You are on page 1of 4

1

A Brief Introduction to the History


of the β-Amyloid Protein (Aβ) of
Alzheimer’s Disease
David H. Small and Colin J. Barrow

Alzheimer’s disease (AD) is the most common (Αβ) is a key step in the pathogenic mechanism of
cause of dementia in the elderly. Typically, the dis- Alzheimer’s disease. In contrast, although the den-
ease progresses in a prolonged, inexorable manner sity of neurofibrillary tangles correlates more
[1]. Patients initially show symptoms of mild cog- closely with the cognitive symptoms, it is now
nitive impairment, which may include some mem- commonly thought that tangles are a secondary
ory loss. As the disease progresses, more severe feature or the underlying disease process [6].
memory loss occurs (e.g., retrograde amnesia)
leading to confusion and lack of orientation. The
patient is often institutionalized in this period, as it 1.1 The Role of Aβ in AD
becomes increasingly difficult for family members
to cope with the constant requirements of care. In Glenner and Wong [7] first identified the major
later stages of the disease, apathy and stupor can protein component of vascular amyloid, which was
occur, and the patient becomes bedridden. a low-molecular-weight, 4-kDa polypeptide, now
The histopathology of AD is characterized by referred to as the β-amyloid protein (Αβ). Subse-
gliosis and tissue atrophy caused by both synaptic quent studies established that the same protein was
and neuronal loss, which are most pronounced in the major component of amyloid plaques [8]. The
the frontal and temporal cortices [2]. Proteinaceous complete amino acid sequence of Αβ led to the
deposits are seen in both the intracellular and extra- identification of its precursor, the β-amyloid pre-
cellular compartments of the brain, typically in the cursor protein (APP) [9].
hippocampus and neocortex. The intracellular APP has features of an integral type I transmem-
deposits consist of neurofibrillary tangles that are brane glycoprotein, with a large ectodomain con-
made up of paired helical filaments of a hyper- taining the N-terminus and a small cytoplasmic
phosphorylated form of the cytoskeletal protein tau domain containing the C-terminus (Fig. 1.1).
[3]. Extracellular amyloid plaques are found most Multiple mRNA splicing of exons can generate sev-
commonly in the hippocampus and neocortex and eral different isoforms of APP that lack domains
may be diffuse or compact in nature [4]. Amyloid homologous to Kunitz-type protease inhibitors
is also deposited as cerebral amyloid angiopathy (KPI domain) and the OX-2 antigen as well as a
within small- to medium-sized arterioles [5]. domain encoded by an exon that regulates O-linked
Although neurofibrillary tangles are associated glycosylation by chondroitin sulfate. The Αβ
with a number of different types of neurodegenera- sequence itself comprises part of the ectodomain of
tive disease, the presence of numerous compact or the protein and extends into, but not all the way
neuritic amyloid plaques is a hallmark feature of through, the transmembrane domain [9, 10].
Alzheimer’s disease. For this reason, it may be Soon after its identification, APP was shown
argued that accumulation of the β-amyloid protein to undergo ectodomain shedding by an enzyme

1
2 D.H. Small and C.J. Barrow

associated with aggregation of the peptide [22].


N c APP Recent studies support the view that the most toxic
species are the low-molecular-weight, soluble
β-cleavage α-cleavage oligomers of Αβ [23].
C99 C83
Despite many studies that have shown that Αβ
γ-cleavage γ-cleavage
can disrupt biochemical events within neurons,
direct proof that the accumulation of Αβ is the
Aβ p3
cause of AD has been lacking. Nevertheless, evi-
aggregation
dence that this is the case has slowly been accumu-
Neurotoxicity
lating. Some of the strongest evidence that Αβ
AD
accumulation is the cause, rather than an epiphe-
Aβ oligomer
nomenon, of AD has come from the finding of
FIGURE 1.1. Proteolytic processing of APP and the role of familial AD mutations present in the APP gene
Aβ in AD. APP can be proteolytically cleaved via two [24]. All of these mutations have been found to
different processing pathways. Cleavage by α-secretase cluster around the Αβ sequence, and all of them
and γ-secretase generates C-terminal fragments known have so far been shown to directly or indirectly
as C83 and p3, whereas cleavage by β-secretase cause an increase in forms of Αβ that aggregate
(BACE1) and γ-secretase generates C99 and Aβ. [25]. For example, although the most commonly
According to the amyloid hypothesis, Aβ aggregates
produced form of Αβ contains 40-amino-acid
into amyloid fibrils or low-molecular-weight oligomers
residues (Αβ40), a minor form containing 42 residues
that are neurotoxic. The resulting neurotoxicity causes
neurodegeneration and leads to dementia. is also formed. This minor form aggregates into
amyloid fibrils much more readily than Αβ40 [26].
The first mutation to be identified in the APP gene,
the London mutation, involves a single base change
at codon 717, which encodes a form of APP that is
dubbed the α-secretase. The α-secretase cleaves more readily cleaved to produce Αβ42. To date, at
APP within the Αβ sequence, adjacent to lysine-16, least 10 familial AD mutations are known to occur
thereby destroying the sequence [11, 12]. Recently in APP [27].
studies suggest that enzymes of the ADAM family The direct involvement of APP and Αβ in the
of metalloproteases are responsible for this activity pathogenesis of AD is also strongly supported by
[13, 14]. Other studies have demonstrated that studies on transgenic mice. A number of transgenic
APP can also be cleaved at the N- and C-terminal lines have been developed in which human APP is
ends of the Αβ sequence by enzymes dubbed β- expressed [28]. Many of these mice develop amy-
and γ-secretase, respectively, to generate the full- loid plaques. In addition, other features of AD
length Αβ sequence [15]. Amyloidogenic process- pathology such as neuritic dystrophy, abnormal tau
ing by β- and γ-secretase is a normal, albeit minor, phosphorylation, gliosis, synaptic loss, and behav-
pathway of APP processing. The β-secretase has ioral abnormalities have been observed. Although
been unequivocally identified as an aspartyl pro- human APP mice do not develop neurofibrillary
tease termed BACE1 (an acronym for β-site APP tangles, this is probably due to differences between
cleaving enzyme-1) [16–19]. The γ-secretase com- mouse tau and human tau isoforms. Indeed, in
prises a complex of several proteins including double transgenic mice expressing both mutant
presenilin-1, presenilin-2, Aph1, Pen2, and nicas- human tau and APP, Αβ is seen to increase tau
trin. However, other protein components of this deposition [29].
complex may also exist [15]. Mutations in the APP gene account for only a
There is considerable evidence that the accumu- very small percentage of all familial Alzheimer’s
lation of Αβ in the brain is toxic to neurons and that disease (FAD) cases. Shortly after the identifica-
this toxicity underlies the neurodegeneration that tion of the first familial AD mutation in the APP
occurs in AD (Fig. 1.1) [20]. Aβ peptides are toxic gene, mutations were identified in two other genes,
to cells in culture [21], and this toxicity is PS1 encoding presenilin-1 and PS2 encoding
1. Brief Introduction to the History of the β-Amyloid Protein 3

presenilin-2, located on chromosomes 14 and 1, 1.3 Current Status of the Aβ


respectively [30, 31]. Both presenilin proteins are
components of the γ-secretase complex, and famil-
Hypothesis of AD
ial AD mutations within the PS1 and PS2 genes
There is now very strong evidence that accumula-
alter γ-secretase processing in a way that leads to
tion of oligomeric or fibrillar Αβ in the brain is a
the production of more Αβ42 [32].
key event in the pathogenesis of AD. Perhaps the
In general, mutations in the APP, PS1, and
most important unresolved question is the mecha-
PS2 genes lead to early-onset forms of AD. In
nism by which Αβ causes its neurotoxic effect. It is
contrast, the apolipoprotein E (apoE) gene
also unclear what form of aggregated Αβ is the
located on chromosome 19 is a risk factor for
most neurotoxic. Another major question is how
late-onset AD [33]. There are three forms of
many unidentified genetic risk factors there are and
apoE, termed E2, E3, and E4, encoded by three
how these risk factors affect Αβ production, aggre-
allelic variants ε2, ε3, and ε4. The ε4 variant is a
gation, or clearance. If anti-Αβ therapies can be
risk factor for late-onset AD, whereas the ε2 may
used successfully for the treatment of AD, then the
be protective. Although the reason for this is still
remaining concerns about the role of Αβ in the
unknown, it is undoubtedly related to Aβ pro-
pathogenesis of AD will have been answered.
duction, aggregation, or clearance from the
brain. Individuals with the ε4 allele have more
Aβ deposition within the brain [34]. In addition, References
APP x apoE knockout transgenic mice develop
1. Storey E, Kinsella GJ, Slavin MJ. The neuropsycho-
little amyloid deposition in their brains, unlike
logical diagnosis of Alzheimer’s disease. J Alzheimers
normal APP mice [35]. Thus, studies on the role Dis 2001; 3:261-285.
of apoE in AD provide strong support for the Aβ 2. Probst A, Langui D, Ulrich J. Alzheimer’s disease: a
hypothesis. description of the structural lesions. Brain Pathol
1991; 1:229-239.
3. Iqbal K, Alonso Adel C, Chen S, et al. Tau pathology
1.2 Anti-Αβ Therapies for AD in Alzheimer’s disease and other tauopathies.
Biochim Biophys Acta 2005; 1739:198-210.
The idea that Aβ is a primary causative agent in 4. Wisniewski HM, Wegiel J, Kotula L. Review. David
AD leads inevitably to the view that an effective Oppenheimer Memorial Lecture 1995: Some neu-
therapy based on inhibiting the production, aggre- ropathological aspects of Alzheimer’s disease and its
relevance to other disciplines. Neuropathol Appl
gation, clearance, or toxicity of Aβ may be achiev-
Neurobiol 1996; 22:3-11.
able. One of the most promising but controversial 5. Castellani RJ, Smith MA, Perry G, Friedland RP.
approaches in recent years has been Αβ immuniza- Cerebral amyloid angiopathy: major contributor or
tion. Studies show that in transgenic mice, immu- decorative response to Alzheimer’s disease patho-
nization with Αβ42 leads to the generation of genesis. Neurobiol Aging 2004; 25:599-602.
an immune response [36]. Anti-amyloid antibod- 6. Small DH, McLean CA. Alzheimer’s disease and the
ies bind to amyloid plaques and appear to facilitate amyloid beta protein: What is the role of amyloid?
their removal from the brain, leading to an J Neurochem 1999; 73:443-449.
improvement in cognitive performance compared 7. Glenner GG, Wong CW. Alzheimer’s disease: initial
with nonimmunized control animals. Unfortu- report of the purification and characterization of a
nately, clinical trials of this approach in humans novel cerebrovascular amyloid protein. Biochem
Biophys Res Commun 1984; 120:885-890.
have been halted because a small percentage of
8. Masters CL, Simms G, Weinman NA, et al. Amyloid
individuals immunized with Αβ have developed a plaque core protein in Alzheimer’s disease and Down
severe meningoencephalitis [37]. Nevertheless, syndrome. Proc Natl Acad Sci U S A 1985; 82:4245-
there is some evidence that patients who develop a 4249.
strong immune response to Αβ without the associ- 9. Kang J, Lemaire HG, Unterbeck A, et al. The precur-
ated brain inflammation may benefit from this sor of Alzheimer’s disease amyloid A4 protein resem-
approach [38]. bles a cell-surface receptor. Nature 1987; 325:733-736.
4 D.H. Small and C.J. Barrow

10. Wilquet V, De Strooper B. Amyloid-beta precursor 24. Hardy JA, Higgins GA. Alzheimer’s disease: the
protein processing in neurodegeneration. Curr Opin amyloid cascade hypothesis. Science 1992; 256:184-
Neurobiol 2004; 14:582-588. 185.
11. Weidemann A, Konig G, Bunke D, et al. 25. Scheuner D, Eckman C, Jensen M, et al. Secreted
Identification, biogenesis, and localization of precur- amyloid beta-protein similar to that in the senile
sors of Alzheimer’s disease A4 amyloid protein. Cell plaques of Alzheimer’s disease is increased in vivo
1989; 57:115-126. by the presenilin 1 and 2 and APP mutations linked
12. Esch FS, Keim PS, Beattie EC, et al. Cleavage of to familial Alzheimer’s disease. Nat Med 1996;
amyloid beta peptide during constitutive processing 2:864-870.
of its precursor. Science 1990; 248:1122-1124. 26. Jarrett JT, Lansbury PT, Jr. Seeding “one-dimen-
13. Buxbaum JD, Liu KN, Luo Y, et al. Evidence that sional crystallization” of amyloid: a pathogenic
tumor necrosis factor alpha converting enzyme is mechanism in Alzheimer’s disease and scrapie? Cell
involved in regulated alpha-secretase cleavage of the 1993; 73:1055-1058.
Alzheimer amyloid protein precursor. J Biol Chem 27. Bertram L, Tanzi RE. The current status of
1998; 273:27765-27767. Alzheimer’s disease genetics: what do we tell the
14. Lammich S, Kojro E, Postina R, et al. Constitutive and patients? Pharmacol Res 2004; 50:385-396.
regulated alpha-secretase cleavage of Alzheimer’s 28. Hock BJ Jr., Lamb BT. Transgenic mouse models of
amyloid precursor protein by a disintegrin metal- Alzheimer’s disease. Trends Genet 2001; 17:S7-12.
loprotease. Proc Natl Acad Sci U S A 1999; 96: 29. Lewis J, Dickson DW, Lin WL, et al. Enhanced
3922-3927. neurofibrillary degeneration in transgenic mice
15. Nunan J, Small DH. Regulation of APP cleavage by expressing mutant tau and APP. Science 2001;
alpha-, beta- and gamma-secretases. FEBS Lett 293:1487-1491.
2000; 483:6-10. 30. Sherrington R, Rogaev EI, Liang Y, et al. Cloning of
16. Vassar R, Bennett BD, Babu-Khan S, et al. Beta-sec- a gene bearing missense mutations in early-onset
retase cleavage of Alzheimer’s amyloid precursor familial Alzheimer’s disease. Nature 1995; 375:754-
protein by the transmembrane aspartic protease 760.
BACE. Science 1999; 286:735-741. 31. Levy-Lahad E, Wasco W, Poorkaj P, et al. Candidate
17. Lin X, Koelsch G, Wu S, et al. Human aspartic pro- gene for the chromosome 1 familial Alzheimer’s dis-
tease memapsin 2 cleaves the beta-secretase site of ease locus. Science 1995; 269:973-977.
beta-amyloid precursor protein. Proc Natl Acad Sci 32. Saunders AM, Strittmatter WJ, Schmechel D, et al.
U S A 2000; 97:1456-1460. Association of apolipoprotein E allele epsilon 4 with
18. Sinha S, Anderson JP, Barbour R, et al. Purification late-onset familial and sporadic Alzheimer’s disease.
and cloning of amyloid precursor protein beta-secre- Neurology 1993; 43:1467-1472.
tase from human brain. Nature 1999; 402:537-540. 33. Schmechel DE, Saunders AM, Strittmatter WJ, et al.
19. Yan R, Bienkowski MJ, Shuck ME, et al. Membrane- Increased amyloid beta-peptide deposition in cere-
anchored aspartyl protease with Alzheimer’s disease bral cortex as a consequence of apolipoprotein E
beta-secretase activity. Nature 1999; 402:533-537. genotype in late-onset Alzheimer’s disease. Proc
20. Small DH, Mok SS, Bornstein JC. Alzheimer’s dis- Natl Acad Sci U S A 1993; 90:9649-9653.
ease and Abeta toxicity: from top to bottom. Nat Rev 34. Bales KR, Verina T, Dodel RC, et al. Lack of
Neurosci 2001; 2:595-598. apolipoprotein E dramatically reduces amyloid beta-
21. Yankner BA, Dawes LR, Fisher S, et al. Neurotox- peptide deposition. Nat Genet 1997; 17:263-264.
icity of a fragment of the amyloid precursor associ- 35. Schenk D, Barbour R, Dunn W, et al. Immunization
ated with Alzheimer’s disease. Science 1989; with amyloid-beta attenuates Alzheimer’s-disease-
245:417-420. like pathology in the PDAPP mouse. Nature 1999;
22. Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, 400:173-177.
et al. Structure-activity analyses of beta-amyloid 36. Nicoll JA, Wilkinson D, Holmes C, et al.
peptides: contributions of the beta 25-35 region to Neuropathology of human Alzheimer’s disease after
aggregation and neurotoxicity. J Neurochem 1995; immunization with amyloid-beta peptide: a case
64:253-265. report. Nat Med 2003; 9:448-452.
23. Walsh DM, Selkoe DJ. Deciphering the molecular 37. Hock C, Konietzko U, Streffer JR, et al. Antibodies
basis of memory failure in Alzheimer’s disease. against beta-amyloid slow cognitive decline in
Neuron 2004; 44:181-193. Alzheimer’s disease. Neuron 2003; 38:547-554.

You might also like