γ-Secretase Inhibitors – from Molecular Probes to New Therapeutics?

https://doi.org/10.1016/S0079-6468(02)41003-XGet rights and content

Publisher Summary

The hallmark lesions in Alzheimer's disease (AD) brains described by Alois Alzheimer are extracellular proteinaceous deposits, found either as amyloid plaques in the brain parenchyma or as vascular amyloid surrounding the brain blood vessels, and intracellular neurofibrillary tangles consisting of an abnormally phosphorylated microtubule-associated protein tau. Over the past several years, extraordinary progress has been made in understanding mechanistic details of the amyloid hypothesis of AD. A part of this understanding has arisen as a result of the availability of molecular probes targeted toward γ-secretase. Although the final proof of the identity of γ-secretase by reconstitution of the γ-secretase complex in vitro is a difficult task, substantial evidence generated by affinity labeling of presenilins by transition state analog inhibitors points toward a critical role of polypeptides in the enzyme complex.

Introduction

In 1906, the neuropathologist Alois Alzheimer drew the attention of the scientific community to neuropathological changes that he had observed in the brains of elderly patients and suggested a possible connection to the neuronal dysfunction underlying the disease [1], which was later to bear his name. Subsequent progress in the understanding of the molecular basis of Alzheimer's disease (AD) proceeded at a relatively slow rate. During the last decade, however, new genetic and biochemical evidence has provided a rationale for a potential disease-modifying therapeutic approach. Since both age and genetic predisposition are the major known risk factors for AD, the increased life span of the population of the Western world in recent decades has resulted in an urgent need for an effective therapy. One of the main aims will be to prevent or slow down the rapid decline in cognitive function and memory, which is a consequence of this irreversible and progressive neurodegenerative disease affecting the central nervous system (CNS).

The hallmark lesions in AD brains described by Alois Alzheimer are extracellular proteinaceous deposits found either as amyloid plaques in the brain parenchyma or as vascular amyloid surrounding the brain blood vessels, and intracellular neurofibrillary tangles consisting of an abnormally phosphorylated microtubule-associated protein tau [2]. Until 1984, the identity of the main component of the extracellular deposits was unknown, but amino acid sequencing of the major peptide entity purified from vascular amyloid [3] or senile plaques [4] revealed that they are composed primarily of the 40–42 amino acid amyloid-β (Aβ) peptide. The Aβ peptide is a product of post-translational processing, which is derived by sequential proteolytic cleavage of a type I transmembrane protein, the β-amyloid precursor protein (βAPP) [5], by enzymes referred to as β- and γ-secretase [6] (Figure 3.1). β-Secretase cleaves within the βAPP ectodomain close to the extracellular membrane surface to generate a membrane-bound intermediate, the β-C-terminal fragment (β-CTF, C99). A predominant alternative processing pathway, which precludes Aβ peptide formation, involves cleavage of βAPP within the Aβ sequence by a protease termed α-secretase, producing an alternative membrane-bound stub, the α-C-terminal fragment (α-CTF, C83). This activity appears to be mediated by members of the disintegrin and metalloprotease family TACE [7] and ADAM-10 [8] and leads to the secretion of the soluble βAPP ectodomain as secretory βAPP [9]. Both processing events (β- and α-secretase cleavage) generate truncated membrane-bound substrates for a third protease, γ-secretase. These substrates are cleaved within the lipid bilayer by γ-secretase to release either Aβ peptides as a product of sequential β/γ-cleavage or a peptide termed p3 as a result of α/γ-cleavage [6].

A causative role for the Aβ peptide, and especially the more hydrophobic C-terminally elongated variant Aβ(1–42), in AD in which accumulation in extracellular protein deposits occurs, has been postulated (the so-called amyloid cascade hypothesis) [10]. This hypothesis is substantially supported by the identification of autosomal dominant mutations causing familial Alzheimer's disease (FAD) which is an inherited form of AD leading to an early onset of the disease. Such FAD mutations were found either in the βAPP gene itself, clustering around β- and γ-secretase cleavage sites of the corresponding Aβ peptide sequence (Figure 3.2) or in two alternative genes encoding presenilin 1 and 2 (PS1 and PS2) (Figure 3.3), two proteins which were shown to have high homology with each other. Mechanistically, all these mutations increase the production of Aβ(1–42) peptide, which is more hydrophobic, more prone to aggregation and is deposited earlier in the time course of the disease than the shorter (and more predominant) species Aβ(1–40) (for review see 10, 11). Although the prevalence of FAD is much lower than of late onset sporadic AD, the pathology observed in both types is similar, and it seems likely that the disease mechanisms involved may be the same. It is noteworthy that the occurrence of familial and sporadic forms of several neurodegenerative diseases has been observed, including Parkinsonism and amyotrophic lateral sclerosis (ALS) [12]. If one considers a causative role for Aβ peptide in the manifestation of AD, inhibition of either of the two critical enzymes involved in Aβ peptide generation provides target opportunities to develop drugs which could slow down or even halt the progression of the disease.

Section snippets

β-Secretase

β-Secretase (β-site APP cleaving enzyme, BACE1) has been cloned 13, 14, 15, 16, 17 and was shown to be a type I transmembrane protein with homology to the pepsin family of aspartyl proteases. BACE1 contains two catalytic sites characterised by the D(T/S)G(T/S) motif within its large luminal domain. Like other aspartyl proteases, BACE1 has a propeptide (Pro) domain which is removed by constitutive N-terminal processing in the Golgi apparatus 18, 19 to generate mature BACE. This endoproteolytic

γ-Secretase and presenilins

Whereas BACE has been characterised intensively using either overexpressing cell lines [18] or purified recombinant enzyme 21, 28, the critical enzyme which releases the Aβ peptide from the membrane-substrate, γ-secretase, has proven more elusive. Genetic evidence has linked γ-secretase to presenilins 1 and 2 (PS1 and PS2) which were identified in 1995 by mutations causing FAD 32, 33, 34, 35. Intriguingly, mutations in the PS1 gene appear to account for the majority of all known FAD cases to

γ-Secretase inhibitors

The first reported γ-secretase inhibitors were peptidyl aldehydes, which had originally been developed as calpain inhibitors 59, 60. These compounds, for example, compound (1), were weak inhibitors of Aβ production in vitro (IC50's 5–200 μM). Since peptidyl aldehydes have been shown to inhibit a variety of proteases including cysteine, serine and aspartyl, the identification of these inhibitors did not allow assignment of γ-secretase to a distinct mechanistic class of protease. In order to

Probing the nature of γ-secretase: active site-directed photoaffinity probes

The identification of potent, selective inhibitors of γ-secretase has provided an opportunity to design tools that will allow further characterisation of this elusive protease. At Merck, compound (5) was used as a starting point to design and synthesise photoaffinity probes, which would be targeted to the active site of γ-secretase. Since compound (5) is a minor diastereoisomer of a compound that originated from the HIV protease inhibitor programme, a new synthetic route had to be developed to

γ-Secretase and regulated intramembrane proteolysis

The cleavage of βAPP by γ-secretase within its transmembrane domain would appear to be an extraordinary processing event. Within the last few years, however, significant evidence has accumulated which suggests that similar cleavage events are observed in a variety of biological systems. These events have been classified as regulated intramembrane proteolysis (RIP) [87], a signalling paradigm involving transmembrane domain-containing proteins which undergo proteolysis within their transmembrane

Conclusions and prospective

Over the last several years, extraordinary progress has been made in understanding mechanistic details of the amyloid hypothesis of AD. Part of this understanding has arisen as a result of the availability of molecular probes targeted toward γ-secretase. Although the final proof of the identity of γ-secretase by reconstitution of the γ-secretase complex in vitro will be a difficult task, substantial evidence generated by affinity labelling of presenilins by transition state analogue inhibitors

Acknowledgements

The authors would like to thank Drs Karl Gibson, Mark Shearman and Ian Churcher for proof-reading this manuscript, and for their helpful comments.

References (131)

  • M. Goedert et al.

    Curr. Opin. Neurobiol.

    (1991)
  • G.G. Glenner et al.

    Biochem. Biophys. Res. Commun.

    (1984)
  • J.D. Buxbaum et al.

    J. Biol. Chem.

    (1998)
  • A. Weidemann et al.

    Cell

    (1989)
  • J. Hardy

    Trends Neurosci.

    (1997)
  • I. Hussain et al.

    Mol. Cell. Neurosci.

    (1999)
  • M. Haniu et al.

    J. Biol. Chem.

    (2000)
  • A. Capell et al.

    J. Biol. Chem.

    (2000)
  • B.D. Bennett et al.

    J. Biol. Chem.

    (2000)
  • X.P. Shi et al.

    J. Biol. Chem.

    (2001)
  • I. Hussain et al.

    Mol. Cell. Neurosci.

    (2000)
  • F. Gruninger-Leitch et al.

    J. Biol. Chem.

    (2002)
  • G. Thinakaran et al.

    Neuron

    (1996)
  • A. Doan et al.

    Neuron

    (1996)
  • S.H. Kim et al.

    J. Biol. Chem.

    (2001)
  • G. Thinakaran et al.

    J. Biol. Chem.

    (1997)
  • H. Steiner et al.

    J. Biol. Chem.

    (1998)
  • T. Ratovitski et al.

    J. Biol. Chem.

    (1997)
  • A. Capell et al.

    J. Biol. Chem.

    (1998)
  • G. Yu et al.

    J. Biol. Chem.

    (1998)
  • Y. Hu et al.

    Dev. Cell

    (2002)
  • H. Lopez-Schier et al.

    Dev. Cell

    (2002)
  • J. Higaki et al.

    Neuron

    (1995)
  • H. Klafki et al.

    J. Biol. Chem.

    (1996)
  • T. Dyrks et al.

    FEBS Lett.

    (1993)
  • D. Seiffert et al.

    J. Biol. Chem.

    (2000)
  • A. Nadin et al.

    Tetrahedron

    (2001)
  • D. Beher et al.

    J. Biol. Chem.

    (2001)
  • M.S. Brown et al.

    Cell

    (2000)
  • J.R. Lee et al.

    Cell

    (2001)
  • S. Urban et al.

    Cell

    (2001)
  • A. Weihofen et al.

    J. Biol. Chem.

    (2000)
  • A. Alzheimer et al.

    Psychiatrie Psychisch.-Gerichtl. Med.

    (1907)
  • C.L. Masters et al.

    Proc. Natl Acad. Sci. USA

    (1985)
  • J. Kang et al.

    Nature (London)

    (1987)
  • C. Haass et al.

    Nature (London)

    (1992)
  • S. Lammich et al.

    Proc. Natl Acad. Sci. USA

    (1999)
  • J. Hardy

    Proc. Natl Acad. Sci. USA

    (1997)
  • D.L. Price et al.

    Science (Washington, DC)

    (1998)
  • R. Vassar et al.

    Science (Washington, DC)

    (1999)
  • X. Lin et al.

    Proc. Natl Acad. Sci. USA

    (2000)
  • S. Sinha et al.

    Nature (London)

    (1999)
  • R. Yan et al.

    Nature (London)

    (1999)
  • H. Cai et al.

    Nat. Neurosci.

    (2001)
  • A.J. Saunders et al.

    Science (Washington, DC)

    (1998)
  • M. Farzan et al.

    Proc. Natl Acad. Sci. USA

    (2000)
  • Y. Luo et al.

    Nat. Neurosci.

    (2001)
  • S. Kitazume et al.

    Proc. Natl Acad. Sci. USA

    (2001)
  • M. Mullan et al.

    Nat. Genet.

    (1992)
  • C. Haass et al.

    Nat. Med.

    (1995)
  • Cited by (10)

    • Selected non-steroidal anti-inflammatory drugs and their derivatives target γ-secretase at a novel site: Evidence for an allosteric mechanism

      2004, Journal of Biological Chemistry
      Citation Excerpt :

      Only certain classes of NSAIDs and derivatives thereof appear to be associated with this phenomenon (29, 39), and their potencies are modest. Furthermore, these compounds do not share any of the structural features that are found in potent γ-secretase inhibitors (reviewed by Harrison and Beher (47)), and not all NSAIDs modulate the enzyme. Thus far, it is only known that the activity for selective inhibition of Aβ(1–42) can be separated from cyclooxygenase inhibition, because inactive derivatives of NSAIDs such as R-flurbiprofen can still act as γ(42)-secretase inhibitors.

    • Links between amyloid and Tau biology in Alzheimer's disease and their cholinergic aspects

      2007, Exploring the Vertebrate Central Cholinergic Nervous System
    View all citing articles on Scopus
    View full text