Abstract

The γ-secretase protein complex executes the intramembrane proteolysis of amyloid precursor protein (APP), which releases Alzheimer disease β-amyloid peptide. In addition to APP, γ-secretase also cleaves several other type I membrane protein substrates including Notch1 and N-cadherin. γ-Secretase is made of four integral transmembrane protein subunits: presenilin (PS), nicastrin, APH1, and PEN2. Multiple lines of evidence indicate that a heteromer of PS-derived N- and C-terminal fragments functions as the catalytic subunit of γ-secretase. Only limited information is available on the domains within each subunit involved in the recognition and recruitment of diverse substrates and the transfer of substrates to the catalytic site. Here, we performed mutagenesis of two domains of PS1, namely the first luminal loop domain (LL1) and the second transmembrane domain (TM2), and analyzed PS1 endoproteolysis as well as the catalytic activities of PS1 toward APP, Notch, and N-cadherin. Our results show that distinct residues within LL1 and TM2 domains as well as the length of the LL1 domain are critical for PS1 endoproteolysis, but not for PS1 complex formation with nicastrin, APH1, and PEN2. Furthermore, our experimental PS1 mutants formed γ-secretase complexes with distinct catalytic properties toward the three substrates examined in this study; however, the mutations did not affect PS1 interaction with the substrates. We conclude that the N-terminal LL1 and TM2 domains are critical for PS1 endoproteolysis and the coordination between the putative substrate-docking site and the catalytic core of the γ-secretase.

Highlights

  • By introducing mutations in LL1 and TM2, we identified residues that are critical for PS1 processing and/or ␥-secretase activity toward three substrates, APP, Notch, and N-cadherin (Table 1)

  • We find that PS1 endoproteolysis is severely impaired in four LL1 mutants (MutBLL1, 8aaLL1, 16aaLL1, and 19/22AlaLL1) and three TM2 mutants (TM2-aa1/4, TM2aa16/22, and 18/22AlaTM2) (Figs. 2 and 4)

  • The results of our study show that LL1 and TM2 of PS1 are critical for PS1 endoproteolysis and for substrate catalysis and/or coordinating the transfer of docked substrates to the catalytic site of ␥-secretase for proteolysis

Read more

Summary

EXPERIMENTAL PROCEDURES

Plasmids and Retrovirus-mediated Gene Expression—The cDNA fragments encoding PS1 mutants were generated as Asp718 and PflMI fragments by two-step PCR mutagenesis and cloned by exchanging the corresponding segment in human WT PS1 expression plasmid. With the exception of LL1Ins and 28/32Ala, all other LL1 mutants we examined failed to generate NICD and accumulated Notch⌬EMV-6myc precursor at levels comparable with that found in PS1Ϫ/Ϫ/PS2Ϫ/Ϫ vector MEF lacking ␥-secretase activity (Fig. 3B, middle panel). Western blot analysis of lysates showed that despite the differences in the steady-state levels of PS1 NTF and CTF generated from the TM2 mutants, stable expression of each mutant restored nicastrin maturation and PEN2 protein stability, suggesting that each mutant was able to interact with these subunits. Analysis of APP processing following transient expression of full-length APPSwe showed a marked loss of ␥-secretase activity toward processing of APP in the TM2 mutants aa1/4, aa16/22, 1/3Ala, and 18/22Ala, evidenced by the accumulation of APP CTFs and reduced secretion of A␤40 peptides (Fig. 5A). Detergent-resistant lipid raft association of PS1 variants tested in this study largely reflects their ability to assemble with other ␥-secretase subunits and their endoproteolysis status

DISCUSSION
MutA ϩϩ ϩ ϩ
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call