT, Zheng JB, Guenette SY, Selkoe DJ. The intracellular domain of the beta-amyloid precursor protein is stabilized by Fe65 and translocates to the nucleus in a notch-like manner. J Biol Chem. 2001;276:40288–40292. doi: 10.1074/jbc.C100447200. [DOI] [PubMed] [Google Scholar]
  • 25.Cao X, Sudhof TC. A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60. Science. 2001;293:115–120. doi: 10.1126/science.1058783. [DOI] [PubMed] [Google Scholar]
  • 26.von Rotz RC, Kohli BM, Bosset J, Meier M, Suzuki T, Nitsch RM, Konietzko U. The APP intracellular domain forms nuclear multiprotein complexes and regulates the transcription of its own precursor. J Cell Sci. 2004;117:4435–4448. doi: 10.1242/jcs.01323. [DOI] [PubMed] [Google Scholar]
  • 27.Baek SH, Ohgi KA, Rose DW, Koo EH, Glass CK, Rosenfeld MG. Exchange of N-CoR corepressor and Tip60 coactivator complexes links gene expression by NF-kappaB and beta-amyloid precursor protein. Cell. 2002;110:55–67. doi: 10.1016/s0092-8674(02)00809-7. [DOI] [PubMed] [Google Scholar]
  • 28.Cao X, Sudhof TC. Dissection of amyloid-beta precursor protein-dependent transcriptional transactivation. J Biol Chem. 2004;279:24601–24611. doi: 10.1074/jbc.M402248200. [DOI] [PubMed] [Google Scholar]
  • 29.Pardossi-Piquard R, Petit A, Kawarai T, Sunyach C, Alves da Costa C, Vincent B, Ring S, D’Adamio L, Shen J, Muller U, St George Hyslop P, Checler F. Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betaAPP and APLP. Neuron. 2005;46:541–554. doi: 10.1016/j.neuron.2005.04.008. [DOI] [PubMed] [Google Scholar]
  • 30.Ozaki T, Li Y, Kikuchi H, Tomita T, Iwatsubo T, Nakagawara A. The intracellular domain of the amyloid precursor protein (AICD) enhances the p53-mediated apoptosis. Biochem Biophys Res Commun. 2006;351:57–63. doi: 10.1016/j.bbrc.2006.09.162. [DOI] [PubMed] [Google Scholar]
  • 31.Alves da Costa C, Sunyach C, Pardossi-Piquard R, Sevalle J, Vincent B, Boyer N, Kawarai T, Girardot N, St George-Hyslop P, Checler F. Presenilin-dependent gamma-secretase-mediated control of p53-associated cell death in Alzheimer’s disease. J Neurosci. 2006;26:6377–6385. doi: 10.1523/JNEUROSCI.0651-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang YW, Wang R, Liu Q, Zhang H, Liao FF, Xu H. Presenilin/gamma-secretase-dependent processing of beta-amyloid precursor protein regulates EGF receptor expression. Proc Natl Acad Sci U S A. 2007;104:10613–10618. doi: 10.1073/pnas.0703903104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hebert SS, Serneels L, Tolia A, Craessaerts K, Derks C, Filippov MA, Muller U, De Strooper B. Regulated intramembrane proteolysis of amyloid precursor protein and regulation of expression of putative target genes. EMBO Rep. 2006;7:739–745. doi: 10.1038/sj.embor.7400704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Waldron E, Isbert S, Kern A, Jaeger S, Martin AM, Hebert SS, Behl C, Weggen S, De Strooper B, Pietrzik CU. Increased AICD generation does not result in increased nuclear translocation or activation of target gene transcription. Exp Cell Res. 2008;314:2419–2433. doi: 10.1016/j.yexcr.2008.05.003. [DOI] [PubMed] [Google Scholar]
  • 35.Ring S, Weyer SW, Kilian SB, Waldron E, Pietrzik CU, Filippov MA, Herms J, Buchholz C, Eckman CB, Korte M, Wolfer DP, Muller UC. The secreted beta-amyloid precursor protein ectodomain APPs alpha is sufficient to rescue the anatomical, behavioral, and electrophysiological abnormalities of APP-deficient mice. J Neurosci. 2007;27:7817–7826. doi: 10.1523/JNEUROSCI.1026-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Goodger ZV, Rajendran L, Trutzel A, Kohli BM, Nitsch RM, Konietzko U. Nuclear signaling by the APP intracellular domain occurs predominantly through the amyloidogenic processing pathway. J Cell Sci. 2009;122:3703–3714. doi: 10.1242/jcs.048090. [DOI] [PubMed] [Google Scholar]
  • 37.Ghosal K, Vogt DL, Liang M, Shen Y, Lamb BT, Pimplikar SW. Alzheimer’s disease-like pathological features in transgenic mice expressing the APP intracellular domain. Proc Natl Acad Sci U S A. 2009;106:18367–18372. doi: 10.1073/pnas.0907652106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hussain I, Powell D, Howlett DR, Tew DG, Meek TD, Chapman C, Gloger IS, Murphy KE, Southan CD, Ryan DM, Smith TS, Simmons DL, Walsh FS, Dingwall C, Christie G. Identification of a novel aspartic protease (Asp 2) as beta-secretase. Mol Cell Neurosci. 1999;14:419–427. doi: 10.1006/mcne.1999.0811. [DOI] [PubMed] [Google Scholar]
  • 39.Sinha S, Anderson JP, Barbour R, Basi GS, Caccavello R, Davis D, Doan M, Dovey HF, Frigon N, Hong J, Jacobson-Croak K, Jewett N, Keim P, Knops J, Lieberburg I, Power M, Tan H, Tatsuno G, Tung J, Schenk D, Seubert P, Suomensaari SM, Wang S, Walker D, Zhao J, McConlogue L, John V. Purification and cloning of amyloid precursor protein beta-secretase from human brain. Nature. 1999;402:537–540. doi: 10.1038/990114. [DOI] [PubMed] [Google Scholar]
  • 40.Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, Luo Y, Fisher S, Fuller J, Edenson S, Lile J, Jarosinski MA, Biere AL, Curran E, Burgess T, Louis JC, Collins F, Treanor J, Rogers G, Citron M. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286:735–741. doi: 10.1126/science.286.5440.735. [DOI] [PubMed] [Google Scholar]
  • 41.Yan R, Bienkowski MJ, Shuck ME, Miao H, Tory MC, Pauley AM, Brashier JR, Stratman NC, Mathews WR, Buhl AE, Carter DB, Tomasselli AG, Parodi LA, Heinrikson RL, Gurney ME. Membrane-anchored aspartyl protease with Alzheimer’s disease beta-secretase activity. Nature. 1999;402:533–537. doi: 10.1038/990107. [DOI] [PubMed] [Google Scholar]
  • 42.Nalivaeva NN, Fisk LR, Belyaev ND, Turner AJ. Amyloid-degrading enzymes as therapeutic targets in Alzheimer’s disease. Curr Alzheimer Res. 2008;5:212–224. doi: 10.2174/156720508783954785. [DOI] [PubMed] [Google Scholar]
  • 43.Miners JS, Baig S, Palmer J, Palmer LE, Kehoe PG, Love S. Abeta-degrading enzymes in Alzheimer’s disease. Brain Pathol. 2008;18:240–252. doi: 10.1111/j.1750-3639.2008.00132.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zhao J, Fu Y, Yasvoina M, Shao P, Hitt B, O’Connor T, Logan S, Maus E, Citron M, Berry R, Binder L, Vassar R. Beta-site amyloid precursor protein cleaving enzyme 1 levels become elevated in neurons around amyloid plaques: implications for Alzheimer’s disease pathogenesis. J Neurosci. 2007;27:3639–3649. doi: 10.1523/JNEUROSCI.4396-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tanzi RE, Bertram L. Twenty years of the Alzheimer’s disease amyloid hypothesis: a genetic perspective. Cell. 2005;120:545–555. doi: 10.1016/j.cell.2005.02.008. [DOI] [PubMed] [Google Scholar]
  • 46.De Strooper B, Saftig P, Craessaerts K, Vanderstichele H, Guhde G, Annaert W, Von Figura K, Van Leuven F. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature. 1998;391:387–390. doi: 10.1038/34910. [DOI] [PubMed] [Google Scholar]
  • 47.Xia W, Zhang J, Ostaszewski BL, Kimberly WT, Seubert P, Koo EH, Shen J, Selkoe DJ. Presenilin 1 regulates the processing of beta-amyloid precursor protein C-terminal fragments and the generation of amyloid beta-protein in endoplasmic reticulum and golgi. Biochemistry. 1998;37:16465–16471. doi: 10.1021/bi9816195. [DOI] [PubMed] [Google Scholar]
  • 48.Chen F, Yang DS, Petanceska S, Yang A, Tandon A, Yu G, Rozmahel R, Ghiso J, Nishimura M, Zhang DM, Kawarai T, Levesque G, Mills J, Levesque L, Song YQ, Rogaeva E, Westaway D, Mount H, Gandy S, St George-Hyslop P, Fraser PE. Carboxyl-terminal fragments of Alzheimer beta-amyloid precursor protein accumulate in restricted and unpredicted intracellular compartments in presenilin 1-deficient cells. J Biol Chem. 2000;275:36794–36802. doi: 10.1074/jbc.M006986200. [DOI] [PubMed] [Google Scholar]
  • 49.Wolfe MS, De Los Angeles J, Miller DD, Xia W, Selkoe DJ. Are presenilins intramembrane-cleaving proteases? Implications for the molecular mechanism of Alzheimer’s disease. Biochemistry. 1999;38:11223–11230. doi: 10.1021/bi991080q. [DOI] [PubMed] [Google Scholar]
  • 50.Yu G, Nishimura M, Arawaka S, Levitan D, Zhang L, Tandon A, Song YQ, Rogaeva E, Chen F, Kawarai T, Supala A, Levesque L, Yu H, Yang DS, Holmes E, Milman P, Liang Y, Zhang DM, Xu DH, Sato C, Rogaev E, Smith M, Janus C, Zhang Y, Aebersold R, Farrer LS, Sorbi S, Bruni A, Fraser P, St George-Hyslop P. Nicastrin modulates presenilin-mediated notch/glp-1 signal transduction and betaAPP processing. Nature. 2000;407:48–54. doi: 10.1038/35024009. [DOI] [PubMed] [Google Scholar]
  • 51.Goutte C, Tsunozaki M, Hale VA, Priess JR. APH-1 is a multipass membrane protein essential for the Notch signaling pathway in Caenorhabditis elegans embryos. Proc Natl Acad Sci U S A. 2002;99:775–779. doi: 10.1073/pnas.022523499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lee SF, Shah S, Li H, Yu C, Han W, Yu G. Mammalian APH-1 interacts with presenilin and nicastrin and is required for intramembrane proteolysis of amyloid-beta precursor protein and Notch. J Biol Chem. 2002;277:45013–45019. doi: 10.1074/jbc.M208164200. [DOI] [PubMed] [Google Scholar]
  • 53.Francis R, McGrath G, Zhang J, Ruddy DA, Sym M, Apfeld J, Nicoll M, Maxwell M, Hai B, Ellis MC, Parks AL, Xu W, Li J, Gurney M, Myers RL, Himes CS, Hiebsch R, Ruble C, Nye JS, Curtis D. aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell. 2002;3:85–97. doi: 10.1016/s1534-5807(02)00189-2. [DOI] [PubMed] [Google Scholar]
  • 54.Steiner H, Winkler E, Edbauer D, Prokop S, Basset G, Yamasaki A, Kostka M, Haass C. PEN-2 is an integral component of the gamma-secretase complex required for coordinated expression of presenilin and nicastrin. J Biol Chem. 2002;277:39062–39065. doi: 10.1074/jbc.C200469200. [DOI] [PubMed] [Google Scholar]
  • 55.Haass C, Steiner H. Alzheimer disease gamma-secretase: a complex story of GxGD-type presenilin pro teases. Trends Cell Biol. 2002;12:556–562. doi: 10.1016/s0962-8924(02)02394-2. [DOI] [PubMed] [Google Scholar]
  • 56.De Strooper B. Aph-1, Pen-2, and Nicastrin with presenilin generate an active gamma-secretase complex. Neuron. 2003;38:9–12. doi: 10.1016/s0896-6273(03)00205-8. [DOI] [PubMed] [Google Scholar]
  • 57.Wolfe MS, Xia W, Ostaszewski BL, Diehl TS, Kimberly WT, Selkoe DJ. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature. 1999;398:513–517. doi: 10.1038/19077. [DOI] [PubMed] [Google Scholar]
  • 58.Kimberly WT, Xia W, Rahmati T, Wolfe MS, Selkoe DJ. The transmembrane aspartates in presenilin 1 and 2 are obligatory for gamma-secretase activity and amyloid beta-protein generation. J Biol Chem. 2000;275:3173–3178. doi: 10.1074/jbc.275.5.3173. [DOI] [PubMed] [Google Scholar]
  • 59.Shah S, Lee SF, Tabuchi K, Hao YH, Yu C, LaPlant Q, Ball H, Dann CE, 3rd, Sudhof T, Yu G. Nicastrin functions as a gamma-secretase-substrate receptor. Cell. 2005;122:435–447. doi: 10.1016/j.cell.2005.05.022. [DOI] [PubMed] [Google Scholar]
  • 60.Chavez-Gutierrez L, Tolia A, Maes E, Li T, Wong PC, de Strooper B. Glu(332) in the Nicastrin ectodomain is essential for gamma-secretase complex maturation but not for its activity. J Biol Chem. 2008;283:20096–20105. doi: 10.1074/jbc.M803040200. [DOI] [PubMed] [Google Scholar]
  • 61.Zhao G, Liu Z, Ilagan MX, Kopan R. Gamma-secretase composed of PS1/Pen2/Aph1 a can cleave notch and amyloid precursor protein in the absence of nicastrin. J Neurosci. 2010;30:1648–1656. doi: 10.1523/JNEUROSCI.3826-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dries DR, Shah S, Han YH, Yu C, Yu S, Shearman MS, Yu G. Glu-333 of nicastrin directly participates in gamma-secretase activity. J Biol Chem. 2009;284:29714–29724. doi: 10.1074/jbc.M109.038737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Serneels L, Van Biervliet J, Craessaerts K, Dejaegere T, Horre K, Van Houtvin T, Esselmann H, Paul S, Schafer MK, Berezovska O, Hyman BT, Sprangers B, Sciot R, Moons L, Jucker M, Yang Z, May PC, Karran E, Wiltfang J, D’Hooge R, De Strooper B. Gamma-secretase heterogeneity in the Aph1 subunit: relevance for Alzheimer’s disease. Science. 2009;324:639–642. doi: 10.1126/science.1171176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen AC, Guo LY, Ostaszewski BL, Selkoe DJ, LaVoie MJ. Aph-1 associates directly with full-length and C-terminal fragments of gamma-secretase substrates. J Biol Chem. 2010;285:11378–11391. doi: 10.1074/jbc.M109.088815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Thinakaran G, Borchelt DR, Lee MK, Slunt HH, Spitzer L, Kim G, Ratovitsky T, Davenport F, Nordstedt C, Seeger M, Hardy J, Levey AI, Gandy SE, Jenkins NA, Copeland NG, Price DL, Sisodia SS. Endoproteolysis of presenilin 1 and accumulation of processed derivatives in vivo. Neuron. 1996;17:181–190. doi: 10.1016/s0896-6273(00)80291-3. [DOI] [PubMed] [Google Scholar]
  • 66.Sato T, Diehl TS, Narayanan S, Funamoto S, Ihara Y, De Strooper B, Steiner H, Haass C, Wolfe MS. Active gamma-secretase complexes contain only one of each component. J Biol Chem. 2007;282:33985–33993. doi: 10.1074/jbc.M705248200. [DOI] [PubMed] [Google Scholar]
  • 67.Vetrivel KS, Zhang YW, Xu H, Thinakaran G. Pathological and physiological functions of presenilins. Mol Neurodegener. 2006;1:4. doi: 10.1186/1750-1326-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Serneels L, Dejaegere T, Craessaerts K, Horre K, Jorissen E, Tousseyn T, Hebert S, Coolen M, Martens G, Zwijsen A, Annaert W, Hartmann D, De Strooper B. Differential contribution of the three Aph1 genes to gamma-secretase activity in vivo. Proc Natl Acad Sci U S A. 2005;102:1719–1724. doi: 10.1073/pnas.0408901102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754–760. doi: 10.1038/375754a0. [DOI] [PubMed] [Google Scholar]
  • 70.Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K. Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science. 1995;269:973–977. doi: 10.1126/science.7638622. [DOI] [PubMed] [Google Scholar]
  • 71.Lai MT, Chen E, Crouthamel MC, DiMuzio-Mower J, Xu M, Huang Q, Price E, Register RB, Shi XP, Donoviel DB, Bernstein A, Hazuda D, Gardell SJ, Li YM. Presenilin-1 and presenilin-2 exhibit distinct yet overlapping gamma-secretase activities. J Biol Chem. 2003;278:22475–22481. doi: 10.1074/jbc.M300974200. [DOI] [PubMed] [Google Scholar]
  • 72.Mastrangelo P, Mathews PM, Chishti MA, Schmidt SD, Gu Y, Yang J, Mazzella MJ, Coomaraswamy J, Horne P, Strome B, Pelly H, Levesque G, Ebeling C, Jiang Y, Nixon RA, Rozmahel R, Fraser PE, St George-Hyslop P, Carlson GA, Westaway D. Dissociated phenotypes in presenilin transgenic mice define functionally distinct gamma-secretases. Proc Natl Acad Sci U S A. 2005;102:8972–8977. doi: 10.1073/pnas.0500940102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ, Tonegawa S. Skeletal and CNS defects in presenilin-1-deficient mice. Cell. 1997;89:629–639. doi: 10.1016/s0092-8674(00)80244-5. [DOI] [PubMed] [Google Scholar]
  • 74.Herreman A, Hartmann D, Annaert W, Saftig P, Craessaerts K, Serneels L, Umans L, Schrijvers V, Checler F, Vanderstichele H, Baekelandt V, Dressel R, Cupers P, Huylebroeck D, Zwijsen A, Van Leuven F, De Strooper B. 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 U S A. 1999;96:11872–11877. doi: 10.1073/pnas.96.21.11872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, Kieu C, Papior P, Baeuerle PA, Munz M, Gires O. Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol. 2009;11:162–171. doi: 10.1038/ncb1824. [DOI] [PubMed] [Google Scholar]
  • 76.Huysseune S, Kienlen-Campard P, Hebert S, Tasiaux B, Leroy K, Devuyst O, Brion JP, De Strooper B, Octave JN. Epigenetic control of aquaporin 1 expression by the amyloid precursor protein. FASEB J. 2009;23:4158–4167. doi: 10.1096/fj.09-140012. [DOI] [PubMed] [Google Scholar]
  • 77.Zhou S, Zhou H, Walian PJ, Jap BK. CD147 is a regulatory subunit of the gamma-secretase complex in Alzheimer’s disease amyloid beta-peptide production. Proc Natl Acad Sci U S A. 2005;102:7499–7504. doi: 10.1073/pnas.0502768102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chen F, Hasegawa H, Schmitt-Ulms G, Kawarai T, Bohm C, Katayama T, Gu Y, Sanjo N, Glista M, Rogaeva E, Wakutani Y, Pardossi-Piquard R, Ruan X, Tandon A, Checler F, Marambaud P, Hansen K, Westaway D, St George-Hyslop P, Fraser P. TMP21 is a presenilin complex component that modulates gamma-secretase but not epsilon-secretase activity. Nature. 2006;440:1208–1212. doi: 10.1038/nature04667. [DOI] [PubMed] [Google Scholar]
  • 79.Verdile G, Gandy SE, Martins RN. The role of pre-senilin and its interacting proteins in the biogenesis of Alzheimer’s beta amyloid. Neurochem Res. 2007;32:609–623. doi: 10.1007/s11064-006-9131-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Huovila AP, Turner AJ, Pelto-Huikko M, Karkkainen I, Ortiz RM. Shedding light on ADAM metalloproteinases. Trends Biochem Sci. 2005;30:413–422. doi: 10.1016/j.tibs.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 81.Schlondorff J, Becherer JD, Blobel CP. Intracellular maturation and localization of the tumour necrosis factor alpha convertase (TACE) Biochem J. 2000;347(Pt 1):131–138. [PMC free article] [PubMed] [Google Scholar]
  • 82.Jolly-Tornetta C, Wolf BA. Protein kinase C regulation of intracellular and cell surface amyloid precursor protein (APP) cleavage in CHO695 cells. Biochemistry. 2000;39:15282–15290. doi: 10.1021/bi001723y. [DOI] [PubMed] [Google Scholar]
  • 83.Skovronsky DM, Moore DB, Milla ME, Doms RW, Lee VM. Protein kinase C-dependent alpha-secretase competes with beta-secretase for cleavage of amyloid-beta precursor protein in the trans-golgi network. J Biol Chem. 2000;275:2568–2575. doi: 10.1074/jbc.275.4.2568. [DOI] [PubMed] [Google Scholar]
  • 84.Stephens DJ, Austen BM. Metabolites of the beta-amyloid precursor protein generated by beta-secretase localise to the trans-golgi network and late endosome in 293 cells. J Neurosci Res. 1996;46:211–225. doi: 10.1002/(SICI)1097-4547(19961015)46:2<211::AID-JNR9>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 85.Daugherty BL, Green SA. Endosomal sorting of amyloid precursor protein-P-selectin chimeras influences secretase processing. Traffic. 2001;2:908–916. doi: 10.1034/j.1600-0854.2001.21206.x. [DOI] [PubMed] [Google Scholar]
  • 86.Walter J, Fluhrer R, Hartung B, Willem M, Kaether C, Capell A, Lammich S, Multhaup G, Haass C. Phosphorylation regulates intracellular trafficking of beta-secretase. J Biol Chem. 2001;276:14634–14641. doi: 10.1074/jbc.M011116200. [DOI] [PubMed] [Google Scholar]
  • 87.Chyung JH, Selkoe DJ. Inhibition of receptor-mediated endocytosis demonstrates generation of amyloid beta-protein at the cell surface. J Biol Chem. 2003;278:51035–51043. doi: 10.1074/jbc.M304989200. [DOI] [PubMed] [Google Scholar]
  • 88.Kinoshita A, Fukumoto H, Shah T, Whelan CM, Irizarry MC, Hyman BT. Demonstration by FRET of BACE interaction with the amyloid precursor protein at the cell surface and in early endosomes. J Cell Sci. 2003;116:3339–3346. doi: 10.1242/jcs.00643. [DOI] [PubMed] [Google Scholar]
  • 89.Kim SH, Yin YI, Li YM, Sisodia SS. Evidence that assembly of an active gamma-secretase complex occurs in the early compartments of the secretory pathway. J Biol Chem. 2004;279:48615–48619. doi: 10.1074/jbc.C400396200. [DOI] [PubMed] [Google Scholar]
  • 90.Vetrivel KS, Cheng H, Lin W, Sakurai T, Li T, Nukina N, Wong PC, Xu H, Thinakaran G. Association of gamma-secretase with lipid rafts in post-golgi and endosome membranes. J Biol Chem. 2004;279:44945–44954. doi: 10.1074/jbc.M407986200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Pasternak SH, Bagshaw RD, Guiral M, Zhang S, Ackerley CA, Pak BJ, Callahan JW, Mahuran DJ. Presenilin-1, nicastrin, amyloid precursor protein, and gamma-secretase activity are co-localized in the lysosomal membrane. J Biol Chem. 2003;278:26687–26694. doi: 10.1074/jbc.m304009200. [DOI] [PubMed] [Google Scholar]
  • 92.Jutras I, Laplante A, Boulais J, Brunet S, Thinakaran G, Desjardins M. Gamma-secretase is a functional component of phagosomes. J Biol Chem. 2005;280:36310–36317. doi: 10.1074/jbc.M504069200. [DOI] [PubMed] [Google Scholar]
  • 93.Fukumori A, Okochi M, Tagami S, Jiang J, Itoh N, Nakayama T, Yanagida K, Ishizuka-Katsura Y, Morihara T, Kamino K, Tanaka T, Kudo T, Tanii H, Ikuta A, Haass C, Takeda M. Presenilin-dependent gamma-secretase on plasma membrane and endosomes is functionally distinct. Biochemistry. 2006;45:4907–4914. doi: 10.1021/bi052412w. [DOI] [PubMed] [Google Scholar]
  • 94.Chyung JH, Raper DM, Selkoe DJ. Gamma-secretase exists on the plasma membrane as an intact complex that accepts substrates and effects intramembrane cleavage. J Biol Chem. 2005;280:4383–4392. doi: 10.1074/jbc.M409272200. [DOI] [PubMed] [Google Scholar]
  • 95.Hansson CA, Frykman S, Farmery MR, Tjernberg LO, Nilsberth C, Pursglove SE, Ito A, Winblad B, Cowburn RF, Thyberg J, Ankarcrona M. Nicastrin, presenilin, APH-1, and PEN-2 form active gamma-secretase complexes in mitochondria. J Biol Chem. 2004;279:51654–51660. doi: 10.1074/jbc.M404500200. [DOI] [PubMed] [Google Scholar]
  • 96.Carey RM, Balcz BA, Lopez-Coviella I, Slack BE. Inhibition of dynamin-dependent endocytosis increases shedding of the amyloid precursor protein ectodomain and reduces generation of amyloid beta protein. BMC Cell Biol. 2005;6:30. doi: 10.1186/1471-2121-6-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Cataldo AM, Barnett JL, Pieroni C, Nixon RA. Increased neuronal endocytosis and protease delivery to early endosomes in sporadic Alzheimer’s disease: neuropathologic evidence for a mechanism of increased beta-amyloidogenesis. J Neurosci. 1997;17:6142–6151. doi: 10.1523/JNEUROSCI.17-16-06142.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman BT, Nixon RA. Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer’s disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. Am J Pathol. 2000;157:277–286. doi: 10.1016/s0002-9440(10)64538-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Huse JT, Pijak DS, Leslie GJ, Lee VM, Doms RW. Maturation and endosomal targeting of beta-site amyloid precursor protein-cleaving enzyme. The Alzheimer’s disease beta-secretase. J Biol Chem. 2000;275:33729–33737. doi: 10.1074/jbc.M004175200. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang M, Haapasalo A, Kim DY, Ingano LA, Pettingell WH, Kovacs DM. Presenilin/gamma-secretase activity regulates protein clearance from the endocytic recycling compartment. FASEB J. 2006;20:1176–1178. doi: 10.1096/fj.05-5531fje. [DOI] [PubMed] [Google Scholar]
  • 101.Kaether C, Schmitt S, Willem M, Haass C. Amyloid precursor protein and Notch intracellular domains are generated after transport of their precursors to the cell surface. Traffic. 2006;7:408–415. doi: 10.1111/j.1600-0854.2006.00396.x. [DOI] [PubMed] [Google Scholar]
  • 102.Urra S, Escudero CA, Ramos P, Lisbona F, Allende E, Covarrubias P, Parraguez JI, Zampieri N, Chao MV, Annaert W, Bronfman FC. 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. 2007;282:7606–7615. doi: 10.1074/jbc.M610458200. [DOI] [PubMed] [Google Scholar]
  • 103.Vaccari T, Lu H, Kanwar R, Fortini ME, Bilder D. Endosomal entry regulates Notch receptor activation in Drosophila melanogaster. J Cell Biol. 2008;180:755–762. doi: 10.1083/jcb.200708127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Gupta-Rossi N, Six E, LeBail O, Logeat F, Chastagner P, Olry A, Israel A, Brou C. Monoubiquitination and endocytosis direct gamma-secretase cleavage of activated Notch receptor. J Cell Biol. 2004;166:73–83. doi: 10.1083/jcb.200310098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Bossy-Wetzel E, Schwarzenbacher R, Lipton SA. Molecular pathways to neurodegeneration. Nat Med. 2004;10 (Suppl):S2–S9. doi: 10.1038/nm1067. [DOI] [PubMed] [Google Scholar]
  • 106.Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362:329–344. doi: 10.1056/NEJMra0909142. [DOI] [PubMed] [Google Scholar]
  • 107.von Arnim CA, Kinoshita A, Peltan ID, Tangredi MM, Herl L, Lee BM, Spoelgen R, Hshieh TT, Ranganathan S, Battey FD, Liu CX, Bacskai BJ, Sever S, Irizarry MC, Strickland DK, Hyman BT. The low density lipoprotein receptor-related protein (LRP) is a novel beta-secretase (BACE1) substrate. J Biol Chem. 2005;280:17777–17785. doi: 10.1074/jbc.M414248200. [DOI] [PubMed] [Google Scholar]
  • 108.Kim DY, Carey BW, Wang H, Ingano LA, Binshtok AM, Wertz MH, Pettingell WH, He P, Lee VM, Woolf CJ, Kovacs DM. BACE1 regulates voltage-gated sodium channels and neuronal activity. Nat Cell Biol. 2007;9:755–764. doi: 10.1038/ncb1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Araki Y, Miyagi N, Kato N, Yoshida T, Wada S, Nishimura M, Komano H, Yamamoto T, De Strooper B, Yamamoto K, Suzuki T. Coordinated metabolism of Alcadein and amyloid beta-protein precursor regulates FE65-dependent gene transactivation. J Biol Chem. 2004;279:24343–24354. doi: 10.1074/jbc.M401925200. [DOI] [PubMed] [Google Scholar]
  • 110.Okochi M, Steiner H, Fukumori A, Tanii H, Tomita T, Tanaka T, Iwatsubo T, Kudo T, Takeda M, Haass C. Presenilins mediate a dual intramembranous gamma-secretase cleavage of Notch-1. EMBO J. 2002;21:5408–5416. doi: 10.1093/emboj/cdf541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Walsh DM, Fadeeva JV, LaVoie MJ, Paliga K, Eggert S, Kimberly WT, Wasco W, Selkoe DJ. gamma-Secretase cleavage and binding to FE65 regulate the nuclear translocation of the intracellular C-terminal domain (ICD) of the APP family of proteins. Biochemistry. 2003;42:6664–6673. doi: 10.1021/bi027375c. [DOI] [PubMed] [Google Scholar]
  • 112.Hemming ML, Elias JE, Gygi SP, Selkoe DJ. Proteomic profiling of gamma-secretase substrates and mapping of substrate requirements. PLoS Biol. 2008;6:e257. doi: 10.1371/journal.pbio.0060257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.McCarthy JV, Twomey C, Wujek P. Presenilin-dependent regulated intramembrane proteolysis and gamma-secretase activity. Cell Mol Life Sci. 2009;66:1534–1555. doi: 10.1007/s00018-009-8435-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mumm JS, Schroeter EH, Saxena MT, Griesemer A, Tian X, Pan DJ, Ray WJ, Kopan R. A ligand-induced extracellular cleavage regulates gamma-secretase-like proteolytic activation of Notch1. Mol Cell. 2000;5:197–206. doi: 10.1016/s1097-2765(00)80416-5. [DOI] [PubMed] [Google Scholar]
  • 115.Nakahara S, Saito T, Kondo N, Moriwaki K, Noda K, Ihara S, Takahashi M, Ide Y, Gu J, Inohara H, Katayama T, Tohyama M, Kubo T, Taniguchi N, Miyoshi E. A secreted type of beta1,6 N-acetylglucosaminyltransferase V (GnT-V), a novel angiogenesis inducer, is regulated by gamma-secretase. FASEB J. 2006;20:2451–2459. doi: 10.1096/fj.05-5066com. [DOI] [PubMed] [Google Scholar]
  • 116.Meyer EL, Strutz N, Gahring LC, Rogers SW. Glutamate receptor subunit 3 is modified by site-specific limited proteolysis including cleavage by gamma-secretase. J Biol Chem. 2003;278:23786–23796. doi: 10.1074/jbc.M301360200. [DOI] [PubMed] [Google Scholar]
  • 117.Scheinfeld MH, Ghersi E, Laky K, Fowlkes BJ, D’Adamio L. Processing of beta-amyloid precursor-like protein-1 and -2 by gamma-secretase regulates transcription. J Biol Chem. 2002;277:44195–44201. doi: 10.1074/jbc.M208110200. [DOI] [PubMed] [Google Scholar]
  • 118.Minopoli G, de Candia P, Bonetti A, Faraonio R, Zambrano N, Russo T. The beta-amyloid precursor protein functions as a cytosolic anchoring site that prevents Fe65 nuclear translocation. J Biol Chem. 2001;276:6545–6550. doi: 10.1074/jbc.M007340200. [DOI] [PubMed] [Google Scholar]
  • 119.Okamoto I, Kawano Y, Murakami D, Sasayama T, Araki N, Miki T, Wong AJ, Saya H. Proteolytic release of CD44 intracellular domain and its role in the CD44 signaling pathway. J Cell Biol. 2001;155:755–762. doi: 10.1083/jcb.200108159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Taniguchi Y, Kim SH, Sisodia SS. Presenilin-dependent “gamma-secretase” processing of deleted in colorectal cancer (DCC) J Biol Chem. 2003;278:30425–30428. doi: 10.1074/jbc.C300239200. [DOI] [PubMed] [Google Scholar]
  • 121.Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature. 1998;393:382–386. doi: 10.1038/30756. [DOI] [PubMed] [Google Scholar]
  • 122.Saxena MT, Schroeter EH, Mumm JS, Kopan R. Murine notch homologs (N1–4) undergo presenilin-dependent proteolysis. J Biol Chem. 2001;276:40268–40273. doi: 10.1074/jbc.M107234200. [DOI] [PubMed] [Google Scholar]
  • 123.Shimizu K, Chiba S, Hosoya N, Kumano K, Saito T, Kurokawa M, Kanda Y, Hamada Y, Hirai H. Binding of Delta1, Jagged1, and Jagged2 to Notch2 rapidly induces cleavage, nuclear translocation, and hyperphosphorylation of Notch2. Mol Cell Biol. 2000;20:6913–6922. doi: 10.1128/mcb.20.18.6913-6922.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mizutani T, Taniguchi Y, Aoki T, Hashimoto N, Honjo T. Conservation of the biochemical mechanisms of signal transduction among mammalian Notch family members. Proc Natl Acad Sci U S A. 2001;98:9026–9031. doi: 10.1073/pnas.161269998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Fiuza UM, Arias AM. Cell and molecular biology of Notch. J Endocrinol. 2007;194:459–474. doi: 10.1677/JOE-07-0242. [DOI] [PubMed] [Google Scholar]
  • 126.Ikeuchi T, Sisodia SS. The Notch ligands, Delta1 and Jagged2, are substrates for presenilin-dependent “gamma-secretase” cleavage. J Biol Chem. 2003;278:7751–7754. doi: 10.1074/jbc.C200711200. [DOI] [PubMed] [Google Scholar]
  • 127.LaVoie MJ, Selkoe DJ. The Notch ligands, Jagged and Delta, are sequentially processed by alpha-secretase and presenilin/gamma-secretase and release signaling fragments. J Biol Chem. 2003;278:34427–34437. doi: 10.1074/jbc.M302659200. [DOI] [PubMed] [Google Scholar]
  • 128.Bland CE, Kimberly P, Rand MD. Notch-induced proteolysis and nuclear localization of the Delta ligand. J Biol Chem. 2003;278:13607–13610. doi: 10.1074/jbc.C300016200. [DOI] [PubMed] [Google Scholar]
  • 129.Six E, Ndiaye D, Laabi Y, Brou C, Gupta-Rossi N, Israel A, Logeat F. The Notch ligand Delta1 is sequentially cleaved by an ADAM protease and gamma-secretase. Proc Natl Acad Sci U S A. 2003;100:7638–7643. doi: 10.1073/pnas.1230693100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Marambaud P, Shioi J, Serban G, Georgakopoulos A, Sarner S, Nagy V, Baki L, Wen P, Efthimiopoulos S, Shao Z, Wisniewski T, Robakis NK. A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. EMBO J. 2002;21:1948–1956. doi: 10.1093/emboj/21.8.1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Parisiadou L, Fassa A, Fotinopoulou A, Bethani I, Efthimiopoulos S. Presenilin 1 and cadherins: stabilization of cell-cell adhesion and proteolysis-dependent regulation of transcription. Neurodegener Dis. 2004;1:184–191. doi: 10.1159/000080984. [DOI] [PubMed] [Google Scholar]
  • 132.Ferber EC, Kajita M, Wadlow A, Tobiansky L, Niessen C, Ariga H, Daniel J, Fujita Y. A role for the cleaved cytoplasmic domain of E-cadherin in the nucleus. J Biol Chem. 2008;283:12691–12700. doi: 10.1074/jbc.M708887200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Marambaud P, Wen PH, Dutt A, Shioi J, Takashima A, Siman R, Robakis NK. A CBP binding transcriptional repressor produced by the PS1/epsilon-cleavage of N-cadherin is inhibited by PS1 FAD mutations. Cell. 2003;114:635–645. doi: 10.1016/j.cell.2003.08.008. [DOI] [PubMed] [Google Scholar]
  • 134.Anders L, Mertins P, Lammich S, Murgia M, Hartmann D, Saftig P, Haass C, Ullrich A. Furin-, ADAM 10-, and gamma-secretase-mediated cleavage of a receptor tyrosine phosphatase and regulation of beta-catenin’s transcriptional activity. Mol Cell Biol. 2006;26:3917–3934. doi: 10.1128/MCB.26.10.3917-3934.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Haapasalo A, Kim DY, Carey BW, Turunen MK, Pettingell WH, Kovacs DM. Presenilin/gamma-secretase-mediated cleavage regulates association of leukocyte-common antigen-related (LAR) receptor tyrosine phosphatase with beta-catenin. J Biol Chem. 2007;282:9063–9072. doi: 10.1074/jbc.M611324200. [DOI] [PubMed] [Google Scholar]
  • 136.Louvi A, Artavanis-Tsakonas S. Notch signalling in vertebrate neural development. Nat Rev Neurosci. 2006;7:93–102. doi: 10.1038/nrn1847. [DOI] [PubMed] [Google Scholar]
  • 137.Linggi B, Cheng QC, Rao AR, Carpenter G. The ErbB-4 s80 intracellular domain is a constitutively active tyrosine kinase. Oncogene. 2006;25:160–163. doi: 10.1038/sj.onc.1209003. [DOI] [PubMed] [Google Scholar]
  • 138.Srinivasan R, Gillett CE, Barnes DM, Gullick WJ. Nuclear expression of the c-erbB-4/HER-4 growth factor receptor in invasive breast cancers. Cancer Res. 2000;60:1483–1487. [PubMed] [Google Scholar]
  • 139.Ni CY, Murphy MP, Golde TE, Carpenter G. Gamma-secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Science. 2001;294:2179–2181. doi: 10.1126/science.1065412. [DOI] [PubMed] [Google Scholar]
  • 140.Lee HJ, Jung KM, Huang YZ, Bennett LB, Lee JS, Mei L, Kim TW. Presenilin-dependent gamma-secretase-like intramembrane cleavage of ErbB4. J Biol Chem. 2002;277:6318–6323. doi: 10.1074/jbc.M110371200. [DOI] [PubMed] [Google Scholar]
  • 141.Williams CC, Allison JG, Vidal GA, Burow ME, Beckman BS, Marrero L, Jones FE. The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone. J Cell Biol. 2004;167:469–478. doi: 10.1083/jcb.200403155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Vidal GA, Naresh A, Marrero L, Jones FE. Presenilin-dependent gamma-secretase processing regulates multiple ERBB4/HER4 activities. J Biol Chem. 2005;280:19777–19783. doi: 10.1074/jbc.M412457200. [DOI] [PubMed] [Google Scholar]
  • 143.Lai C, Feng L. Implication of gamma-secretase in neuregulin-induced maturation of oligodendrocytes. Biochem Biophys Res Commun. 2004;314:535–542. doi: 10.1016/j.bbrc.2003.12.131. [DOI] [PubMed] [Google Scholar]
  • 144.Sardi SP, Murtie J, Koirala S, Patten BA, Corfas G. Presenilin-dependent ErbB4 nuclear signaling regulates the timing of astrogenesis in the developing brain. Cell. 2006;127:185–197. doi: 10.1016/j.cell.2006.07.037. [DOI] [PubMed] [Google Scholar]
  • 145.Bu G, Cam J, Zerbinatti C. LRP in amyloid-beta production and metabolism. Ann N Y Acad Sci. 2006;1086:35–53. doi: 10.1196/annals.1377.005. [DOI] [PubMed] [Google Scholar]
  • 146.Polavarapu R, An J, Zhang C, Yepes M. Regulated intramembrane proteolysis of the low-density lipoprotein receptor-related protein mediates ischemic cell death. Am J Pathol. 2008;172:1355–1362. doi: 10.2353/ajpath.2008.070975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Majdan M, Lachance C, Gloster A, Aloyz R, Zeindler C, Bamji S, Bhakar A, Belliveau D, Fawcett J, Miller FD, Barker PA. Transgenic mice expressing the intra-cellular domain of the p75 neurotrophin receptor undergo neuronal apoptosis. J Neurosci. 1997;17:6988–6998. doi: 10.1523/JNEUROSCI.17-18-06988.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kenchappa RS, Zampieri N, Chao MV, Barker PA, Teng HK, Hempstead BL, Carter BD. Ligand-dependent cleavage of the P75 neurotrophin receptor is necessary for NRIF nuclear translocation and apoptosis in sympathetic neurons. Neuron. 2006;50:219–232. doi: 10.1016/j.neuron.2006.03.011. [DOI] [PubMed] [Google Scholar]
  • 149.Yaar M, Zhai S, Pilch PF, Doyle SM, Eisenhauer PB, Fine RE, Gilchrest BA. Binding of beta-amyloid to the p75 neurotrophin receptor induces apoptosis. A possible mechanism for Alzheimer’s disease. J Clin Invest. 1997;100:2333–2340. doi: 10.1172/JCI119772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Costantini C, Rossi F, Formaggio E, Bernardoni R, Cecconi D, Della-Bianca V. Characterization of the signaling pathway downstream p75 neurotrophin receptor involved in beta-amyloid peptide-dependent cell death. J Mol Neurosci. 2005;25:141–156. doi: 10.1385/JMN:25:2:141. [DOI] [PubMed] [Google Scholar]
  • 151.Yan Z, Feng J. Alzheimer’s disease: interactions between cholinergic functions and beta-amyloid. Curr Alzheimer Res. 2004;1:241–248. doi: 10.2174/1567205043331992. [DOI] [PubMed] [Google Scholar]
  • 152.Wu CK, Thal L, Pizzo D, Hansen L, Masliah E, Geula C. Apoptotic signals within the basal forebrain cholin-ergic neurons in Alzheimer’s disease. Exp Neurol. 2005;195:484–496. doi: 10.1016/j.expneurol.2005.06.020. [DOI] [PubMed] [Google Scholar]
  • 153.Schliebs R. Basal forebrain cholinergic dysfunction in Alzheimer’s disease – interrelationship with beta-amyloid, inflammation and neurotrophin signaling. Neurochem Res. 2005;30:895–908. doi: 10.1007/s11064-005-6962-9. [DOI] [PubMed] [Google Scholar]
  • 154.Yeo TT, Chua-Couzens J, Butcher LL, Bredesen DE, Cooper JD, Valletta JS, Mobley WC, Longo FM. Absence of p75NTR causes increased basal forebrain cholinergic neuron size, choline acetyltransferase activity, and target innervation. J Neurosci. 1997;17:7594–7605. doi: 10.1523/JNEUROSCI.17-20-07594.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Volosin M, Song W, Almeida RD, Kaplan DR, Hempstead BL, Friedman WJ. Interaction of survival and death signaling in basal forebrain neurons: roles of neurotrophins and proneurotrophins. J Neurosci. 2006;26:7756–7766. doi: 10.1523/JNEUROSCI.1560-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Podlesniy P, Kichev A, Pedraza C, Saurat J, Encinas M, Perez B, Ferrer I, Espinet C. 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. 2006;169:119–131. doi: 10.2353/ajpath.2006.050787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Neeper M, Schmidt AM, Brett J, Yan SD, Wang F, Pan YC, Elliston K, Stern D, Shaw A. Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J Biol Chem. 1992;267:14998–15004. [PubMed] [Google Scholar]
  • 158.Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B. RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med. 2003;9:907–913. doi: 10.1038/nm890. [DOI] [PubMed] [Google Scholar]
  • 159.Arancio O, Zhang HP, Chen X, Lin C, Trinchese F, Puzzo D, Liu S, Hegde A, Yan SF, Stern A, Luddy JS, Lue LF, Walker DG, Roher A, Buttini M, Mucke L, Li W, Schmidt AM, Kindy M, Hyslop PA, Stern DM, Du Yan SS. RAGE potentiates Abeta-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004;23:4096–4105. doi: 10.1038/sj.emboj.7600415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Galichet A, Weibel M, Heizmann CW. Calcium-regulated intramembrane proteolysis of the RAGE receptor. Biochem Biophys Res Commun. 2008;370:1–5. doi: 10.1016/j.bbrc.2008.02.163. [DOI] [PubMed] [Google Scholar]
  • 161.Moore SW, Tessier-Lavigne M, Kennedy TE. Netrins and their receptors. Adv Exp Med Biol. 2007;621:17–31. doi: 10.1007/978-0-387-76715-4_2. [DOI] [PubMed] [Google Scholar]
  • 162.Parent AT, Barnes NY, Taniguchi Y, Thinakaran G, Sisodia SS. Presenilin attenuates receptor-mediated signaling and synaptic function. J Neurosci. 2005;25:1540–1549. doi: 10.1523/JNEUROSCI.3850-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Tomita T, Tanaka S, Morohashi Y, Iwatsubo T. Presenilin-dependent intramembrane cleavage of ephrin-B1. Mol Neurodegener. 2006;1:2. doi: 10.1186/1750-1326-1-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Georgakopoulos A, Litterst C, Ghersi E, Baki L, Xu C, Serban G, Robakis NK. Metalloproteinase/Presenilin1 processing of ephrinB regulates EphB-induced Src phosphorylation and signaling. EMBO J. 2006;25:1242–1252. doi: 10.1038/sj.emboj.7601031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Waschbusch D, Born S, Niediek V, Kirchgessner N, Tamboli IY, Walter J, Merkel R, Hoffmann B. Presenilin 1 affects focal adhesion site formation and cell force generation via c-Src transcriptional and posttranslational regulation. J Biol Chem. 2009;284:10138–10149. doi: 10.1074/jbc.M806825200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Litterst C, Georgakopoulos A, Shioi J, Ghersi E, Wisniewski T, Wang R, Ludwig A, Robakis NK. Ligand binding and calcium influx induce distinct ectodomain/gamma-secretase-processing pathways of EphB2 receptor. J Biol Chem. 2007;282:16155–16163. doi: 10.1074/jbc.M611449200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Inoue E, Deguchi-Tawarada M, Togawa A, Matsui C, Arita K, Katahira-Tayama S, Sato T, Yamauchi E, Oda Y, Takai Y. Synaptic activity prompts gamma-secretase-mediated cleavage of EphA4 and dendritic spine formation. J Cell Biol. 2009;185:551–564. doi: 10.1083/jcb.200809151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ito K, Okamoto I, Araki N, Kawano Y, Nakao M, Fujiyama S, Tomita K, Mimori T, Saya H. Calcium influx triggers the sequential proteolysis of extracellular and cytoplasmic domains of E-cadherin, leading to loss of beta-catenin from cell-cell contacts. Oncogene. 1999;18:7080–7090. doi: 10.1038/sj.onc.1203191. [DOI] [PubMed] [Google Scholar]
  • 169.Kim DY, Ingano LA, Kovacs DM. Nectin-1alpha, an immunoglobulin-like receptor involved in the formation of synapses, is a substrate for presenilin/gamma-secretase-like cleavage. J Biol Chem. 2002;277:49976–49981. doi: 10.1074/jbc.M210179200. [DOI] [PubMed] [Google Scholar]
  • 170.Kim DY, Ingano LA, Carey BW, Pettingell WH, Kovacs DM. Presenilin/gamma-secretase-mediated cleavage of the voltage-gated sodium channel beta2-subunit regulates cell adhesion and migration. J Biol Chem. 2005;280:23251–23261. doi: 10.1074/jbc.M412938200. [DOI] [PubMed] [Google Scholar]
  • 171.Cai J, Jiang WG, Grant MB, Boulton M. Pigment epithelium-derived factor inhibits angiogenesis via regulated intracellular proteolysis of vascular endothelial growth factor receptor 1. J Biol Chem. 2006;281:3604–3613. doi: 10.1074/jbc.M507401200. [DOI] [PubMed] [Google Scholar]
  • 172.Pelletier L, Guillaumot P, Freche B, Luquain C, Christiansen D, Brugiere S, Garin J, Manie SN. Gamma-secretase-dependent proteolysis of CD44 promotes neoplastic transformation of rat fibroblastic cells. Cancer Res. 2006;66:3681–3687. doi: 10.1158/0008-5472.CAN-05-3870. [DOI] [PubMed] [Google Scholar]
  • 173.Arasada RR, Carpenter G. Secretase-dependent tyrosine phosphorylation of Mdm2 by the ErbB-4 intracellular domain fragment. J Biol Chem. 2005;280:30783–30787. doi: 10.1074/jbc.M506057200. [DOI] [PubMed] [Google Scholar]
  • 174.Junttila TT, Sundvall M, Lundin M, Lundin J, Tanner M, Harkonen P, Joensuu H, Isola J, Elenius K. Cleavable ErbB4 isoform in estrogen receptor-regulated growth of breast cancer cells. Cancer Res. 2005;65:1384–1393. doi: 10.1158/0008-5472.CAN-04-3150. [DOI] [PubMed] [Google Scholar]
  • 175.Sachse CC, Kovacs DM, Kim DY. BACE1 and presenilin/gamma-secretase cleave the beta subunits of the voltage-gated potassium channels. Soc Neurosci Abstr, Program No. P854.22 2010 [Google Scholar]
  • 176.Sturm RA, Teasdale RD, Box NF. Human pigmentation genes: identification, structure and consequences of polymorphic variation. Gene. 2001;277:49–62. doi: 10.1016/s0378-1119(01)00694-1. [DOI] [PubMed] [Google Scholar]
  • 177.Oetting WS, King RA. Molecular basis of albinism: mutations and polymorphisms of pigmentation genes associated with albinism. Hum Mutat. 1999;13:99–115. doi: 10.1002/(SICI)1098-1004(1999)13:2<99::AID-HUMU2>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 178.Wang R, Tang P, Wang P, Boissy RE, Zheng H. Regulation of tyrosinase trafficking and processing by pre-senilins: partial loss of function by familial Alzheimer’s disease mutation. Proc Natl Acad Sci U S A. 2006;103:353–358. doi: 10.1073/pnas.0509822102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yan R, McCarthy J. Presenilin and γ-secretase activity: a viable therapeutic target for Alzheimer’s disease? CSTT. 2010;5:128–140. [Google Scholar]
  • 180.Wolfe MS. Inhibition and modulation of gamma-secretase for Alzheimer’s disease. Neurotherapeutics. 2008;5:391–398. doi: 10.1016/j.nurt.2008.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Petit A, Bihel F, Alves da Costa C, Pourquie O, Checler F, Kraus JL. New protease inhibitors prevent gamma-secretase-mediated production of Abeta40/42 without affecting Notch cleavage. Nat Cell Biol. 2001;3:507–511. doi: 10.1038/35074581. [DOI] [PubMed] [Google Scholar]
  • 182.Kounnas MZ, Danks AM, Cheng S, Tyree C, Ackerman E, Zhang X, Ahn K, Nguyen P, Comer D, Mao L, Yu C, Pleynet D, Digregorio PJ, Velicelebi G, Stauderman KA, Comer WT, Mobley WC, Li YM, Sisodia SS, Tanzi RE, Wagner SL. Modulation of gamma-secretase reduces beta-amyloid deposition in a transgenic mouse model of Alzheimer’s disease. Neuron. 2010;67:769–780. doi: 10.1016/j.neuron.2010.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Li T, Wen H, Brayton C, Laird FM, Ma G, Peng S, Placanica L, Wu TC, Crain BJ, Price DL, Eberhart CG, Wong PC. Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. J Neurosci. 2007;27:10849–10859. doi: 10.1523/JNEUROSCI.2152-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Araki Y, Tomita S, Yamaguchi H, Miyagi N, Sumioka A, Kirino Y, Suzuki T. Novel cadherin-related membrane proteins, Alcadeins, enhance the X11-like protein-mediated stabilization of amyloid beta-protein precursor metabolism. J Biol Chem. 2003;278:49448–49458. doi: 10.1074/jbc.M306024200. [DOI] [PubMed] [Google Scholar]
  • 185.Eggert S, Paliga K, Soba P, Evin G, Masters CL, Weidemann A, Beyreuther K. The proteolytic processing of the amyloid precursor protein gene family members APLP-1 and APLP-2 involves alpha-, beta-, gamma-, and epsilon-like cleavages: modulation of APLP-1 processing by n-glycosylation. J Biol Chem. 2004;279:18146–18156. doi: 10.1074/jbc.M311601200. [DOI] [PubMed] [Google Scholar]
  • 186.May P, Reddy YK, Herz J. Proteolytic processing of low density lipoprotein receptor-related protein mediates regulated release of its intracellular domain. J Biol Chem. 2002;277:18736–18743. doi: 10.1074/jbc.M201979200. [DOI] [PubMed] [Google Scholar]
  • 187.Hoe HS, Rebeck GW. Regulation of ApoE receptor proteolysis by ligand binding. Brain Res Mol Brain Res. 2005;137:31–39. doi: 10.1016/j.molbrainres.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 188.Stoeck A, Shang L, Dempsey PJ. Sequential and gamma-secretase-dependent processing of the betacellulin precursor generates a palmitoylated intracellular-domain fragment that inhibits cell growth. J Cell Sci. 2010;123:2319–2331. doi: 10.1242/jcs.060830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Blair CR, Stone JB, Wells RG. The type III TGF-beta receptor betaglycan transmembrane-cytoplasmic domain fragment is stable after ectodomain cleavage and is a substrate of the intramembrane protease gamma-secretase. Biochim Biophys Acta. 2011;1813:332–339. doi: 10.1016/j.bbamcr.2010.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Andersson CX, Fernandez-Rodriguez J, Laos S, Baeckstrom D, Haass C, Hansson GC. Shedding and gamma-secretase-mediated intramembrane proteolysis of the mucin-type molecule CD43. Biochem J. 2005;387:377–384. doi: 10.1042/BJ20041387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Andersson CX, Fernandez-Rodriguez J, Laos S, Sikut R, Sikut A, Baeckstrom D, Hansson GC. CD43 has a functional NLS, interacts with beta-catenin, and affects gene expression. Biochem Biophys Res Commun. 2004;316:12–17. doi: 10.1016/j.bbrc.2004.02.011. [DOI] [PubMed] [Google Scholar]
  • 192.Mambole A, Baruch D, Nusbaum P, Bigot S, Suzuki M, Lesavre P, Fukuda M, Halbwachs-Mecarelli L. The cleavage of neutrophil leukosialin (CD43) by cathepsin G releases its extracellular domain and triggers its intramem-brane proteolysis by presenilin/gamma-secretase. J Biol Chem. 2008;283:23627–23635. doi: 10.1074/jbc.M710286200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Lammich S, Okochi M, Takeda M, Kaether C, Capell A, Zimmer AK, Edbauer D, Walter J, Steiner H, Haass C. Presenilin-dependent intramembrane proteolysis of CD44 leads to the liberation of its intracellular domain and the secretion of an Abeta-like peptide. J Biol Chem. 2002;277:44754–44759. doi: 10.1074/jbc.M206872200. [DOI] [PubMed] [Google Scholar]
  • 194.Murakami D, Okamoto I, Nagano O, Kawano Y, Tomita T, Iwatsubo T, De Strooper B, Yumoto E, Saya H. Presenilin-dependent gamma-secretase activity mediates the intramembranous cleavage of CD44. Oncogene. 2003;22:1511–1516. doi: 10.1038/sj.onc.1206298. [DOI] [PubMed] [Google Scholar]
  • 195.Cui W, Ke JZ, Zhang Q, Ke HZ, Chalouni C, Vignery A. The intracellular domain of CD44 promotes the fusion of macrophages. Blood. 2006;107:796–805. doi: 10.1182/blood-2005-05-1902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Wilhelmsen K, van der Geer P. Phorbol 12-myristate 13-acetate-induced release of the colony-stimulating factor 1 receptor cytoplasmic domain into the cytosol involves two separate cleavage events. Mol Cell Biol. 2004;24:454–464. doi: 10.1128/MCB.24.1.454-464.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Schulte A, Schulz B, Andrzejewski MG, Hundhausen C, Mletzko S, Achilles J, Reiss K, Paliga K, Weber C, John SR, Ludwig A. Sequential processing of the trans-membrane chemokines CX3CL1 and CXCL16 by alpha-and gamma-secretases. Biochem Biophys Res Commun. 2007;358:233–240. doi: 10.1016/j.bbrc.2007.04.100. [DOI] [PubMed] [Google Scholar]
  • 198.Chu J, Jeffries S, Norton JE, Capobianco AJ, Bresnick EH. Repression of activator protein-1-mediated transcriptional activation by the Notch-1 intracellular domain. J Biol Chem. 2002;277:7587–7597. doi: 10.1074/jbc.M111044200. [DOI] [PubMed] [Google Scholar]
  • 199.Omerovic J, Puggioni EM, Napoletano S, Visco V, Fraioli R, Frati L, Gulino A, Alimandi M. Ligand-regulated association of ErbB-4 to the transcriptional co-activator YAP65 controls transcription at the nuclear level. Exp Cell Res. 2004;294:469–479. doi: 10.1016/j.yexcr.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 200.Cowan JW, Wang X, Guan R, He K, Jiang J, Baumann G, Black RA, Wolfe MS, Frank SJ. Growth hormone receptor is a target for presenilin-dependent gamma-secretase cleavage. J Biol Chem. 2005;280:19331–19342. doi: 10.1074/jbc.M500621200. [DOI] [PubMed] [Google Scholar]
  • 201.Carey BW, Kim DY, Kovacs DM. Presenilin/gamma-secretase and alpha-secretase-like peptidases cleave human MHC class I proteins. Biochem J. 2007;401:121–127. doi: 10.1042/BJ20060847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Saleh AZ, Fang AT, Arch AE, Neupane D, El Fiky A, Krolewski JJ. Regulated proteolysis of the IFNaR2 subunit of the interferon-alpha receptor. Oncogene. 2004;23:7076–7086. doi: 10.1038/sj.onc.1207955. [DOI] [PubMed] [Google Scholar]
  • 203.El Fiky A, Arch AE, Krolewski JJ. Intracellular domain of the IFNaR2 interferon receptor subunit mediates transcription via Stat2. J Cell Physiol. 2005;204:567–573. doi: 10.1002/jcp.20305. [DOI] [PubMed] [Google Scholar]
  • 204.McElroy B, Powell JC, McCarthy JV. The insulin-like growth factor 1 (IGF-1) receptor is a substrate for gamma-secretase-mediated intramembrane proteolysis. Biochem Biophys Res Commun. 2007;358:1136–1141. doi: 10.1016/j.bbrc.2007.05.062. [DOI] [PubMed] [Google Scholar]
  • 205.Elzinga BM, Twomey C, Powell JC, Harte F, McCarthy JV. Interleukin-1 receptor type 1 is a substrate for gamma-secretase-dependent regulated intramembrane pro-teolysis. J Biol Chem. 2009;284:1394–1409. doi: 10.1074/jbc.M803108200. [DOI] [PubMed] [Google Scholar]
  • 206.Kuhn PH, Marjaux E, Imhof A, De Strooper B, Haass C, Lichtenthaler SF. Regulated intramembrane proteolysis of the interleukin-1 receptor II by alpha-, beta-, and gamma-secretase. J Biol Chem. 2007;282:11982–11995. doi: 10.1074/jbc.M700356200. [DOI] [PubMed] [Google Scholar]
  • 207.Chalaris A, Gewiese J, Paliga K, Fleig L, Schneede A, Krieger K, Rose-John S, Scheller J. ADAM17-mediated shedding of the IL6R induces cleavage of the membrane stub by gamma-secretase. Biochim Biophys Acta. 2010;1803:234–245. doi: 10.1016/j.bbamcr.2009.12.001. [DOI] [PubMed] [Google Scholar]
  • 208.Kasuga K, Kaneko H, Nishizawa M, Onodera O, Ikeuchi T. Generation of intracellular domain of insulin receptor tyrosine kinase by gamma-secretase. Biochem Biophys Res Commun. 2007;360:90–96. doi: 10.1016/j.bbrc.2007.06.022. [DOI] [PubMed] [Google Scholar]
  • 209.Niwa M, Sidrauski C, Kaufman RJ, Walter P. A role for presenilin-1 in nuclear accumulation of Ire1 fragments and induction of the mammalian unfolded protein response. Cell. 1999;99:691–702. doi: 10.1016/s0092-8674(00)81667-0. [DOI] [PubMed] [Google Scholar]
  • 210.Bloch L, Sineshchekova O, Reichenbach D, Reiss K, Saftig P, Kuro-o M, Kaether C. Klotho is a substrate for alpha-, beta- and gamma-secretase. FEBS Lett. 2009;583:3221–3224. doi: 10.1016/j.febslet.2009.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Maretzky T, Schulte M, Ludwig A, Rose-John S, Blobel C, Hartmann D, Altevogt P, Saftig P, Reiss K. L1 is sequentially processed by two differently activated metal-loproteases and presenilin/gamma-secretase and regulates neural cell adhesion, cell migration, and neurite outgrowth. Mol Cell Biol. 2005;25:9040–9053. doi: 10.1128/MCB.25.20.9040-9053.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Kinoshita A, Shah T, Tangredi MM, Strickland DK, Hyman BT. The intracellular domain of the low density lipoprotein receptor-related protein modulates transactiva-tion mediated by amyloid precursor protein and Fe65. J Biol Chem. 2003;278:41182–41188. doi: 10.1074/jbc.M306403200. [DOI] [PubMed] [Google Scholar]
  • 213.Liu CX, Ranganathan S, Robinson S, Strickland DK. gamma-Secretase-mediated release of the low density lipoprotein receptor-related protein 1B intracellular domain suppresses anchorage-independent growth of neu-roglioma cells. J Biol Chem. 2007;282:7504–7511. doi: 10.1074/jbc.M608088200. [DOI] [PubMed] [Google Scholar]
  • 214.Zou Z, Chung B, Nguyen T, Mentone S, Thomson B, Biemesderfer D. Linking receptor-mediated endocytosis and cell signaling: evidence for regulated intramembrane proteolysis of megalin in proximal tubule. J Biol Chem. 2004;279:34302–34310. doi: 10.1074/jbc.M405608200. [DOI] [PubMed] [Google Scholar]
  • 215.Mi K, Johnson GV. Regulated proteolytic processing of LRP6 results in release of its intracellular domain. J Neurochem. 2007;101:517–529. doi: 10.1111/j.1471-4159.2007.04447.x. [DOI] [PubMed] [Google Scholar]
  • 216.Julian J, Dharmaraj N, Carson DD. MUC1 is a substrate for gamma-secretase. J Cell Biochem. 2009;108:802–815. doi: 10.1002/jcb.22292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Uemura K, Kihara T, Kuzuya A, Okawa K, Nishimoto T, Ninomiya H, Sugimoto H, Kinoshita A, Shimohama S. Characterization of sequential N-cadherin cleavage by ADAM10 and PS1. Neurosci Lett. 2006;402:278–283. doi: 10.1016/j.neulet.2006.04.018. [DOI] [PubMed] [Google Scholar]
  • 218.Wong HK, Sakurai T, Oyama F, Kaneko K, Wada K, Miyazaki H, Kurosawa M, De Strooper B, Saftig P, Nukina N. beta Subunits of voltage-gated sodium channels are novel substrates of beta-site amyloid precursor protein-cleaving enzyme (BACE1) and gamma-secretase. J Biol Chem. 2005;280:23009–23017. doi: 10.1074/jbc.M414648200. [DOI] [PubMed] [Google Scholar]
  • 219.Bao J, Wolpowitz D, Role LW, Talmage DA. Back signaling by the Nrg-1 intracellular domain. J Cell Biol. 2003;161:1133–1141. doi: 10.1083/jcb.200212085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Bao J, Lin H, Ouyang Y, Lei D, Osman A, Kim TW, Mei L, Dai P, Ohlemiller KK, Ambron RT. Activity-dependent transcription regulation of PSD-95 by neuregulin-1 and Eos. Nat Neurosci. 2004;7:1250–1258. doi: 10.1038/nn1342. [DOI] [PubMed] [Google Scholar]
  • 221.Coolen MW, van Loo KM, van Bakel NN, Ellenbroek BA, Cools AR, Martens GJ. Reduced Aph-1 b expression causes tissue- and substrate-specific changes in gamma-secretase activity in rats with a complex phenotype. FASEB J. 2006;20:175–177. doi: 10.1096/fj.05-4337fje. [DOI] [PubMed] [Google Scholar]
  • 222.Gowrishankar K, Zeidler MG, Vincenz C. Release of a membrane-bound death domain by gamma-secretase processing of the p75NTR homolog NRADD. J Cell Sci. 2004;117:4099–4111. doi: 10.1242/jcs.01263. [DOI] [PubMed] [Google Scholar]
  • 223.Jung KM, Tan S, Landman N, Petrova K, Murray S, Lewis R, Kim PK, Kim DS, Ryu SH, Chao MV, Kim TW. Regulated intramembrane proteolysis of the p75 neurotrophin receptor modulates its association with the TrkA receptor. J Biol Chem. 2003;278:42161–42169. doi: 10.1074/jbc.M306028200. [DOI] [PubMed] [Google Scholar]
  • 224.Kanning KC, Hudson M, Amieux PS, Wiley JC, Bothwell M, Schecterson LC. Proteolytic processing of the p75 neurotrophin receptor and two homologs generates C-terminal fragments with signaling capability. J Neurosci. 2003;23:5425–5436. doi: 10.1523/JNEUROSCI.23-13-05425.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Zampieri N, Xu CF, Neubert TA, Chao MV. Cleavage of p75 neurotrophin receptor by alpha-secretase and gamma-secretase requires specific receptor domains. J Biol Chem. 2005;280:14563–14571. doi: 10.1074/jbc.M412957200. [DOI] [PubMed] [Google Scholar]
  • 226.Domeniconi M, Zampieri N, Spencer T, Hilaire M, Mellado W, Chao MV, Filbin MT. MAG induces regulated intramembrane proteolysis of the p75 neurotrophin receptor to inhibit neurite outgrowth. Neuron. 2005;46:849–855. doi: 10.1016/j.neuron.2005.05.029. [DOI] [PubMed] [Google Scholar]
  • 227.Ceni C, Kommaddi RP, Thomas R, Vereker E, Liu X, McPherson PS, Ritter B, Barker PA. The p75NTR intracellular domain generated by neurotrophin-induced receptor cleavage potentiates Trk signaling. J Cell Sci. 2010;123:2299–2307. doi: 10.1242/jcs.062612. [DOI] [PubMed] [Google Scholar]
  • 228.Rajagopal C, Stone KL, Mains RE, Eipper BA. Secretion stimulates intramembrane proteolysis of a secretory granule membrane enzyme. J Biol Chem. 2010;285:34632–34642. doi: 10.1074/jbc.M110.145334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Kaimori JY, Nagasawa Y, Menezes LF, Garcia-Gonzalez MA, Deng J, Imai E, Onuchic LF, Guay-Woodford LM, Germino GG. Polyductin undergoes notch-like processing and regulated release from primary cilia. Hum Mol Genet. 2007;16:942–956. doi: 10.1093/hmg/ddm039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Hiesberger T, Gourley E, Erickson A, Koulen P, Ward CJ, Masyuk TV, Larusso NF, Harris PC, Igarashi P. Proteolytic cleavage and nuclear translocation of fibrocystin is regulated by intracellular Ca2+ and activation of protein kinase C. J Biol Chem. 2006;281:34357–34364. doi: 10.1074/jbc.M606740200. [DOI] [PubMed] [Google Scholar]
  • 231.Bonn S, Seeburg PH, Schwarz MK. Combinatorial expression of alpha- and gamma-protocadherins alters their presenilin-dependent processing. Mol Cell Biol. 2007;27:4121–4132. doi: 10.1128/MCB.01708-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Haas IG, Frank M, Veron N, Kemler R. Presenilin-dependent processing and nuclear function of gamma-protocadherins. J Biol Chem. 2005;280:9313–9319. doi: 10.1074/jbc.M412909200. [DOI] [PubMed] [Google Scholar]
  • 233.Hambsch B, Grinevich V, Seeburg PH, Schwarz MK. {gamma}-Protocadherins, presenilin-mediated release of C-terminal fragment promotes locus expression. J Biol Chem. 2005;280:15888–15897. doi: 10.1074/jbc.M414359200. [DOI] [PubMed] [Google Scholar]
  • 234.Chow JP, Fujikawa A, Shimizu H, Suzuki R, Noda M. Metalloproteinase- and gamma-secretase-mediated cleavage of protein-tyrosine phosphatase receptor type Z. J Biol Chem. 2008;283:30879–30889. doi: 10.1074/jbc.M802976200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Seki M, Watanabe A, Enomoto S, Kawamura T, Ito H, Kodama T, Hamakubo T, Aburatani H. Human ROBO1 is cleaved by metalloproteinases and gamma-secretase and migrates to the nucleus in cancer cells. FEBS Lett. 2010;584:2909–2915. doi: 10.1016/j.febslet.2010.05.009. [DOI] [PubMed] [Google Scholar]
  • 236.Hermey G, Sjogaard SS, Petersen CM, Nykjaer A, Gliemann J. Tumour necrosis factor alpha-converting enzyme mediates ectodomain shedding of Vps10p-domain receptor family members. Biochem J. 2006;395:285–293. doi: 10.1042/BJ20051364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Nyborg AC, Ladd TB, Zwizinski CW, Lah JJ, Golde TE. Sortilin, SorCS1b, and SorLA Vps10 p sorting receptors, are novel gamma-secretase substrates. Mol Neurodegener. 2006;1:3. doi: 10.1186/1750-1326-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Bohm C, Seibel NM, Henkel B, Steiner H, Haass C, Hampe W. SorLA signaling by regulated intramembrane proteolysis. J Biol Chem. 2006;281:14547–14553. doi: 10.1074/jbc.M601660200. [DOI] [PubMed] [Google Scholar]
  • 239.Schulz JG, Annaert W, Vandekerckhove J, Zimmermann P, De Strooper B, David G. Syndecan 3 intramembrane proteolysis is presenilin/gamma-secretase-dependent and modulates cytosolic signaling. J Biol Chem. 2003;278:48651–48657. doi: 10.1074/jbc.M308424200. [DOI] [PubMed] [Google Scholar]
  • 240.Marron MB, Singh H, Tahir TA, Kavumkal J, Kim HZ, Koh GY, Brindle NP. Regulated proteolytic processing of Tie1 modulates ligand responsiveness of the receptor-tyrosine kinase Tie2. J Biol Chem. 2007;282:30509–30517. doi: 10.1074/jbc.M702535200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Kornilova AY, Kim J, Laudon H, Wolfe MS. Deducing the transmembrane domain organization of presenilin-1 in gamma-secretase by cysteine disulfide cross-linking. Biochemistry. 2006;45:7598–7604. doi: 10.1021/bi060107k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Fortna RR, Crystal AS, Morais VA, Pijak DS, Lee VM, Doms RW. Membrane topology and nicastrin-enhanced endoproteolysis of APH-1, a component of the gamma-secretase complex. J Biol Chem. 2004;279:3685–3693. doi: 10.1074/jbc.M310505200. [DOI] [PubMed] [Google Scholar]
  • 243.Crystal AS, Morais VA, Pierson TC, Pijak DS, Carlin D, Lee VM, Doms RW. Membrane topology of gamma-secretase component PEN-2. J Biol Chem. 2003;278:20117–20123. doi: 10.1074/jbc.M213107200. [DOI] [PubMed] [Google Scholar]

RESOURCES