Elsevier

Pharmacological Research

Volume 50, Issue 4, October 2004, Pages 411-418
Pharmacological Research

Identification of the role of presenilins beyond Alzheimer’s disease

https://doi.org/10.1016/j.phrs.2003.12.026Get rights and content

Abstract

Mutations in the genes encoding presenilin 1 (PS1) and presenilin 2 (PS2) account for the majority of the cases of familial early-onset Alzheimer’s disease (FAD). Presenilins (PSs) facilitate the intramembraneous cleavage of amyloid precursor protein (APP), coined γ-secretase cleavage, which generates β-amyloid peptides (Aβ). Considerable evidence suggests that FAD-linked PS variants exert their pathogenic influence by selectively elevating the levels of highly fibrillogenic Aβ42 peptides. In addition, numerous other functions have been ascribed to PSs based on subcellular localization, protein interactions, loss of function studies, and intramembraneous γ-secretase cleavage of growing number of substrates. This review summarizes the diverse physiological functions that are regulated by PSs beyond APP metabolism.

Introduction

Since the first description of a genetic link between PS and AD, these homologous polytopic membrane proteins have become the focus of several research groups interested in the molecular mechanisms of Alzheimer’s disease (AD). PS1 functions as a multiprotein complex comprised of at least three other transmembrane proteins namely nicastrin, Aph-1 and Pen-2. Biogenesis of these four proteins and their assembly into high molecular weight complex are highly regulated processes that depend on the availability of each of the component polypeptides in stoichiometric concentrations [1], [2], [3]. PS1 is synthesized as a 42- to 43-kDa polypeptide that undergoes endoproteolytic processing within the cytoplasmic loop connecting putative transmembrane segments 6 and 7 to generate stable 27- to 28-kDa N-terminal (NTF) and 16- to 17-kDa C-terminal (CTF) proteolytic derivatives [4]. In transfected cells only a fraction of overexpressed PS1 is converted to stable NTF and CTF, whereas the majority of nascent PS1 polypeptide is rapidly degraded, indicating that the accumulation of PS1 NTF and CTF is tightly regulated and saturable [4], [5]. Thus, endoproteolysis of PSs is a highly conserved process and, arguably, a processing event that regulates the accumulation of fragments, and at least some biological functions of PS. Direct evidence that the NTF/CTF assembly is part of the active γ-secretase enzyme complex came from studies that developed transition state analogue inhibitors of aspartyl proteases. Two such reagents specifically bound to PS1 NTF and CTF, and not to PS1 holoprotein, supporting the notion that endoproteolytic derivatives are the biologically active forms of PS1 [6], [7]. Recent evidence shows that PS1 endoproteolysis and accumulation of fragments are regulated by the availability of nicastrin, Aph-1 and Pen-2 [1], [2], [3]. Human PS1 expressed in yeast along with these three cofactors undergoes proteolytic processing to generate stable PS1 NTF and CTF [8]. Furthermore, coexpression of all four proteins is sufficient to overcome the limitation in generating excess PS-derived NTF and CTF in transfected cells [1], [2], [3].

Genetic, biochemical, and pharmacological evidence points to an active role for nicastrin, Aph-1 and Pen-2 in PS1-mediated intramembraneous γ-secretase processing of select type I membrane proteins. All three proteins were first identified by genetic screens performed in Caenorhabditis elegans and subsequently shown to be stoichiometric components of high molecular weight complexes that exhibit γ-secretase activity. Interestingly, each of these proteins appears to be codependent for biogenesis, maturation and stability. For example, the highly glycosylated type I membrane protein nicastrin does not mature and exit the ER in cells lacking PS1 expression [9]. Conversely, PS1 fails to undergo endoproteolysis to generate stable NTF and CTF in nicastrin−/− cells [10]. Thus, it is clear now that γ-secretase activity depends on stoichiometric expression of each of these components and their proper assembly and trafficking to appropriate subcellular location. Details on the assembly of the γ-secretase complex are only beginning to emerge, and available evidence supports the formation of an early intermediate sub-complex of Aph-1 and nicastrin [11]. Unfortunately, several pharmacological inhibitors selected based on their ability to inhibit Aβ production do not seem to markedly affect PS1 complex formation and localization [12], [13], thus presenting researchers with the challenge of developing additional unique reagents to explore the details of the PS1/γ-secretase complex assembly process.

Section snippets

Intramembraneous cleavage of type I membrane proteins

In the past few years there has been accumulating interest in understanding regulated cleavage of type I membrane proteins within their transmembrane domains (TMs). In keeping with the nomenclature of previously described α- and β-secretase cleavage of APP within the extracellular domain, the intramembraneous cleavage of APP was termed the “γ-secretase” cleavage. In the case of APP, γ-secretase cleavage at two major sites within the TM was identified, which result in the generation of Aβ

Presenilin-interacting proteins

As described above, PS-derived NTF and CTF are components of high molecular weight complexes. Over the past few years several investigators employed yeast two-hybrid assays and candidate approaches to identify a growing number of proteins that interact with various domains of PS1 or PS2 [31]. The list of PS-interacting proteins includes: members of a family of armadillo-related proteins, including β-catenin (see the following); cell-surface transmembrane protein E-cadherin; filamin, an

Functional role for PS1 in regulating β-catenin

The most interesting information uncovered by the two-hybrid binding studies employing PS1 is the association with members of a family of armadillo-related proteins. First described by Zhou et al. [34], PS has been shown to interact with β-catenin, γ-catenin, δ-catenin, p0001, and neural-specific plakophilin. Since β-catenin is a multifunctional protein involved in Wnt signal transduction, cell adhesion and tumor progression, the functional significance of PS β-catenin interaction has been

PS1 and neurogenesis

Notch activity is essential for a wide variety of cell fate decisions during development [45]. In the adult, Notch activity also plays a fundamental role in the adult in regulating neurite outgrowth, maintenance of the hematopoietic system, etc. A recent review by Selkoe and Kopan [24] has detailed description of how PS1 function impacts on Notch activity during development. Notch receptors and ligands continue to be expressed in post-mitotic cortical neurons, and cytoplasmic domain of

PS1 function in cell adhesion and synapse formation

PS1 is ubiquitously expressed in peripheral tissue and in the nervous system. Several studies have investigated the subcellular localization PS1 in neurons using biochemical methods, immunostaining, and immunoelectron microscopy [54], [55], [56], [57]. In addition to the expected localization in intracellular membranes (such as the ER and Golgi) based on cell culture studies, PS1-drived fragments in neurons were also found in small synaptic vesicles, synaptic plasma membranes, synaptic adhesion

PS and cellular substrates of memory

The development of transgenic mouse models based on genes that are mutated in individuals with familial early-onset Alzheimer’s disease has greatly advanced our understanding of the pathogenesis of this debilitating disorder [65]. Despite the success of these efforts, little attention has been directed towards the understanding of basic physiology of memory storage in these animal models [66]. Few studies have investigated synaptic transmission and LTP, which contribute to several forms of

Regulation of Ca2+ homeostasis and apoptosis

Nearly a decade ago it was observed that inositol trisphosphate (IP3)-mediated intracellular Ca2+ release was enhanced in fibroblasts from patients with AD [74]. Neither voltage-dependent Ca2+ influx, nor release of endoplasmic reticulum Ca2+ stores by exposure to thapsigargin was different for AD and control fibroblasts. Although this study included fibroblasts derived from familial and nonfamilial cases of AD, subsequent studies demonstrated significant potentiation of IP3-evoked Ca2+ release

Conclusions

Inhibition of Aβ production by interfering with PS1/γ-secretase activity is considered as one of the potential therapeutic strategy for the treatment of Alzheimer’s disease, and several inhibitors that target to PS-derived fragments and lower Aβ production have been developed. However, the accumulating evidence that APP, Notch and other unidentified transmembrane proteins share similar intramembraneous proteolytic processing pathways compels us to reconsider the merit of this strategy. Based on

Acknowledgements

The authors are supported by grants from the National Institutes of Health, the Alzheimer’s Association and American Health Assistance Foundation.

References (105)

  • E.L. Meyer et al.

    Glutamate receptor subunit 3 is modified by site-specific limited proteolysis including cleavage by γ-secretase

    J. Biol. Chem.

    (2003)
  • G. Van Gassen et al.

    Binding partners of Alzheimer’s disease proteins: are they physiologically relevant?

    Neurobiol. Dis.

    (2000)
  • B.J. Passer et al.

    Interaction of Alzheimer’s presenilin-1 and presenilin-2 with Bcl-(XL). A potential role in modulating the threshold of cell death

    J. Biol. Chem.

    (1999)
  • D.E. Kang et al.

    Presenilin couples the paired phosphorylation of β-catenin independent of axin: implications for β-catenin activation in tumorigenesis

    Cell

    (2002)
  • E. Noll et al.

    Presenilin affects arm/β-catenin localization and function in Drosophila

    Dev. Biol.

    (2000)
  • S.M. Janicki et al.

    Presenilin overexpression arrests cells in the G1 phase of the cell cycle. Arrest potentiated by the Alzheimer’s disease PS2(N141I)mutant

    Am. J. Pathol.

    (1999)
  • J.E. Meredith et al.

    γ-Secretase activity is not involved in presenilin-mediated regulation of β-catenin

    Biochem. Biophys. Res. Commun.

    (2002)
  • C.A. Saura et al.

    The non-conserved hydrophilic loop domain of presenilin (PS) is neither required for PS endoproteolysis nor enhanced Aβ42 production mediated by familial Alzheimer’s disease-linked PS variants

    J. Biol. Chem.

    (2000)
  • O. Berezovska et al.

    Notch1 inhibits neurite outgrowth in post-mitotic primary neurons

    Neuroscience

    (1999)
  • O. Berezovska et al.

    The Alzheimer-related gene presenilin 1 facilitates notch 1 in primary mammalian neurons

    Brain Res. Mol. Brain Res.

    (1999)
  • J.L. Franklin et al.

    Autonomous and non-autonomous regulation of mammalian neurite development by Notch1 and Delta1

    Curr. Biol.

    (1999)
  • D.J. Figueroa et al.

    Presenilin-dependent γ-secretase activity modulates neurite outgrowth

    Neurobiol. Dis.

    (2002)
  • A. Georgakopoulos et al.

    Presenilin-1 forms complexes with the cadherin/catenin cell-cell adhesion system and is recruited to intercellular and synaptic contacts

    Mol. Cell

    (1999)
  • Y. Goda

    Cadherins communicate structural plasticity of presynaptic and post-synaptic terminals

    Neuron

    (2002)
  • H. Togashi et al.

    Cadherin regulates dendritic spine morphogenesis

    Neuron

    (2002)
  • S. Murase et al.

    Depolarization drives β-catenin into neuronal spines promoting changes in synaptic structure and function

    Neuron

    (2002)
  • D.Y. Kim et al.

    Nectin-1α, an immunoglobulin-like receptor involved in the formation of synapses, is a substrate for presenilin/γ-secretase-like cleavage

    J. Biol. Chem.

    (2002)
  • W.G. Annaert et al.

    Interaction with telencephalin and the amyloid precursor protein predicts a ring structure for presenilins

    Neuron

    (2001)
  • S. Lammich et al.

    Presenilin-dependent intramembrane proteolysis of CD44 leads to the liberation of its intracellular domain and the secretion of an Aβ-like peptide

    J. Biol. Chem.

    (2002)
  • G.R. Seabrook et al.

    Transgenic animals relevant to Alzheimer’s disease

    Neuropharmacology

    (1999)
  • A. Parent et al.

    Synaptic transmission and hippocampal long-term potentiation in transgenic mice expressing FAD-linked presenilin 1

    Neurobiol. Dis.

    (1999)
  • P.A. Barrow et al.

    Functional phenotype in transgenic mice expressing mutant human presenilin-1a

    Neurobiol. Dis.

    (2000)
  • S.H. Zaman et al.

    Enhanced synaptic potentiation in transgenic mice expressing presenilin 1 familial Alzheimer’s disease mutation is normalized with a benzodiazepine

    Neurobiol. Dis.

    (2000)
  • I. Schneider et al.

    Mutant presenilins disturb neuronal calcium homeostasis in the brain of transgenic mice, decreasing the threshold for excitotoxicity and facilitating long-term potentiation

    J. Biol. Chem.

    (2001)
  • R. Pybus et al.

    Enhanced long-term potentiation in the hippocampus of rats expressing mutant presenillin-1 is age related

    Neurobiol. Dis.

    (2003)
  • R.A. Morton et al.

    Impairment in hippocampal long-term potentiation in mice under-expressing the Alzheimer’s disease related gene presenilin-1

    Neurosci. Lett.

    (2002)
  • M.A. Leissring et al.

    Presenilin-2 mutations modulate amplitude and kinetics of inositol 1,4,5-trisphosphate-mediated calcium signals

    J. Biol. Chem.

    (1999)
  • M.A. Leissring et al.

    Subcellular mechanisms of presenilin-mediated enhancement of calcium signaling

    Neurobiol. Dis.

    (2001)
  • A. Cedazo-Minguez et al.

    The presenilin 1 DeltaE9 mutation gives enhanced basal phospholipase C activity and a resultant increase in intracellular calcium concentrations

    J. Biol. Chem.

    (2002)
  • A.S. Yoo et al.

    Presenilin-mediated modulation of capacitative calcium entry

    Neuron

    (2000)
  • I.F. Smith et al.

    Ca(2+) stores and capacitative Ca(2+) entry in human neuroblastoma (SH-SY5Y) cells expressing a familial Alzheimer’s disease presenilin-1 mutation

    Brain Res.

    (2002)
  • J. Herms et al.

    Capacitive calcium entry is directly attenuated by mutant presenilin-1, independent of the expression of the amyloid precursor protein

    J. Biol. Chem.

    (2003)
  • P. Vito et al.

    Requirement of the familial Alzheimer’s disease gene PS2 for apoptosis. Opposing effect of ALG-3

    J. Biol. Chem.

    (1996)
  • P. Vito et al.

    Generation of anti-apoptotic presenilin-2 polypeptides by alternative transcription, proteolysis, and caspase-3 cleavage

    J. Biol. Chem.

    (1997)
  • S.L. Chan et al.

    Presenilin-1 mutations sensitize neurons to DNA damage-induced death by a mechanism involving perturbed calcium homeostasis and activation of calpains and caspase-12

    Neurobiol. Dis.

    (2002)
  • N. Takasugi et al.

    The role of presenilin cofactors in the γ-secretase complex

    Nature

    (2003)
  • W.T. Kimberly et al.

    γ-Secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • Kim SH, Ikeuchi T, Yu C, Sisodia SS. Regulated hyperaccumulation of presenilin-1 and the “γ-secretase” complex:...
  • Y.M. Li et al.

    Photoactivated γ-secretase inhibitors directed to the active site covalently label presenilin 1

    Nature

    (2000)
  • W.P. Esler et al.

    Transition-state analogue inhibitors of γ-secretase bind directly to presenilin-1

    Nat. Cell Biol.

    (2000)
  • Cited by (60)

    • Trafficking in neurons: Searching for new targets for Alzheimer's disease future therapies

      2013, European Journal of Pharmacology
      Citation Excerpt :

      Moreover, the substrates APP and Notch were shown to compete for proteolysis but not for binding to PS under saturating condition (Schroeter et al., 2003). Several studies demonstrate that certain PS domains have different functions independently of their role in the γ-secretase complex (Thinakaran and Parent, 2004). For example, the hydrophilic loop of PS1 represents the domain of interaction with β-catenin, and this association functions as a negative regulator of the Wnt/β-catenin signaling pathway (Kang et al., 2002; Soriano et al., 2001).

    • Mouse models of Alzheimer's disease

      2012, Brain Research Bulletin
    • Intraperitoneal injection of JNK-specific inhibitor SP600125 inhibits the expression of presenilin-1 and Notch signaling in mouse brain without induction of apoptosis

      2012, Brain Research
      Citation Excerpt :

      PS1, PS2, and γ-secretase also cleave a variety of other type 1 transmembrane proteins which all release intracellular fragments (ICD) with the ability to interact with transcription co-activators (Koo and Kopan, 2004; Kopan and Goate, 2000). Hence PS1 and PS2 may affect the expression of many genes through intramembrane proteolysis (Thinakaran and Parent, 2004). Therefore, we have studied the transcriptional control of the PS1 gene.

    • Aberrant regulation of alternative pre-mRNA splicing in schizophrenia

      2010, Neurochemistry International
      Citation Excerpt :

      The presenilins (PSs) have been identified as causative genes for familial Alzheimer's disease (AD); they are required for the regulation of the intramembrane proteolysis of amyloid precursor protein, another causative gene for familial AD. In addition, PSs have an important role in Notch, ERBB4, E-cadherin and WNT signaling and are also associated with schizophrenia (Ide et al., 2004; Law et al., 2007; Lee et al., 2002; Marambaud et al., 2002; Roperch et al., 1998; Sardi et al., 2006; Soriano et al., 2001; Takashima et al., 1998; Thinakaran and Parent, 2004; Wei and Hemmings, 2000). Furthermore, the PS2 gene has been genetically associated with SZ (Zhang et al., 2009).

    View all citing articles on Scopus
    View full text