Sei sulla pagina 1di 20

J Neural Transm (2006) 113: 1625–1644

DOI 10.1007/s00702-006-0579-2

Review
The significance of the cholinergic system in the brain
during aging and in Alzheimer’s disease

R. Schliebs, T. Arendt

Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany

Received: September 11, 2006 = Accepted: September 27, 2006 = published online October 13, 2006
# Springer-Verlag 2006

Summary Acetylcholine is widely distributed in the nervous system and thalamus and spinal cord. Moreover, cholinergic projection
has been implicated to play a critical role in cerebral cortical develop-
ment, cortical activity, controlling cerebral blood flow and sleep-wake
neurons comprise also motor neurons in the spinal cord
cycle as well as in modulating cognitive performances and learning and (cranial nerves 3–7, and 9–12) as well as cholinergic neu-
memory processes. Cholinergic neurons of the basal forebrain complex rons in the sympathic and parasympathic nervous system
have been described to undergo moderate degenerative changes during
(see e.g., Martinez-Murillo and Rodrigo, 1995).
aging, resulting in cholinergic hypofunction that has been related to the
progressing memory deficits with aging. Basal forebrain cholinergic cell The cholinergic neurons in the basal forebrain and in the
loss is also a consistent feature of Alzheimer’s disease, which has been pontine reticular formation are forming parts of the ascend-
suggested to cause, at least partly, the cognitive deficits observed, and ing reticular activating system. The basal forebrain cholin-
has led to the formulation of the cholinergic hypotheses of geriatric
memory dysfunction. Impaired cortical cholinergic neurotransmission ergic complex comprising medial septum, horizontal and
may also contribute to b-amyloid plaque pathology and increase phos- vertical diagonal band of Broca, and nucleus basalis of
phorylation of tau protein the main component of neurofibrillar tangles Meynert provides the mayor cholinergic projections to the
in Alzheimer’s disease. Understanding the molecular mechanisms under-
lying the interrelationship between cortical cholinergic dysfunction,
cerebral cortex and hippocampus, while the pontine cholin-
b-amyloid formation and deposition, and tau pathology in Alzheimer’s ergic system acts mainly through thalamic intralaminar
disease, would allow to derive potential treatment strategies to pharma- nuclei and provides only a minor innervation of the cortex.
cologically intervene in the disease-causing signaling cascade.
In particular, neurons located in the medial septum inner-
Keywords: Alzheimer, cholinergic system, beta-amyloid, tau, nerve vate predominantly the hippocampus, while those of the
growth factor
vertical and horizontal diagonal band project to the anterior
cingulate cortex and olfactory bulb, respectively. Choliner-
Brain cholinergic system, cognition gic neurons of the nucleus basalis of Meynert provide effer-
and dementing disorders ents to the amygdala and throughout the rest of the cortical
mantle (Bigl et al., 1990; Fig. 1). They display a continuum
Central cholinergic system of large cells that express nerve growth factor (NGF) and
its cognate receptors, calbindin, and glutamate receptors.
Central cholinergic neurons can be subdivided into projec- Intermingled with these cholinergic cells are smaller in-
tion neurons located mainly in the forebrain and upper brain terneurons containing NADPH-diaphorase, GABA, calcium
stem, and interneurons present in the caudate-putamen, binding proteins and several inhibitory neuropeptides in-
nucleus accumbens, hippocampus, cerebral cortex, hypo- cluding galanin (Mufson et al., 2003). The GABAergic neu-
rons within the basal forebrain system act as interneurons
Correspondence: Dr. Reinhard Schliebs, Department of Neurochemistry, that make connections with cholinergic neurons (Frotscher,
Paul Flechsig Institute for Brain Research, University of Leipzig, Jahnallee
59, 04109 Leipzig, Germany 1992) or may project to the hippocampus (Toth et al.,
e-mail: schre@medizin.uni-leipzig.de 1993).
1626 R. Schliebs, T. Arendt

produce dose-dependent deleterious effects on cognitive


performance of a broad spectrum of learned behaviour in
rats (Fibiger, 1991; Fr€olich et al., 1989) as well as in
humans (Christensen et al., 1992; Molchan et al., 1992),
providing evidence for the important role of the central
cholinergic system in realizing cognitive processes. Cholin-
ergic lesions by electrolytic or excitotoxic methods as well
as application of the cholinergic immunolesion strategy
(Wiley et al., 1991) that produces a more specific and
selective destruction of basal forebrain cholinergic cells
(Roßner et al., 1995; Heckers et al., 1994; reviewed in
Schliebs, 2005a), and stimulation of basal forebrain choli-
nergic cells provided experimental tools to further empha-
Fig. 1. Cholinergic innervation of the cortical mantle. The basal forebrain
size this view. Behavioral studies on such animal models
cholinergic complex comprising medial septum, horizontal and vertical
diagonal band of Broca, and nucleus basalis of Meynert provides the demonstrated that lesion-induced damage to basal forebrain
mayor cholinergic projections to the cerebral cortex and hippocampus. cholinergic system, and their cholinergic projections to the
Neurons located in the medial septum innervate predominantly the neocortex induced deficits related to cognitive impairments,
hippocampus, while those of the vertical and horizontal diagonal band
project to the anterior cingulate cortex and olfactory bulb (limbic cortex), especially on attentional processes (Sarter and Bruno, 1997),
respectively. Cholinergic neurons of the nucleus basalis of Meynert as well as on learning and memory processes (Dunnett and
provide efferents to the amygdala and throughout the rest of the cortical Fibiger, 1993; Everitt and Robbins, 1997; Wenk, 1993;
mantle. Individual cells in the nucleus basalis=diagonal band complex
have a restricted axonal projections to the cerebral cortex in the rat.
Woolf, 1997; for review, see also, McKinney, 2005).
Although the projection as a whole is very widespread, the projection of
individual cells is limited to a very restricted area within the cortex, not
more than 1–2 mm in diameter (modified according to Arendt, 1999) Cholinergic system in postnatal development and aging
A number of anatomical, electrophysiological, chemical,
Physiologically, the brain cholinergic system plays a role and behavioral studies have highlighted also a critical role
in controlling cerebral blood flow (Biesold et al., 1989; for acetylcholine in cortical development, and its impor-
Barbelivien et al., 1999; Sato et al., 2004), cortical activity tance in establishing synaptic contacts in networks of cells
(Detari et al., 1999; Lucas-Meunier et al., 2003), and sleep- that will subserve complex cognitive functions in adult-
wake cycle (Lee et al., 2005) as well as in modulating hood (see e.g., Berger-Sweeney, 2003). There is ample evi-
cognitive function and cortical plasticity (Arendt and Bigl, dence that acetylcholine plays a role in structural and
1986; Bigl and Schliebs, 1998; Schliebs and Bigl, 1996; functional remodeling of cortical circuits. In particular,
Schliebs et al., 1996, for review, see e.g. McKinney, 2005). the involvement of acetylcholine in visual cortex plasticity
has extensively been investigated over the years (Roßner
et al., 1994; Schliebs et al., 1982; Walch et al., 1989; for
Central cholinergic function in learning and memory
review, see e.g., Gu, 2003).
Already for a long time it is known that agents such as The occurance of abnormalities in the control of cortical
tropan alkaloids that are today known to block muscarinic cholinergic input very early in life such as disruption of
cholinergic receptor sites, result in memory deficits and trophic factor support (Mufson et al., 2002; Ginsberg et al.,
cognitive disturbancies. The involvement of central cholin- 2006b), has been assumed to mediate early-life cognitive
ergic function in learning and memory was first postulated limitation, a disease stage that is suggested to subsequent-
by Deutsch (1971), and later further corroborated by phar- ly develop into mild cognitive impairment (MCI; Sarter
macological studies (Drachman and Leavitt, 1974) demon- and Bruno, 2004) and Alzheimer disease. Enhancement of
strating that centrally acting anticholinergic drugs impaired cholinergic neurotransmission in subjects with MCI who
cognitive performance in young healthy patients to a level demonstrate an increased risk of developing Alzheimer’s
detectable in dementing disorders, while enhancement of disease, has been shown to specifically improve hippocam-
central cholinergic function improved the performance of pal function, thus further supporting the suggestion that
aged patients (Drachman, 1977). cholinergic deficits in MCI are functionally relevant (Gron
A vast number of experimental studies then has con- et al., 2006), but may appear to be modified by compensa-
firmed that defined administrations of anticholinergic drugs tory neurochemical mechanisms in the cholinergic system
Cholinergic transmission and Alzheimer’s disease 1627

(DeKosky et al., 2002). Cholinergic challenges in Alzheimer progression (Nordberg, 2001). While there is no doubt that
patients and in subjects with MCI differentially affected severe loss of cortical cholinergic innervation exists already
hippocampal activation as revealed by functional magnetic in the initial stages of presenile (early onset) as well as in
resonance imaging (MRI). These data support the sug- the advanced stages of late-onset Alzheimer’s disease, the
gestion of a key role of the cholinergic system in the func- assumption of cholinergic denervation being an early and
tional processes that lead to Alzheimer’s disease (Goekoop initial stage also in mild, late-onset Alzheimer’s disease has
et al., 2006). been debated (for review and references, see Mesulam,
2004). The cholinergic changes in late-onset Alzheimer’s
disease should also be considered in the light of normal
Cholinergic dysfunction in Alzheimer’s disease
age-related decline in cholinergic function. During aging
In late 1970s several groups provided evidence of severe cholinergic neurons of the basal forebrain complex, have
deficiency of presynaptic cholinergic markers in the ce- been described to undergo moderate degenerative changes,
rebral cortex of patients with early-onset of Alzheimer’s including cell atrophy, downregulation of choline acetyl-
disease (Davies and Malloney, 1976; Bowen et al., 1976; transferase (ChAT) activity, and mild cell loss detectable in
Perry et al., 1977a, b; see also Bigl et al., 1990). The cor- both animals and humans, resulting in cholinergic hypo-
relation of clinical dementia ratings with the reductions in function that has been related to the progressing memory
a number of cortical cholinergic markers such as choline deficits with aging (Bigl et al., 1990; Wenk, 1993; Arendt
acetyltransferase, muscarinic and nicotinic acetylcholine et al., 1987). A correlation of the loss of cholinergic neu-
receptor binding as well as levels of acetylcholine (Bierer rons of the basal forebrain and the extent of cognitive distur-
et al., 1995; Fr€
olich and Riederer, 1992; Gsell et al., 1993, bance as assessed by scoring the Mini mental state (MMS)
1997, 2004; Moll et al., 1990; Nordberg, 1992; Sofic et al., in both healthy individuals and a number of various demen-
1989), suggested an association of cholinergic hypofunc- ting disorders, revealed that cognitive deficits are detect-
tion with cognitive deficits, which led to the formulation of able not earlier before at least 30% of the total cholinergic
the cholinergic hypothesis of memory dysfunction in senes- basal forebrain cells have degenerated (Fig. 2). This indi-
cence and in Alzheimer’s disease (see, Bartus, 2000). This cates a high capacity of the basal forebrain cholinergic
is further supported by in vivo measurements of choliner- system with regard to respond to neuronal degeneration.
gic receptor expression by positron emission tomography Degeneration of basal forebrain cholinergic cells has
(PET) at different stages of Alzheimer’s disease demon- also been observed in a number of other dementing disor-
strating receptor abnormalities that change with disease ders (Table 1), such as Parkinsons’s disease (Arendt et al.,

Fig. 2. Correlation of the loss of cholinergic


neurons of the basal forebrain and the extent
of cognitive disturbance as assessed by scor-
ing the Mini mental state (MMS). This rela-
tionship is independent of the genesis of the
neurodegeneration. Healthy individuals dis-
play MMS scores of around 30, while
MMS scores of 24 or lower are considered
to represent a dementing state. In order to
reach a MMS score of 24, a loss of choliner-
gic neurons by about of 30% is required,
indicating a high capacity of the cholinergic
system to respond to neurodegenerative pro-
cesses (modified according to Arendt, 1999)
1628 R. Schliebs, T. Arendt

Table 1. Disorders with cognitive deficits which are accompanied by First evidence of a link between cholinergic dysfunction
cholinergic neuronal cell loss in the nucleus basalis Meynert
and APP processing was provided by observations of a co-
Alzheimer’s disease localization of acetylcholinesterase (AChE) with b-amy-
Parkinson’s disease loid deposits in Alzheimer brains (Mesulam, 1986; Moran
Parkinsonism with dementing complex of Guam
Striatonigral degeneration et al., 1993), which was further validated by in vitro studies
Down syndrome (Nitsch et al., 1992). Secretion of sAPPa was enhanced by
Dementia pugilistica electrical stimulation of tissue slices from rat brain, which
Pick’s disease
Wernicke-Korsakoff syndrome
could be blocked by the sodium-channel antagonist tetro-
Presenile argyrophilic subcortical dystrophy dotoxin (Nitsch et al., 1993). Selective activation of M1=
progressive supranuclear palsy M3-but not M2=M4-mAChR increased sAPPa secretion
Jakob-Creutzfeld disease
and decreased total b-amyloid formation both in vitro
Olivo-ponto cerebellar atrophy
subacute sklerotic panencephalitis (Ensinger et al., 1993: Farber et al., 1995; Walland et al.,
traumatic brain injury (boxing syndrome) 1997; M€uller et al., 1997) and in vivo in patients with
Alzheimer’s disease (Hock et al., 2003; Nitsch et al., 2003).
A similar effect could be achieved by direct activation of
1983; Jellinger, 1991, 2000; Whitehouse et al., 1983), protein kinase C (PKC) by phorbol ester indicating that
Down-syndrome, progressive supranuclear palsy, Jakob- mAChR mediate their effects on APP processing through
Creutzfeld disease (Arendt et al., 1984b), Korsakoff’s syn- activation of the phosphatidyl inositol signaling pathway
drome (Bohnen et al., 2003; Terry and Buccafusco, 2003) as (Hung et al., 1993). However, inhibition of sAPPa in the
well as after chronic ethanol intake (Arendt, 1994), and presence of PKC inhibitors was not complete (Slack et al.,
traumatic brain injury (Salmond et al., 2005). However, in 1995), indicating that other pathways downstream of the
contrast to many other dementing disorders, in Alzheimer’s acetylcholine receptor including tyrosine kinase (Slack et al.,
disease the cholinergic dysfunctions are accompanied by the 1995) or MAP kinase (Haring et al., 1998; Slack, 2000)
occurance of two major histopathological hallmarks such as may also be involved in the control of APP processing.
b-amyloid plaques deposed extracellularly in cerebral corti- Moreover, inhibitors of AChE were found to increase se-
cal and hippocampal areas and neurofibrillary tangles that cretion of sAPPa in both cortical rat brain slices (Mori
occupy much of the cytoplasm of select cortical pyramidal et al., 1995) and cell culture (Racchi et al., 2001). Selective
neurons. Despite a considerable progress in understanding lesion of basal forebrain cholinergic cells in rat brain
the biochemical processes of formation and deposition of (Roßner et al., 1998a; Lin et al., 1999), or administration of
b-amyloid peptides in Alzheimer’s disease, the molecular selective M1-muscarinic agonists to mice (Seo et al., 2002)
mechanisms of what causes the specific degeneration of provided strong in vivo evidence that cortical APP proces-
basal forebrain cholinergic cells is still a matter of debate. sing is controlled by cholinergic activity originating from
But during the last decade increasing evidence has accumu- the basal forebrain. Scopolamine treatment of transgenic
lated suggesting an interrelationship between basal forebrain Tg2576 mice resulted in increased levels of fibrillar b-amy-
cholinergic neurotransmission, neurotrophin signaling, and loid (1–40) and b-amyloid (1–42), and enhanced a-secre-
metabolism of the amyloid precursor protein (APP), which tase activity, suggesting that chronic suppression of cortical
will be highlighted in the next chapters. muscarinic cholinergic transmission may alter the balance
between a- and b-secretory APP processing by favouring
the amyloidogenic route (Liskowsky and Schliebs, 2006).
Cholinergic control of amyloid precursor
The pathway by which mAChR regulate a-secretase-
protein processing
mediated sAPPa release occurs via PKCa and Src-tyro-
The major constituent of the neuritic plaques is the 4 kDa sine kinase-dependent activation of ERK1=2 (Canet-Aviles
b-amyloid peptide that is derived from a much larger amyloid et al., 2002). Given that a-and b-secretory APP processing
precursor protein (APP) by the sequential action of b- and are differentially regulated through mAChR subtypes, we
g-secretases. Alternatively, the non-amyloidogenic secretory recently examined in SH-SY5Y neuroblastoma cells the sig-
pathway of APP processing includes the proteolytic cleavage naling pathways through which mAChR subtypes control
of APP by the a-secretase between amino acid position 16 b-secretase activity (Z€uchner et al., 2004). The expres-
and 17 of the b-amyloid domain that results into the secretion sion of the b-secretase BACE1 was found to be differen-
of sAPPa and precludes the generation of b-amyloid (for tially regulated in a subtype-specific manner by mAChR.
review on APP processing, see e.g. Selkoe, 2003). Agonist binding to M1=M3-receptors upregulated BACE1
Cholinergic transmission and Alzheimer’s disease 1629

expression through activation of both protein kinase C and strong staining for AChE activity was found throughout
MAP kinase signaling cascades. In contrast, BACE1 ex- the b-amyloid plaques accompanied by local distortions
pression was downregulated by activation of M2-mAChR of the cholinergic fiber network. The amyloid plaque core
and protein kinase A-mediated pathways (Z€ uchner et al., was observed to be associated with intense AChE activity
2004). These results may partly explain the observed dete- and a weakly stained AChE-positive halo (Apelt et al., 2002).
riorations of Alzheimer’s disease patients after initial In doubly transgenic mice overexpressing both human
improvements by AChE-inhibitor or M1-muscarinic ago- AChE and the Swedish mutation of human APP acceler-
nist treatment. ated plaque formation as well as larger plaque size have
Nicotine through action on nicotinic nAChR has also been observed, as compared to single transgenic APP mice
been observed to modulate APP processing both in cell (Rees et al., 2003). These findings provide strong evidence
culture (Efthimiopoulos et al., 1996; Seo et al., 2001) and for an important role of AChE in amyloid deposition.
in vivo (Utsuki et al., 2002) by favouring the non-amyloi- AChE interacts with b-amyloid by the peripheral anionic
dogenic pathway when treated with low doses of nicotine binding site (PAS) of the enzyme (Inestrosa et al., 1996;
(Lahiri et al., 2002). Moreover, inhibition of b-amyloid Inestrosa and Alarcon, 1998; Reyes et al., 1997), which led
fibril formation and disruption of preformed b-amyloid fi- to the suggestion that inhibition of the PAS of the AChE
brils by nicotine was described (Salomon et al., 1996; Ono may represent a potential strategy to intervene in b-amy-
et al., 2002). Chronic treatment of transgenic Tg2576 APP loid formation (Fuentealba et al., 2004).
mice with nicotine significantly reduced b-amyloid plaque However, vice versa, there are also reports that b-amy-
deposition as compared to non-treated controls (Nordberg loid increased AChE in both neuronally differentiated P19
et al., 2002; Hellstrom-Lindahl et al., 2004) suggesting cells and in primary murine cortical cells (Sberna et al.,
beneficial effects of nicotine for Alzheimer’s disease pa- 1997; Saez-Valero et al., 2003; Fodero et al., 2004), with
tients. This is further emphasized by a recent study in b-amyloid (1–42) being more potent than b-amyloid (1–40).
tobacco smoking elderly people who demonstrated less The b-amyloid (1–42)-induced enhancement of AChE was
b-amyloid deposition in the entorhinal cortex as compared found to be mediated through the action b-amyloid on a7
to non-smokers (Court et al., 2005). nicotinic acetylcholine receptors (nAChR), indicating that
the local increase in AChE around amyloid plaques in
Alzheimer’s disease may also be a result of a direct action
Acetylcholinesterase and b-amyloid
of b-amyloid on a7 nAChR located on terminals around
Co-localization of acetylcholinesterase (AChE) with b- senile plaques (Fodero et al., 2004).
amyloid deposits in Alzheimer brains (Mesulam, 1986;
Moran et al., 1993) as well as the capability of AChE to
Butyrylcholinesterase and Alzheimer’s disease
affect APP processing (Lahiri et al., 1997; Mori et al., 1995;
Racchi et al., 2001), and aggregation of b-amyloid pep- Recent studies examining butyrylcholinesterase (BuChE)
tides (Inestrosa et al., 1996) suggested a particular role of in post mortem brain samples from dementia patients in re-
the acetylcholine-degrading enzyme in the pathogenesis of lationship to the progression of cognitive impairment sug-
Alzheimer’s disease (see e.g., Zhang, 2004; Racchi and gested an essential role of BuChE in disease progression.
Govoni, 2003). Both BuChE and AChE appear to exert broader functions
AChE appears to promote the aggregation of b-amyloid in the central nervous system than previously thought,
at least in vitro by forming a complex with the growing which may also have consequences on treatment strategies
b-amyloid fibrils (Alvarez et al., 1997; Bartolini et al., with cholinesterase inhibitors by selectively targeting AChE
2003). This association of AChE with b-amyloid induces and BuChE (Ballard et al., 2005; Lane et al., 2006; Tasker
alterations in some of the enzyme properties (pH sensitiv- et al., 2005; Darreh-Shori et al., 2006).
ity, substrate concentration) and increases the neurotoxic- BuChE, the sister enzyme of AChE, is a serine hydrolase
ity of Alzheimer fibrils (Alvarez et al., 1998; Reyes et al., that catalyses the hydrolysis of esters of choline, including
2004), which correlates with corresponding findings in se- acetylcholine (Ballard et al., 2005; Darvesh et al., 2003),
nile plaques from Alzheimer brains (Geula and Mesulam, but its role in the brain still needs to be elucidated. It
1989; Schatz et al., 1990). A role of AChE in b-amyloid has been associated with cellular proliferation and neurite
plaque formation was also supported by in vivo studies. In growth during the development of the nervous system.
the neocortex of 18-month-old transgenic Tg2576 mice However, the viability of mice deficient of AChE but with
that demonstrated a considerable b-amyloid burden, a normal level of BuChE (Li et al., 2000; Mesulam et al.,
1630 R. Schliebs, T. Arendt

2002a), suggests an essential function of BuChE in the deposit assumes a compact b-sheat conformation, suggest-
brain in overtaking some actions of the AChE and compen- ing a role of BuChE in the transformation of b-amyloid
sating for the loss of AChE. This is further supported by its into neuritic plaques (Mesulam and Geula, 1994; Guillozet
widely distribution in the nervous system. The enzyme is et al., 1997). Indeed, selective inhibition of BuChE (cym-
primarily expressed in neuronal somata and proximal den- serine analogs) in both SK-N-SH-neuroblastoma cells, rat
drites in the cerebral cortex, mainly in deeper layers, in the brain, and double transgenic mice (overexpressing human
hippocampal formation, amygdala, and in many thalamic APPswe and presenilin-1) resulted into elevated acetylcho-
nuclei, while the AChE can be detected in somata and line level, enhanced long-term potentiation and in
axons (Darvesh et al., 1998; Darvesh and Hopkins, 2003). decreased b-amyloid level (Greig et al., 2005).
Particularly, in the thalamus, BuChE and AChE-positive Moreover, BuChE has also been observed in both intra-
neurons displayed both distinct and overlapping distribu- and extracellular neurofibrillary tangles, neurites associ-
tion patterns with some nuclei primarily expressing one ated with plaques and in neuropil threads (Perry et al.,
or the other cholinesterase (Darvesh and Hopkins, 2003). 2003; Gomez-Ramos et al., 1994). Interestingly, studies in
AChE appears localized mainly in neurons, whereas BuChE autopsy-diagnosed dementia (Alzheimer’s disease and de-
is primarily associated with glial cells, as well as with mentia of Lewy bodies) demonstrated that patients with the
endothelial cells and neurons (Ballard, 2002; Darvesh K-variant of BuChE have less severe phosphorylation of
et al., 2003). tau in the temporal cortex (Ballard et al., 2005), which may
It has been hypothesized that AChE and BuChE are co- be of therapeutic relevance. The properties of cholines-
regulators of the duration of action of acetylcholine in terases detected in plaques and tangles were similar to
cholinergic neurotransmission (Darvesh et al., 1998; Xie those found in glial cells (Wright et al., 1993), suggesting
et al., 2000; Mesulam et al., 2002a, b). This suggestion is a glial source of plaque-associated BuChE.
further supported by the different kinetic properties of both Taken together, selective targeting of BuChE might be of
AChE and BuChE (Greig et al., 2005). BuChE is less effi- therapeutic value in Alzheimer’s disease (Darvesh et al.,
cient in hydrolysing acetylcholine at low concentrations but 2003). However, a recent in vivo study failed to observe
highly effective at high acetylcholine levels at which AChE increased synaptic BuChE activity in brain of Alzheimer
becomes subtrate inhibited (Kamal et al., 2006; Giacobini, patients (Kuhl et al., 2006).
2003), indicating a supportive role of acetylcholine hydro-
lysis by BuChE under conditions of high brain activity.
Toxicity of b-amyloid on the cholinergic system
The single gene on chromosome 3 that codes BuChE
displays polymorphisms, resulting in several enzymes with It has been hypothesized that b-amyloid peptides induce
different levels of activity, such as the atypical, the K- and neurodegenerative changes at cholinergic terminals (Auld
J-variants of BuChE (Bartels et al., 1992a, b). Moreover, et al., 2002; Selkoe, 2002; Dolezal and Kasparova, 2003;
BuChE exists in several different molecular forms, includ- Harkany et al., 2000; Yan and Feng, 2004). However, it is
ing monomers and oligomers that consist of identical cat- still a matter of debate whether the loss and degeneration of
alytic subunits (Darvesh et al., 2003). cholinergic terminals are primary events or secondary to
In healthy human brain AChE predominates over BuChE the b-amyloid plaque pathology. The most predominant
(Giacobini, 2003). However, in Alzheimer’s disease, in b-amyloid-peptides detected in Alzheimer plaques are b-
contrast to AChE which is lost early during the course of amyloid (1–40) and b-amyloid (1–42) (Masters et al., 1985;
the disease (see above), BuChE levels, mainly the G1 form, Selkoe et al., 1986). b-amyloid (1–42) is more fibrillogenic
rise with disease progression (Perry et al., 1978; Arendt and displays higher neurotoxicity in vivo than b-amyloid
et al., 1992). The ratio of BuChE to AChE increases strik- (1–40) (Klein et al., 1999). Both peptides are able to
ingly in cortical regions of Alzheimer patients (Arendt form several aggregates with different physicochemical and
et al., 1984a; Perry et al., 1978; Giacobini, 2003). BuChE pharmacological properties: soluble, low-molecular weight
occurs in plaques, tangles, and amyloid-containing vessels monomers (Taylor et al., 2003), oligomers and amyloid-
(Arendt et al., 1992; Geula et al., 1994), and the levels of derived diffusible ligands (ADDLs), respectively (Lambert
BuChE correlate positively with the development of amy- et al., 1998; Kirkitadze et al., 2001; Walsh et al., 2002a;
loid plaques and neurofibrillar tangles in Alzheimer brains Bitan et al., 2003; Chromy et al., 2003; Gong et al., 2003),
(Geula et al., 1994; Geula and Mesulam, 1995; Guillozet protofibrils (Harper et al., 1997; Walsh et al., 1999) in
et al., 1997). The BuChE becomes associated with amyloid equilibrium with insoluble, high-molecular weight fi-
plaques at approximately the same time that the b-amyloid brils (Teplow 1998; Walsh et al., 1999). The insoluble,
Cholinergic transmission and Alzheimer’s disease 1631

high-molecular weight fibrils are major components of the Modulatory role of b-amyloid on cholinergic function
senile plaques in the Alzheimer brain and are assumed to
b-amyloid is also produced under normal conditions and
be the most toxic forms responsible for cholinergic neuro-
secreted in the brain as a soluble peptide (Haass and
degeneration (Arendt et al., 1985; Arendt and Bigl, 1986).
Selkoe, 1993; Shoji et al., 1992), which raised the possibi-
Cell culture experiments have shown that the insoluble
lity that b-amyloid may also play a physiological role. This
b-amyloid fibrils have a neurotoxic effect (Pike et al., 1991;
is supported by cell culture studies providing evidence of a
Lorenzo and Yankner, 1994).
modulatory role of soluble b-amyloid on cholinergic neu-
The toxicity of b-amyloid plaques has been suggested to
rotransmission (see e.g., Auld et al., 2002). For example,
be affected by advanced glycation endproducts (AGEs),
solubilized b-amyloid at pM to nM concentrations strongly
formed by reaction of proteins with reactive sugars or
inhibits the potassium-stimulated release of acetylcholine
dicarbonyl compounds (M€ unch et al., 1994). AGEs detect-
from hippocampal slices (Kar et al., 2004) and decreases
ed in neuritic but not diffuse b-amyloid plaques in aged
activity of choline acetyltransferase but not AChE activity
Alzheimer patients (L€ uth et al., 2005), are able to cross-
in cholinergic SN56 cells (Pedersen et al., 1996). b-amy-
link proteins and to activate glial cells, thus contributing
loid peptides also decrease the intracellular acetylcholine
to plaque stability and plaque-induced inflammation in
concentration (Hoshi et al., 1997) and impair M1-mAChR
Alzheimer’s disease (M€ unch et al., 1997a, b, 1998, 2003).
associated signaling (Kelly et al., 1996) in primary septal
However, transgenic mice that express the Swedish dou-
or cortical cultures. Exposure of PC12 cells to b-amyloid
ble mutation of human APP (APPsw) show a surprisingly
resulted into significant decrase in nAChR which led to the
minimal correlation among the accumulation of insolu-
suggestion that b-amyloid can degenerate nAChR (Guan
ble Ab-peptides, neuronal losses and memory impairments
et al., 2001; Liu et al., 2001) which could explain the loss
(Irizarry et al., 1997; Westerman et al., 2002, for review,
of nAChR binding observed in Alzheimer brains (Nordberg,
see also Schliebs, 2005b). Additionally, there are reports of
1992; Teaktong et al., 2004). Otherwise, there a reports that
cognitively normal individuals with high b-amyloid burden
b-amyloid may affect nAChR by direct binding with high-
in their brains (Delaere et al., 1990). Thus, these novel
affinity to nAChR, in particular to the a7 subtype (Wang
aspects of b-amyloid actions have led to a re-evaluation
et al., 2000a, b), while other studies have been unable con-
of the amyloid hypothesis of Alzheimer’s disease (Hardy
firm these findings (Liu et al., 2001; Guan et al., 2003).
and Selkoe, 2002).
b-amyloid has been observed to act as an agonist of
An alternative hypothesis involves the roles of soluble,
a7 nAChR (Fodero et al., 2004) mediating the activation
low-molecular weight monomers, oligomers and protofi-
of the ERK2 MAP kinase signaling cascade (Dineley et al.,
brils of b-amyloid and their effects on neurons. It has been
2001, 2002), while other groups have reported inhibitory
shown that b-amyloid oligomers are synaptic-specific li-
actions of b-amyloid on a7 nAChR (Dineley et al., 2002;
gands, which are able to inhibit hippocampal long-term
Pettit et al., 2001; Tozaki et al., 2002; Grassi et al., 2003;
potentiation in brain slices and rat brains in vivo (Walsh
Lee and Wang, 2003), which appears to depend on the
et al., 2002b; Wang et al., 2002). Protofibrils induce elec-
concentration of b-amyloid, low concentration can activate,
trophysiological changes and neurotoxicity in cortical neu-
higher concentrations desensitize a7 nAChR (Dineley et al.,
rons (Hartley et al., 1999). In Alzheimer brains, it has been
2002). Therefore, parts of the cholinergic deficits produced
observed that the severity of neurodegeneration correlates
in Alzheimer’s disease as well as in transgenic APP mice
best with the pool of soluble b-amyloid than with the num-
could be attributed to the suppression of cholinergic func-
ber of insoluble b-amyloid plaques (McLean et al., 1999).
tions by b-amyloid peptides apart of cell death (Blusztajn
In different cell and animal models prefibrillar assemblies
and Berse, 2000). This is supported by findings in Alzheimer
of b-amyloid have been shown to induce neurotoxicity, elec-
patients, that the cholinergic deficits coincide with the de-
trophysiological changes and disruption of cognitive func-
position of b-amyloid plaques at the earliest histopatholog-
tion (Hartley et al., 1999; Walsh et al., 1999, 2002; Kayed
ical stages, before the onset of clinical symptoms (Beach
et al., 2003; Wang et al., 2002; Cleary et al., 2005), which
et al., 2000).
may explain why early onset cholinergic dysfunction is in
Recently, it has been shown that memory deficits in
progress before there is considerable plaque formation in
middle-aged transgenic Tg2576 mice that do not exhibit
Alzheimer’s disease. This is further supported by a recent
significant b-amyloid plaque load, are caused by the extra-
cell culture study demonstrating that only soluble oligomeric
cellular accumulation of a 56-kDa soluble b-amyloid as-
but not fibrillar b-amyloid (1–42) forms induced toxicity in
sembly (Ab 56), as Ab 56 purified from the brains of
cholinergic SN56.B5.G4 cells (Heinitz et al., 2006).
1632 R. Schliebs, T. Arendt

impaired Tg2576 mice disrupted memory when admin- while the other three forms contain four microtubule bind-
istered to young rats (Lesne et al., 2006). It has been ing repeats. Single cell gene expression profiling revealed a
suggested that Ab 56 impairs memory independently of shift in the ratio of three-repeat tau to four-repear tau in
plaques, and may contribute to cognitive deficits associated individual human cholinergic basal forebrain neurons with-
with Alzheimer’s disease (Lesne et al., 2006). However, it in the nucleus basalis and CA1 hippocampal neurons dur-
remains to be elucidated whether Ab 56 exerts its action ing the progression of Alzheimer’s disease but not during
by affecting also the cholinergic transmitter system. normal aging (Ginsberg et al., 2006a). While the functional
significance of this shift still remains to be elucidated,
changes in tau isoform expression in vulnerable popula-
Cholinergic dysfunction and tau protein
tions of neurons may contribute to the cholinergic cell loss
Neurofibrillary tangles, one of the hallmarks of Alzheimer’s and cognitive deficits in Alzheimer’s disease.
disease, represent intracellular inclusions formed by aggre-
gates of hyperphosphorylated microtubule-associated tau
Particular vulnerability of basal forebrain
protein and which are found in select neuronal populations
cholinergic neurons to pathogenic stimuli
in Alzheimer’s disease (Kosik et al., 1986; Lee et al., 1991).
While in the brains of Alzheimer patients no tau mutations It is interesting to note that cholinergic cells located in
have been described, pathogenic mutations in the tau genes the pontine reticular formation are relatively preserved in
cause frontotemporal dementia (see e.g. Goedert and Jakes, Alzheimer’s disease indicating that brain cholinergic neu-
2005) suggesting that post-transcriptional alterations in tau rons demonstrate differential sensitivity to pathogenic in-
gene expression may contribute to the cognitive deficits in sults. This is emphasized by cell culture and in vivo studies
Alzheimer’s disease presumably also by interacting with demonstrating that basal forebrain cholinergic neurons are
the cholinergic transmission. Several studies have demon- more susceptible to toxic agents such as aluminium, nitric
strated that activation of nAChR results in a significant in- oxide, ethanol as compared to those in striatum and brain
crease in tau phosphorylation, whereas mAChR activation stem (Arendt et al., 1981; Fass et al., 2000; Julka et al.,
may prevent tau phosphorylation (Hellstrom-Lindahl et al., 1995). RN46A neuroblastoma cells from raphe nucleus,
2000; Wang et al., 2003; see also Rubio et al., 2006, for when differentiated to cholinergic phenotype with ciliary
review). Nicotine-induced increase in tau phosphoryla- neurothrophic factor appeared to be more susceptible to
tion at specific sites that were also hypophosphorylated in b-amyloid than those differentiated to serotoninergic phe-
Alzheimer’s disease, and is presumably mediated mainly notype with brain derived neurothrophic factor (Olesen
through the alpha7 subtype of nAChR (Wang et al., 2003). et al., 1998) suggesting that cholinergic neurons are more
Triple transgenic Alzheimer-like mice that overexpress mu- susceptible to toxic agents than other neuronal cell types,
tated human amyloid precursor protein, presenilin-1 and which may explain why non-cholinergic transmitter sys-
tau (Oddo et al., 2003) developed age-and regional depend- tems such as glutamate or GABA appear less affected in
ent accumulation of plaques and tangles and progressive Alzheimer’s disease (Armstrong et al., 2003).
deficits in cognition (Billings et al., 2005; Kitazawa et al., Cholinergic cells differ from other neuronal cell types by
2005). Chronic nicotine administration to one-month-old utilizing acetyl-CoA not only for energy production but also
triple transgenic Alzheimer-like mice for five months did for acetylcholine synthesis. Thus the particular vulnerability
not change soluble b-amyloid levels but resulted in a strik- and differential susceptibility of cholinergic neurons to var-
ing increase in phosphorylation and aggregation state of ious toxic conditions has been suggested to be due to dif-
tau, a finding that is presumably mediated through the loss ferent ratios of their acetyl-CoA energy producing and
of alpha7 nAChR observed in 6-months-old triple trans- acetylcholine synthesizing capacities. Cholinergic cells ap-
genic mice (Oddo et al., 2005). pear to have a higher demand for energy production and
Cholinergic basal forebrain neurons have been shown to therefore should respond more sensitive to energy (glucose)
demonstrate tau pathology both in patients with mild cog- deprivation (Szutowicz et al., 2006). Indeed, neurotoxic
nitive impairment and in Alzheimer patients (Mufson et al., compounds such as aluminium (Platt et al., 2001), nitric
2002; Mesulam, 2004; Counts and Mufson, 2005; Ginsberg oxide and reactive oxygen species (Law et al., 2001) have
et al., 2006a). In the adult human brain, six tau isoforms are been shown to contribute to the loss of central cholinergic
expressed through alternative splicing of a single tau gene neurons by affecting acetyl-CoA and energy metabolism
on chromosome 17 (Goedert et al., 1989a, b). Three iso- (Jankowska et al., 2000; Szutowicz, 2001). Moreover, acute
forms consist of three tandem microtubule binding repeats, stress has also been found to induce expression of the AChE
Cholinergic transmission and Alzheimer’s disease 1633

gene and to increase brain AChE activity (Kaufer et al., improves neuronal survival, increases sprouting of cholin-
1998), while in cell cultures oxidative stress has been ob- ergic neurons in vitro and in vivo (Dekker et al., 1991;
served to differentially affect transcriptions factors which Hefti et al., 1985) and may rescue cholinergic cells from
were activated by cholinergic stimulation (Li et al., 1996). lesion-induced atrophy (Williams et al., 1986; Wilcox et al.,
In Alzheimer’s disease brain glucose utilisation as well as 1995; Winkler et al., 2000). Administration of exogenous
some key enzymes of glucose metabolism are reduced (Bigl NGF to aged animals was found to reverse the age-related
et al., 1996, 2000). As insulin, insulin-like growth factors atrophic changes (Fischer et al., 1987) and to improve spa-
and their receptors, molecules which are known to regulate tial memory (Fischer et al., 1991). However, the aging-
glucose metabolism in brain, are also affected in Alzheimer’s related cholinergic atrophy and cell loss in normal brain
disease, it has been hypothesized that insulin-dependent as well as in Alzheimer’s disease is not complemented by
functions may be of pathogenetic relevance in sporadic reductions in the levels of NGF (Hellweg et al., 1998;
forms of the disease (Fr€ olich et al., 1998; Bernstein et al., Katoh-Semba et al., 1998; Murase et al., 1993; Narisawa-
1999; Hoyer, 2004). A relationship between glucose meta- Saito et al., 1996) as could be expected, which hints to a
bolism and cholinergic transmission and b-amyloid has also functional loss of aging cholinergic neurons to respond to
been derived from animal experimental approaches (Bigl NGF. Indeed, in aged rats, forebrain cholinergic neurons
et al., 2003; L€ offler et al., 2001; Mehlhorn et al., 1998) demonstrated striking reductions in the retrograde transport
suggesting functional links between cortical cholinergic of NGF, and cholinergic cells that were no longer capable
activity and glucose metabolism in cholinoceptive target re- to transport NGF appeared severely shrunken (Cooper et al.,
gions (for review, see also Schliebs, 2005a). 1994; deLacalle et al., 1996). Thus basal forebrain cholin-
There is evidence that cholinergic cells are also suscepti- ergic neurons seem to be particularly sensitive to failures
ble to inflammatory conditions, and anti-inflammatory drugs in NGF signalling that may induce cellular atrophy and
have been observed to attenuate inflammation-mediated cy- changes in gene expression (Sofroniew et al., 2001). The
totoxicity (see Wenk et al., 2000). The transcription factor recent development of transgenic mice that express a neu-
NFkB is a downstream signal molecule that may be acti- tralizing anti-NGF recombinant antibody (Capsoni et al.,
vated by stimulation of both type 1 interleukin (IL)-1 recep- 2000) added further evidence that failure in NGF signaling
tor and muscarinic cholinergic receptor-mediated signaling contributes to cholinergic neurodegeneration. These anti-
transduction pathway. Thus IL-1b may interfere with the NGFmice in which the levels of antibodies are three orders
cholinergic signal transduction cascade by inhibiting tran- of magnitude higher in adult than in newborn mice, demon-
scription factor activation, thus providing another mecha- strated age-dependent Alzheimer-like neurodegenerative
nism by which IL-1b may induce cholinergic dysfunction in pathology including formation of amyloid plaques, hyper-
Alzheimer’s disease (Schliebs et al., 2006). phosphorylated tau, and neurofibrillary tangles in cortex and
The e4-allele of the apolipoprotein E gene has been at- hippocamous (Capsoni et al., 2000). Aged anti-NGF mice
tributed to constitute a major genetic risk factor to develop also displayed extensive neuronal loss throughout the cor-
late-onset Alzheimer’s disease (Corder et al., 1993, 1998; tex, cholinergic deficits in the basal forebrain, and behav-
Cruz-Sánchez et al., 2000) and is assumed to affect the cho- ioural deficits (Capsoni et al., 2000).
linergic deficit. However, neither the apolipoprotein E knock- In addition to its protective function on basal forebrain
out nor the apolipoprotein E4 transgenic mice displayed any cholinergic cells, NGF signaling has also been shown to
changes in cholinergic parameters as compared to non-trans- influence expression and metabolism of APP (for review,
genic mice, suggesting that apolipoprotein E4 is not dele- see e.g. Isacson et al., 2002). In primary cortical neuronal
terious per se for the cholinergic system in mouse brain cultures enhanced expression of APP and secretion of
(Bronfman et al., 2000; Fagan et al., 1998). Moreover, apol- sAPPa was observed following NGF treatment (Wang
ipoprotein E deficiency did not increase the sensitivity to et al., 2000c). Stimulation of PC12 cells with NGF caused
cholinergic and spatial navigation deficits produced by ex- an increase in endogenous presenilin-1 level (Counts et al.,
citotoxic lesions of the nucleus basalis (Puolivali et al., 2000). 2001), while trophic factor withdrawal-induced death of
PC12 cells, resulted into enhanced APP expression (Araki
and Wurtman, 1998). NGF stimulates APP promotor ac-
Cholinergic dysfunction and nerve
tivity in PC12 cells involving activation of Ras-MAP
growth factor (NGF)
kinase signaling, while NGF-induced secretion of sAPP
NGF is specifically implicated in protection and mainte- is mediated by a Ras-independent mechanism (Villa et al.,
nance of cholinergic neurons in the basal forebrain. It 2001).
1634 R. Schliebs, T. Arendt

The chronic deprivation of NGF in the anti-NGF mice found to potentiate the neurotoxic action of b-amyloid me-
(AD11) leads to basal forebrain cholinergic deficits and to diated through p75NTR (Perini et al., 2002).
the formation and deposition of b-amyloid (Capsoni et al., In contrast to the findings in neuroblastoma cell lines, in
2002), which provides in vivo evidence of a direct link human primary neurons p75NTR was found to protect
between NGF signaling and APP processing. against extracellular b-amyloid toxicity in a phosphatidy-
NGF was also observed to modulate the cholinergic con- linositide 3-kinase-dependent but Akt-independent manner
trol of APP prosessing. In PC12 cells transfected with the (Zhang et al., 2003). This neuroprotective role of p75NTR
M1-mAChR subtype, NGF promoted sAPPa secretion fol- observed in human primary neuronal cell cultures is sup-
lowing M1 muscarinic agonist stimulation (Haring et al., ported by in vivo studies. p75NTR immunoreactivity is de-
1995). Similarly, treatment of mice with the selective M1 creased in basal forebrain cholinergic neurons of Alzheimer
muscarinic agonist RS86 enhanced secretion of sAPPa, while patients (Salehi et al., 2000; Mufson et al., 2002) suggest-
reducing the level of total APP and NGF in corticohippo- ing that a lack of p75NTR predisposes to neuronal degen-
campal regions (Seo et al., 2002). Chronic treatment of eration in Alzheimer’s disease (Mufson et al., 2002). This
transgenic Tg2576 Alzheimer mice with propentofylline, a compares well with observations in APPSwe=PS1 trans-
synthetic xanthine derivative known to stimulate synthesis genic mice with high b-amyloid plaque load, revealing
and release of NGF, resulted into increased NGF mRNA increased p75NTR protein level in the absence of any neu-
level and a major shift in the balance of APP processing ronal cell loss (Jaffar et al., 2001).
from the amyloidogenic to the non-amyloidgenic pathway However, recent observations that the transition from
compared to untreated controls (Chauhan and Siegel, 2003; mild cognitive impairment to Alzheimer’s disease is char-
Chauhan et al., 2005), thus providing also in vivo evidence acterized by a selective reduction of cortical TrkA protein
of a modulatory function of NGF on APP metabolism. but stable expression of cortical p75NTR (Counts et al.,
In NGF-responsive cells, following stimulation of the 2004), indicates that mainly the NGF receptor imbalance
high-affinity NGF receptor, TrkA, we have observed in- produces the deleterious effects on cholinergic basal fore-
creased secretory non-amyloidogenic APP processing, while brain function. However, chronic degeneration of cholin-
a transcriptional regulation of APP expression is mediated ergic basal forebrain neurons may also trigger reactive
through the low-affinity neurotrophin receptor p75NTR mechanisms of repair involving the action of different tro-
(Roßner et al., 1998b). This compares well with findings phic factors such as brain-derived neurotrophic factor and
in Alzheimer patients, a significant reduction in the expres- neurotrophin-3 (Durany et al., 2000).
sion of TrkA has been observed, while TrkB, TrkC and Although the beneficial effects of NGF on neuronal
p75NTR mRNA levels remained unchanged (for review, see maintenance and rescue from damage appeared promising
Salehi et al., 2004). Thus a loss of TrkA-mediated signaling to prove for the therapeutic potentials of NGF (Martinez-
in Alzheimer’s disease may also affect APP processing. Serrano and Bj€orklund, 1998; Rahimi and Juliano, 2001;
Alternatively, the production of b-amyloid may influence Roßner et al., 1996), the intracerebroventricular administra-
the membrane integrity of TrkA-bearing neurons resulting tion of NGF to Alzheimer patients (Eriksdotter-J€ onhagen
into disruption of NGF signaling with severe consequences et al., 1998) and to rodents (Winkler et al., 2000) produced
on maintenaince of basal forebrain cholinergic cells (Salehi a number of unwanted side effects. This may partly be due
et al., 2004). to the diversity of cells that respond to NGF signaling
b-amyloid may damage neurons either directly by inter- (Sofroniew et al., 2001). However, a recent study provided
acting with membrane components that initiate cell death evidence that NGF, when infused intraparenchymally to
signaling or indirectly by activating astro-or microglia which rats with complete unilateral fornix transections, rescued
secrete pro-inflammatory cytokines. In a number of neuro- basal forebrain cholinergic neurons, but did not induce any
blastoma cell lines including PC12, SK-N-BE, NIH3T3, and adverse effects at least over a two-week infusion period
SK-N-MC cells, it has been shown that p75NTR increases the (Tuszynski, 2000).
susceptibility of these cells to b-amyloid toxicity (Rabizadeh
et al., 1994; Yaar et al., 2002; Kuner et al., 1998; Perini et al.,
Enhancing cholinergic transmission as symptomatic
2002). p75NTR is reqired to induce the b-amyloid-mediated
and palliative therapy of Alzheimer’s disease
cytotoxicity by interacting with b-amyloid (Yaar et al.,
2002) and initiating the p75NTR -mediated activation of cas- Based on the assumption that the progressive decline of
pases-8 and 3 (Perini et al., 2002). Moreover, IL-1b and cognitive function in Alzheimer’s disease mainly results
TNFa produced by b-amyloid-activated microglia were from decreased cholinergic neurotransmission, pharmaco-
Cholinergic transmission and Alzheimer’s disease 1635

logical approaches to enhance basal forebrain cholinergic cognitive deficits in Alzheimer’s disease. This is further
pathways have been developed for symptomatic and pallia- emphasized by findings that chronic administration of nico-
tive therapy (Mount and Downtown, 2006). A number of tine to transgenic mice carrying the Swedish mutation of
acetylcholinesterase inhibitors including tacrine, donepezil, human APP, considerably reduced brain beta-amyloid pla-
rivastigmine and galantamine have been proved to enhance que deposition (Nordberg et al., 2002). Recently, long-term
cholinergic synaptic efficacy by preventing the breakdown tobacco smoking has been observed to attenuate b-amyloid
of acetylcholine and thus elevating and=or maintaining its deposition in the entorhinal cortex of normal elderly indi-
level in the synaptic cleft and to prolong its action on viduals (Court et al., 2005) which further underlines the
postsynaptic muscarinic and nicotinic receptors (for review, potential of developing a ‘‘nicotinic’’ treatment strategy.
see, e.g. Giacobini, 2004; Lane et al., 2004; Birks, 2006; Besides the administration of NGF, transplantation of ace-
Mount and Downtown, 2006; Nordberg, 2006). However, tylcholine-producing cells may also provide a useful tool to
acetylcholinesterase inhibitors produce only modest im- restore injured cholinergic neurotransmission. The substan-
provements in cognitive function (Bryson and Benfield, tial and persistent memory losses produced by chronic etha-
1997; Frisoni, 2001; Gauthier, 2002; Giacobini, 2001; nol intake were associated with an impairment of cholinergic
Greenberg et al., 2000; Mohs et al., 2001; Winblad et al., function, and wcould be reversed by cholinergic-rich trans-
2001) together with some unwanted side effects (Wilkinson, plants into cortex and hippocampus (Arendt et al., 1988).
1999; Dunn et al., 2000; Imbimbo, 2001), and a certain Hippocampal grafts of fibroblasts engineered to produce
portion of Alzheimer-patients does not even respond to this acetylcholine have been found to be sufficient to improve
treatment (Birks, 2006; Mount and Downtown, 2006). behavioral performance following unilateral fimbria-fornix
Therefore, it has to taken into consideration that the chron- lesion (Dickinson-Anson et al., 1998). Although fimbria-
ic treatment with AChE inhibitors may also induce com- fornix lesion disrupts a number of hippocampal transmitter
pensatory mechanisms at the cholinergic synapse that are systems, the lesion-induced deficits in water maze perfor-
counteracting to the desired action of the drugs. AChE mance could be attenuated by a target-specific replacement
inhibitor-mediated chronically increased level of acetylcho- of acetylcholine in the hippocampus. Immortalized brain
line at the synaptic cleft should lead to an overactivation of endothelial cells genetically modified to express ChAT
postsynaptic receptors followed by up-regulations of var- and=or the vesicular acetylcholine transporter have also been
ious postsynaptic events including inmediate early gene suggested as a promising tool for improving altered choli-
(IEG)-induced enhanced expression of AChE. Thus, the nergic function in the central nervous system (Malo et al.,
endogenously induced production of AChE may be respon- 1999). More recently, the potential of neural stem cells to
sible for the loss of efficacy of systemically applied AChE acquire cholinergic characteristics has been tested. A neural
inhibitors with the time of treatment (Hoyer and Riederer, stem cell line was transplanted into the septum=diagonal
2003; Riederer and Hoyer, 2005). band nuclei of young and adult rodents previously sub-
Recent progress in understanding the role of BuChE in jected to a lesion of the fimbria-fornix pathway (Doering
dementia (Ballard et al., 2005; Greig et al., 2005; Lane and Snyder, 2000). Subpopulations of the grafted cells
et al., 2006; Tasker et al., 2005), as well as the availability acquired a cholinergic neuronal phenotype and expressed
of selective inhibitors of BuChE (Kamal et al., 2006; Greig ChAT and p75NTR, indicating the potential of this stem cell
et al., 2005) has raised the question whether selective tar- line to rescue damages within the basal forebrain cholinergic
geting of either AChE or BuChE in dementing disorders system. Similarly, it has been demonstrated that condition-
may have greater clinical efficacy (Tasker et al., 2005), ally immortal neuroepithelial stem cells (MHP36), grafted
which however must await further clinical trials. into the cortex or basal forebrain of rats with basal forebrain
An alternative approach to enhance cholinergic neuro- cholinergic lesions produced a restoration of spatial learning
transmission includes the increase of acetylcholine release in the water maze already detectable six weeks after the
by activation of presynaptic nicotinic cholinergic receptors transplantation (Grigoryan et al., 2000), further suggesting
through appropriate nicotinic agonists (Oddo and LaFerla, that the development of immortal human stem cell lines may
2006). Recently, the subtype-selective nicotinic acetylcho- provide a strategy to treat neurodegenerative disorders.
line receptor ligand SIB-1553A has been shown to improve
cognitive deficits in Alzheimer patients in a similar mag-
Conclusions
nitude as the acetylcholinesterase inhibitor donezepil
(Bontempi et al., 2003), suggesting that nicotinic receptor Basal forebrain cholinergic dysfunctions in Alzheimer’s
activation may represent another promising strategy to treat disease appear to play an important role in the pathogenesis
1636 R. Schliebs, T. Arendt

of Alzheimer’s disease. The correlation of clinical demen- Arendt T, Bigl V, Walther F, Sonntag M (1984a) Decreased ratio of CSF
acetylcholinesterase to butyrylcholinesterase activity in Alzheimer’s
tia ratings with the decrease in a number of cholinergic disease. Lancet 1: 173
markers suggested an association of cholinergic hypofunc- Arendt T, Bigl V, Arendt A (1984b) Neurone loss in the nucleus basalis of
tion with cognitive deficits, which has been confirmed by a Meynert in Creutzfeldt-Jakob disease. Acta Neuropathol (Berl) 65:
85–88
number of experimental studies both in animals and hu-
Arendt T, Bigl V, Tennstedt A, Arendt A (1985) Neuronal loss in different
mans. Abnormal changes in the regulation of cholinergic parts of the nucleus basalis is related to neuritic plaque formation in
transmission very early in life has been assumed to con- cortical target areas in Alzheimer’s disease. Neuroscience 14: 1–14
Arendt T, Fischer S, Bigl V, Arendt A (1987) Age related changes in human
tribute to the later development of mild cognitive impair-
nucleus basalis of Meynert in comparison to dementing disorders. In:
ments, further supporting the suggestion of a key role of Trojan S, Stasny F (eds) Ontogenesis of the brain, Vol. 4. Universitas
the cholinergic system in functional processes that lead to Carolina, Praha, pp 53–55
Alzheimer’s disease. While there is strong evidence that Arendt T, Bruckner MK, Lange M, Bigl V (1992) Changes in acetylcho-
linesterase and butyrylcholinesterase in Alzheimer’s disease resemble
cortical cholinergic transmission plays a major role in the embryonic development – a study of molecular forms. Neurochem Int
controling APP processing and b-amyloid plaque for- 21: 381–396
mation as well as phosphorylation of tau protein, there is Armstrong DM, Sheffield R, Mishizen-Eberz AJ, Carter TL, Rissman RA,
Mizukami K, Ikonomovic MD (2003) Plasticity of glutamate and
still a debate whether the cholinergic deficits observed in
GABAA receptors in the hippocampus of patients with Alzheimer’s
Alzheimer’s disease are a primary event or secondary to the disease. Cell Mol Neurobiol 23: 491–505
pathomorphological features of the disease. Auld DS, Kornecook TJ, Bastianetto S, Quirion R (2002) Alzheimer’s
disease and the basal forebrain cholinergic system: relations to b-
amyloid peptides, cognition, and treatment strategies. Prog Neurobiol
68: 209–245
Acknowledgement Ballard CG (2002) Advances in the treatment of Alzheimer’s disease:
benefits of dual cholinesterase inhibition. Eur Neurol 47: 64–70
This study was supported by the Interdisciplinary Centre for Clinical
Ballard C, Morris C, Kalaria R, McKeith I, Perry R, Perry E (2005a) The k
Research (IZKF) at the Medical Faculty of the University of Leipzig.
variant of the butyrylcholinesterase gene is associated with reduced
phosphorylation of tau in dementia patients. Dement Geriatr Cogn
Disord 19: 357–360
References Ballard CG, Greig NH, Guillozet-Bongaarts AL, Enz A, Darvesh S (2005b)
Cholinesterases: roles in the brain during health and disease. Curr
Alvarez A, Alarcon R, Opazo C, Campos EO, Munoz FJ, Calderon FH, Alzheimer Res 2: 307–318
Dajas F, Gentry MK, Doctor BP, De Mello FG, Inestrosa NC (1998) Barbelivien A, Bertrand N, Besret L, Beley A, MacKenzie ET, Dauphin F
Stable complexes involving acetylcholinesterase and amyloid-b pep- (1999) Neurochemical stimulation of the rat substantia innominata
tide change the biochemical properties of the enzyme and increase the increases cerebral blood flow (but not glucose use) through the parallel
neurotoxicity of Alzheimer’s fibrils. J Neurosci 18: 3213–3223 activation of cholinergic and non-cholinergic pathways. Brain Res 840:
Alvarez A, Opazo C, Alarcon R, Garrido J, Inestrosa NC (1997) Acet- 115–124
ylcholinesterase promotes the aggregation of amyloid-beta-peptide Bartels CF, Jensen FS, Lockridge O, van der Spek AF, Rubinstein HM,
fragments by forming a complex with the growing fibrils. J Mol Biol Lubrano T, La Du BN (1992a) DNA mutation associated with the
272: 348–361 human butyrylcholinesterase K-variant and ist linkage to the atypical
Apelt J, Kumar A, Schliebs R (2002) Impairment of cholinergic neuro- variant mutation and other polymorphic sites. Am J Hum Genet 50:
transmission in adult and aged transgenic Tg2576 mouse brain expres- 1086–1103
sing the Swedish mutation of human beta-amyloid precursor protein. Bartels CF, James K, La Du BN (1992b) DNA mutations associated with the
Brain Res 953: 17–30 human butyrylcholinesterase J-variant. Am J Hum Genet 50: 1104–1114
Araki W, Wurtman RJ (1998) Increased expression of amyloid precursor Bartolini M, Bertucci C, Cavrini V, Andrisano V (2003) b-Amyloid
protein and amyloid precursor-like protein 2 during trophic factor aggregation induced by human acetylcholinesterase: inhibition studies.
withdrawal-induced death of neuronal PC12 cells. Mol Brain Res 56: Biochem Pharmacol 65: 407–416
169–177 Bartus RT (2000) On neurodegenerative diseases, models, and treatment
Arendt T (1994) Impairment in memory function and neurodegenerative strategies: lessons learned and lessons forgotten a generation following
changes in the cholinergic basal forebrain system induced by chronic the cholinergic hypothesis. Exp Neurol 163: 495–529
intake of ethanol. J Neural Transm Suppl 44: 173–187 Beach TG, Kuo YM, Spiegel K, Emmerling MR, Sue LI, Kokjohn K, Roher
Arendt T (1999) Pathological anatomy of Alzheimer’s disease (Germ). In: AE (2000) The cholinergic deficit coincides with Ab deposition at the
F€orstl H, Bickel H, Kurz A (eds) Alzheimer Demenz, Grundlagen Klinik earliest histopathologic stages of Alzheimer disease. J Neuropathol
und Therapie. Springer, Berlin Heidelberg New York, pp 87–106 Exp Neurol 59: 308–313
Arendt T, Bigl V (1986) Alzheimer plaques and cortical cholinergic Berger-Sweeney J (2003) The cholinergic basal forebrain system during
innervation. Neuroscience 17: 277–279 development and its influence on cognitive processes: important
Arendt T, Allen Y, Sinden J, Schugens MM, Marchbanks RM, Lantos PL, questions and potential answers. Neurosci Behav Rev 27: 401–411
Gray JA (1988) Cholinergic-rich brain transplants reverse alcohol- Bernstein HG, Ansorge S, Riederer P, Reiser M, Frolich L, Bogerts B
induced memory deficits. Nature 332: 448–450 (1999) Insulin-degrading enzyme in the Alzheimer’s disease brain:
Arendt T, Bigl V, Arendt A, Tennstedt A (1983) Loss of neurons in prominent localization in neurons and senile plaques. Neurosci Lett
the nucleus basalis of Meynert in Alzheimer’s disease, paral- 263: 161–164
ysis agitans and Korsakoff’s Disease. Acta Neuropathol (Berl) 61: Bierer LM, Haroutunian V, Gabriel S, Knott PJ, Carlin LS, Purohit DP, Perl
101–108 DP, Schmeidler J, Kanof P, Davis KL (1995) Neurochemical correlates
Cholinergic transmission and Alzheimer’s disease 1637

of dementia severity in Alzheimer’s disease: relative importance of the Chauhan NB, Siegel GJ, Feinstein DL (2005) Propentofylline attenuates tau
cholinergic deficits. J Neurochem 64: 749–760 hyperphosphorylation in Alzheimer’s Swedish mutant model Tg2576.
Biesold D, Inanami O, Sato A, Sato Y (1989) Stimulation of the nucleus Neuropharmacology 48: 93–104
basalis of Meynert increases cerebral cortical blood flow in rats. Christensen H, Maltby N, Jorm AF, Creasey H, Broe GA (1992) Cholin-
Neurosci Lett 98: 39–44 ergic ‘blockade’ as a model of the cognitive deficits in Alzheimer’s
Bigl M, Bleyl AD, Zedlick D, Arendt T, Bigl V, Eschrich K (1996) Changes disease. Brain 115: 1681–1699
of activity and isozyme pattern of phosphofructokinase in the brains of Chromy BA, Nowak RJ, Lambert MP, Viola KL, Chang L, Velasco PT,
patients with Alzheimer’s disease. J Neurochem 67: 1164–1171 Jones BW, Fernandez SJ, Lacor PN, Horowitz P, Finch CE, Krafft GA,
Bigl M, Beck M, Bleyl AD, Bigl V, Eschrich K (2000) Altered phospho- Klein WL (2003) Self-assembly of Ab (1–42) into globular neurotox-
fructokinase mRNA levels but unchanged isoenzyme pattern in ins. Biochemistry 42: 12749–12760
brains from patients with Alzheimer’s disease. Mol Brain Res 76: Cleary JP, Walsh DM, Hofmeister JJ, Shankar GM, Kuskowski MA, Selkoe
411–414 DJ, Ashe KH (2005) Natural oligomers of the amyloid-b protein
Bigl M, Apelt J, Eschrich K, Schliebs R (2003) Cortical glucose metabolism specifically disrupt cognitive function. Nat Neurosci 8: 79–84
is altered in aged transgenic Tg2576 mice that demonstrate Alzheimer Cooper JD, Lindholm D, Sofroniew MV (1994) Reduced transport of 125I-
plaque pathology. J Neural Transm 110: 77–94 NGF by cholinergic neurons and downregulated TrkA expression in
Bigl V, Schliebs R (1998) Simulation of cholinergic deficits – a novel the medial septum of aged rats. Neuroscience 62: 625–629
experimental approach to study pathogenetic aspects of Alzheimer’s Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC,
disease. J Neural Transm Suppl 53: 237–247 Small GW, Roses AD, Haines JL, Pericak-Vance MA (1993) Gene
Bigl V, Arendt T, Biesold D (1990) The nucleus basalis of Meynert during dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s
aging and in dementing neuropsychiatric disorders. In: Steriade M, disease in late onset families. Science 261: 921–923
Biesold D (eds) Brain cholinergic systems. Oxford University Press, Corder EH, Lannfelt L, Bogdanovic N, Fratiglioni L, Mori H (1998) The
Oxford, pp 364–386 role of APOE polymorphisms in late-onset dementias. Cell Mol Life
Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM (2005) Sci 54: 928–934
Intraneuronal Ab causes the onset of early Alzheimer’s disease-related Counts SE, Lah JJ, Levey AI (2001) The regulation of presenilin-1 by nerve
cognitive deficits in transgenic mice. Neuron 45: 675–688 growth factor. J Neurochem 76: 679–689
Birks J (2006) Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Counts SE, Nadeem M, Wuu J, Ginsberg SD, Saragovi HU, Mufson EJ
Database Syst Rev 25: CD005593 (2004) Reduction of cortical trkA but not p75NTR protein in early-stage
Bitan G, Kirkitadze MD, Lomakin A, Vollers SS, Benedek GB, Teplow Alzheimer’s disease. Ann Neurol 56: 520–531
DB (2003) Amyloid b-protein (Ab) assembly: Ab40 and Ab42 Counts SE, Mufson EJ (2005) The role of nerve growth factor receptors in
oligomerize through distinct pathways. Proc Natl Acad Sci USA cholinergic basal forebrain degeneration in prodromal Alzheimer
100: 330–335 disease. J Neuropathol Exp Neurol 64: 263–272
Blusztajn JK, Berse B (2000) The cholinergic neuronal phenotype in Court JA, Johnson M, Religa D, Keverne J, Kalaria R, Jaros E, McKeith IG,
Alzheimer’s disease. Metab Brain Dis 15: 45–64 Perry R, Naslund J, Perry EK (2005) Attenuation of Ab deposition in
Bohnen NI, Kaufer DI, Ivanco LS, Lopresti B, Koeppe RA, Davis JG, the entorhinal cortex of normal elderly individuals associated with
Mathis CA, Moore RY, DeKosky ST (2003) Cortical cholinergic tobacco smoking. Neuropathol Appl Neurobiol 31: 522–535
function is more severely affected in Parkinsonian dementia than in Cruz-Sanchez FF, Durany N, Thome J, Riederer P, Zambon D (2000)
Alzheimer’s disease: An in vivo positron tomography study. Arch Correlation between Apolipoprotein-E polymorphism and Alzhei-
Neurol 60: 1745–1748 mer’s disease pathology. J Alzheimers Dis 2: 223–229
Bontempi B, Whelan KT, Risbrough VB, Lloyd GK, Menzaghi F (2003) Darreh-Shori T, Brimijoin S, Kadir A, Almkvist O, Nordberg A (2006)
Cognitive enhancing properties and tolerability of cholinergic agents Differential CSF butyrylcholinesterase levels in Alzheimer’s disease
in mice: a comparative study of nicotine, donepezil, and SIB-1553A, a patients with the ApoE epsilon4 allele, in relation to cognitive function
subtype-selective ligand for nicotinic acetylcholine receptors. Neu- and cerebral glucose metabolism. Neurobiol Dis 2006 Sep 12; [Epub
ropsychopharmacology 28: 1235–1246 ahead of print]
Bowen DM, Smith CB, White P, Davison AN (1976) Neurotransmitter- Darvesh S, Hopkins DA (2003) Differential distribution of butyrylcholi-
related enzymes and indices of hypoxia in senile dementia and other nesterase and acetylcholinesterase in the human thalamus. J Comp
abiotrophies. Brain 99: 459–496 Neurol 463: 25–43
Bronfman FC, Moechars D, Van Leuven F (2000) Acetylcholine-positive Darvesh S, Hopkins DA, Geula C (2003) Neurobiology of butyrylcholi-
fibre deafferentation and cell shrinkage in the septohippocampal path- nesterase. Nat Rev Neurosci 4: 131–138
way of aged amyloid precursor protein London mutant transgenic Darvesh S, Grantham DL, Hopkins DA (1998) Distribution of butyrylcho-
mice. Neurobiol Dis 7: 152–168 linesterase in the human amygdala and hippocampal formation. J
Bryson HM, Benfield P (1997) Donepezil. Drugs Aging 10: 234–239 Comp Neurol 393: 374–390
Canet-Aviles RM, Anderton M, Hooper NM, Turner AJ, Vaughan PF (2002) Davies P, Maloney AJ (1976) Selective loss of central cholinergic neurons
Muscarine enhances soluble amyloid precursor protein secretion in in Alzheimer’s disease. Lancet 2: 1403
human neuroblastoma SH-SY5Y by a pathway dependent on protein Dekker AJ, Langdon DJ, Gage FH, Thal LJ (1991) NGF increases cortical
kinase C(alpha), src-tyrosine kinase and extracellular signal-regulated acetylcholine release in rats with lesions of the nucleus basalis.
kinase but not phospholipase C. Mol Brain Res 102: 62–72 Neuroreport 2: 577–580
Capsoni S, Ugolini G, Comparini A, Ruberti F, Berardi N, Cattaneo A DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski S,
(2000) Alzheimer-like neurodegeneration in aged antinerve growth Bennett DA, Cochran EJ, Kordower JH, Mufson EJ (2002) Upregula-
factor transgenic mice. Proc Natl Acad Sci USA 97: 6826–6831 tion of choline acetyltransferase activity in hippocampus and frontal
Capsoni S, Giannotta S, Cattaneo A (2002) b-amyloid plaques in a model cortex of elderly subjects with mild cognitive impairment. Ann Neurol
for sporadic Alzheimer’s disease based on transgenic anti-nerve 51: 145–155
growth factor antibodies. Mol Cell Neurosci 21: 15–28 DeLacalle S, Cooper JD, Svendsen CN, Dunnett SB, Sofroniew MV (1996)
Chauhan NB, Siegel GJ (2003) Effect of PPF and ALCAR on the induction Reduced retrograde labeling with fluorescent tracer accompanies
of NGF-and p75-mRNA and on APP processing in Tg2576 brain. neuronal atrophy of basal forebrain cholinergic neurons in aged rats.
Neurochem Int 43: 225–233 Neuroscience 75: 19–27
1638 R. Schliebs, T. Arendt

Delaere P, Duyckaerts C, Masters C, Beyreuther K, Piette F, Hauw JJ (1990) Fischer W, Wictorin K, Bj€orklund A, Williams LR, Varon S, Gage FH
Large amounts of neocortical bA4 deposits without neuritic plaques (1987) Amelioration of cholinergic atrophy and spatial memory
nor tangles in a psychometrically assessed, nondemented person. impairment in aged rats by nerve growth factor. Nature 329: 65–68
Neurosci Lett 116: 87–93 Fischer W, Bj€orklund A, Chen KS, Gage PH (1991) NGF improves spa-
Detari L, Rasmusson DD, Semba K (1999) The role of basal forebrain tial learning in aged rodents as a function of age. J Neurosci 11:
neurons in tonic and phasic activation of the cerebral cortex. Prog 1889–1906
Neurobiol 58: 249–277 Fodero LR, Mok SS, Losic D, Martin LL, Aguilar MI, Barrow CJ, Livett
Deutsch JA (1971) The cholinergic synapse and the site of memory. Science BG, Small DH (2004) a7-nicotinic acetylcholine receptors mediate an
174: 788–794 Ab (1–42)-induced increase in the level of acetylcholinesterase in
Dickinson-Anson H, Aubert I, Gage FH, Fisher LJ (1998) Hippocampal primary cortical neurones. J Neurochem 88: 1186–1193
grafts of acetylcholine-producing cells are sufficient to improve beha- Fr€olich L, Blum-Degen D, Bernstein HG, Engelsberger S, Humrich J,
vioural performance following a unilateral fimbria-fornix lesion. Neu- Laufer S, Muschner D, Thalheimer A, Turk A, Hoyer S, Zochling R,
roscience 84: 771–781 Boissl KW, Jellinger K, Riederer P (1998) Brain insulin and insulin
Dineley KT, Westerman M, Bui D, Bell K, Ashe KH, Sweatt JD (2001) b- receptors in aging and sporadic Alzheimer’s disease. J Neural Transm
amyloid activates the mitogen-activated protein kinase cascade via 105: 423–438
hippocampal a7 nicotinic acetylcholine receptors: In vitro and in vivo Fr€olich L, Dirr A, Riederer P, Hoyer S (1993) Effects of long-term recovery
mechanisms related to Alzheimer’s disease. J Neurosci 21: 4125–4133 from transient cerebral ischemia in rat brain: tissue levels of acetylcho-
Dineley KT, Bell KA, Bui D, Sweatt JD (2002) b-Amyloid peptide activates line, monoamines, and their metabolites. Neurochem Res 18(12):
a7 nicotinic acetylcholine receptors expressed in Xenopus oocytes. J 1239–1244
Biol Chem 277: 25056–25061 Fr€olich L, Kornhuber J, Hoyer S, Riederer P (1989) Reduction of cortical
Doering LC, Snyder EY (2000) Cholinergic expression by neural stem cell muscarinic receptors after inhibition of pyruvate dehydrogenase com-
line grafted to the adult medial septum=diagonal band complex. plex (PDHc) in rats – a model of dementia? J Neural Transm P-D Sect
J Neurosci Res 61: 597–604 1(1–2): 61
Dolezal V, Kasparova J (2003) b-Amyloid and cholinergic neurons. Fr€olich L, Riederer P (1992) Demenz vom Alzheimer-Typ: biochemische
Neurochem Res 28: 499–506 befunde und €atiologische hypothesen. Therapiewoche 42(9): 500–505
Drachman DA (1977) Memory and cognitive function in man: does the Frotscher M (1992) Specificity of interneuronal connections. Ann Anat 174:
cholinergic system have a specific role? Neurology 27: 783–790 377–382
Drachman DA, Leavitt J (1974) Human memory and the cholinergic Frisoni GB (2001) Treatment of Alzheimer’s disease with acetylcholines-
system. A relationship to aging? Arch Neurol 30: 113–121 terase inhibitors: bridging the gap between evidence and practice.
Dunn NR, Pearce GL, Shakir SA (2000) Adverse effects associated with the J Neurol 248: 551–557
use of donepezil in general practice in England. J Psychopharmacol 14: Fuentealba RA, Farias G, Scheu J, Bronfman M, Marzolo MP, Inestrosa NC
406–408 (2004) Signal transduction during amyloid-beta-peptide neurotoxicity:
Dunnett SB, Fibiger HC (1993) Role of forebrain cholinergic systems in role in Alzheimer disease. Brain Res Rev 47: 275–289
learning and memory: relevance to the cognitive deficits of aging and Gauthier S (2002) Advances in the pharmacology of Alzheimer’s disease.
Alzheimer’s dementia. Prog Brain Res 98: 413–420 CMAJ 166: 616–623
Durany N, Michel T, Kurt J, Cruz-Sanchez FF, Cervas-Navarro J, Riederer Geula C, Greenberg BD, Mesulam MM (1994) Cholinesterase activity in
P (2000) Brain-derived neurotrophic factor and neurotrophin-3 levels the plaques, tangles and angiopathy of Alzheimer’s disease does not
in Alzheimer’s disease brains. Int J Dev Neurosci 18: 807–813 emanate from amyloid. Brain Res 644: 327–330
Efthimiopoulos S, Vassilacopoulou D, Ripellino JA, Tezapsidis N, Robakis Geula C, Mesulam M (1989) Special properties of cholinesterases in the
NK (1996) Cholinergic agonists stimulate secretion of soluble full- cerebral cortex of Alzheimer’s disease. Brain Res 498: 185–189
length amyloid precursor protein in neuroendocrine cells. Proc Natl Geula C, Mesulam MM (1995) Cholinesterases and the pathology of
Acad Sci USA 93: 8046–8050 Alzheimer disease. Alzheimer Dis Assoc Disord 9 Suppl 2: 23–28
Ensinger HA, Doods HN, Immel-Sehr AR, Kuhn FJ, Lambrecht G, Mendla Giacobini E (2001) Do cholinesterase inhibitors have disease-modifying
KD, Muller RE, Mutschler E, Sagrada A, Walther G et al. (1993) WAL effects in Alzheimer’s disease? CNS Drugs 15: 85–91
2014–a muscarinic agonist with preferential neuron-stimulating prop- Giacobini E (2003) Cholinesterases: new roles in brain function and in
erties. Life Sci 52: 473–480 Alzheimer’s disease. Neurochem Res 28: 515–522
Eriksdotter-J€onhagen M, Nordberg A, Amberla K, Backman L, Ebendal T, Giacobini E (2004) Cholinesterase inhibitors: new roles and therapeutic
Meyerson B, Olson L, Seiger-Shigeta M, Theodorsson E, Vittanen M, alternatives. Pharmacol Res 50: 433–440
Winblad, Wahlund LO (1998) Intracerebroventricular infusion of Ginsberg SD, Che S, Counts SE, Mufson EJ (2006a) Shift in the ratio of
nerve growth factor in three patients with Alzheimer’s disease. Dement three-repeat tau and four-repeat tau mRNAs in individual cholinergic
Geriatr Cogn Disord 9: 246–257 basal forebrain neurons in mild cognitive impairment and Alzheimer’s
Everitt BJ, Robbins TW (1997) Central cholinergic systems and cognition. disease. J Neurochem 96: 1401–1408
Annu Rev Psychol 48: 649–684 Ginsberg SD, Che S, Counts SE, Mufson EJ (2006b) Down regulation of trk
Fagan AM, Murphy BA, Patel SN, Kilbridge JF, Mobley WC, Bu G, but not p75NTR gene expression in single cholinergic basal forebrain
Holtzman DM (1998) Evidence for normal aging of the septo-hippo- neurons mark the progression of Alzheimer’s disease. J Neurochem 97:
campal cholinergic system in apoE ( = ) mice but impaired clearance 475–487
of axonal degeneration products following injury. Exp Neurol 151: Goedert M, Jakes R (2005) Mutations causing neurodegenerative tauopa-
314–325 thies. Biochim Biophys Acta 1739: 240–250
Farber SA, Nitsch RM, Schulz JG, Wurtman RJ (1995) Regulated secretion of Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA (1989a)
beta-amyloid precursor protein in rat brain. J Neurosci 15: 7442–7451 Multiple isoforms of human microtubule-associated protein tau:
Fass U, Panickar K, Personett D, Bryan D, Williams K, Gonzales J, Sugaya sequences and localization in neurofibrillary tangles of Alzheimer’s
K, McKinney M (2000) Differential vulnerability of primary cultured disease. Neuron 3: 519–526
cholinergic neurons to nitric oxide excess. Neuro Report 11: 931–936 Goedert M, Spillantini MG, Potier MC, Ulrich J, Crowther RA (1989b)
Fibiger HC (1991) Cholinergic mechanisms in learning, memory and Cloning and sequencing of the cDNA encoding an isoform of
dementia: a review of recent evidence. Trends Neurosci 14: 220–223 microtubule-associated protein tau containing four tandem repeats:
Cholinergic transmission and Alzheimer’s disease 1639

differential expression of tau protein mRNAs in human brain. EMBO J receptor to beta-amyloid precursor protein secretion. J Neurochem
8: 393–399 71: 2094–2103
Goekoop R, Scheltens P, Barkhoh F, Rombouts SARB (2006) Cholinergic Harkany T, Abraham I, Konya C, Nyakas C, Zarandi M, Penke B, Luiten PG
challenge in Alzheimer patients and mild cognitive impairment differ- (2000) Mechanisms of b-amyloid neurotoxicity: perspectives of phar-
entially affects hippocampal activation – a pharmacological fMRI macotherapy. Rev Neurosci 11: 329–382
study. Brain 129: 141–157 Harper JD, Wong SS, Lieber CM, Lansbury PT (1997) Observation of
Gomez-Ramos P, Bouras C, Moran MA (1994) Ultrastructural localization metastable Ab amyloid protofibrils by atomic force microscopy. Chem
of butyrylcholinesterase on neurofibrillary degeneration sites in the Biol 4: 119–125
brains of aged and Alzheimer’s disease patients. Brain Res 640: 17–24 Hartley DM, Walsh DM, Ye CP, Diehl T, Vasquez S, Vassilev PM, Teplow
Gong Y, Chang L, Viola KL, Lacor PN, Lambert MP, Finch CE, Krafft GA, DB, Selkoe DJ (1999) Protofibrillar intermediates of amyloid b-protein
Klein WL (2003) Alzheimer’s disease-affected brain: presence of induce acute electrophysiological changes and progressive neurotoxi-
oligomeric Ab igands (ADDLs) suggests a molecular basis for rever- city in cortical neurons. J Neurosci 19: 8876–8884
sible memory loss. roc Natl Acad Sci USA 100: 10417–10422 Heckers S, Ohtake T, Wiley RG, Lappi DA, Geula C, Mesulam MM (1994)
Grassi F, Palma E, Tonini R, Amici M, Ballivet M, Eusebi F (2003) Amyloid Complete and selective cholinergic denervation of rat neocortex and
b(1–42) peptide alters the gating of human and mouse a-bungarotoxin- hippocampus but not amygdala by an immunotoxin against the p75
sensitive nicotinic receptors. J Physiol 547(Pt 1): 147–157 NGF receptor. J Neurosci 14: 1271–1289
Greenberg SM, Tennis MK, Brown LB, Gomez-Isla T, Hayden DL, Hefti F, Hartikka J, Eckenstein F, Gnahn H, Heumann R, Schwab M (1985)
Schoenfeld DA et al. (2000) Donezepil therapy in clinical practice: Nerve growth factor increases choline acetyltransferase but not surviv-
a randomized crossover study. Arch Neurol 57: 94–99 al or fibre outgrowth of cultured fetal septal cholinergic neurons.
Greig NH, Utsuki T, Ingram DK, Wang Y, Pepeu G, Scali C, Yu QS, Neuroscience 14: 55–68
Mamczarz J, Holloway HW, Giordano T, Chen D, Furukawa K, Heinitz K, Beck M, Schliebs R, Perez-Polo JR (2006) Cholinotoxicity
Sambamurti K, Brossi A, Lahiri DK (2005) Selective butyrylcholin- mediated by soluble oligomers of b-amyloid (1–42) differs from
esterase inhibition elevates brain acetylcholine, augments learning and cholinotoxic effects of oxidative stress as revealed by gene expression
lowers Alzheimer beta-amyloid peptide in rodent. Proc Natl Acad Sci analysis. J Neurochem 98: 1930–1945
USA 102: 17213–17218 Hellstrom-Lindahl E, Moore H, Nordberg A (2000) Increased levels of tau
Gron G, Brandenburg I, Wunderlich AP, Riepe MW (2006) Inhibition of protein in SH-SY5Y cells after treatment with cholinesterase inhibitors
hippocampal function in mild cognitive impairment: targeting the and nicotinic agonists. J Neurochem 74: 777–784
cholinergic hypothesis. Neurobio Aging 27: 78–87 Hellstrom-Lindahl E, Court J, Keverne J, Svedberg M, Lee M, Marutle A,
Grigoryan GA, Gray JA, Rashid T, Chadwick A, Hodges H (2000) Con- Thomas A, Perry E, Bednar I, Nordberg A (2004) Nicotine reduces Ab
dionally immortal neuroepithelial stem cell grafts restored patial in the brain and cerebral vessels of APPsw mice. Eur J Neurosci 19:
learning in rats with lesions at the source of cholinergic forebrain 2703–2710
projections. Restor Neurol Neurosci 17: 1 Hellweg R, Gericke CA, Jendroska K, Hartung HD, Cervos-Navarro J
Gsell W, Jungkunz G, Riederer P (2004) Functional neurochemistry of (1998) NGF content in the cerebral cortex of non-demented patients
Alzheimer’s disease. Curr Pharmacol Design 10: 265–293 with amyloid-plaques and in symptomatic Alzheimer’s disease. Int J
Gsell W, Strein I, Krause U, Riederer P (1997) Neurochemical abnor- Dev Neurosci 16: 787–794
malities in Alzheimer’s disease and Parkinson’s disease – a compara- Hock C, Maddalena A, Raschig A, Muller-Spahn F, Eschweiler G, Hager K,
tive review. J Neural Transm Suppl 51: 45–59 Heuser I, Hampel H, Muller-Thomsen T, Oertel W, Wienrich M,
Gsell W, Moll G, Sofic E, Riederer P (1993) Cholinergic and monoamin- Signorell A, Gonzalez-Agosti C, Nitsch RM (2003) Treatment with
ergic neurotransmitter systems in patients with Alzheimer’s disease the selective muscarinic m1 agonist talsaclidine decreases cerebrospi-
and senile dementia of the Alzheimer type: a critical evaluation. In: nal fluid levels of A beta 42 in patients with Alzheimer’s disease.
Maurer K (ed) Dementias – neurochemistry, neuropathology, neuroi- Amyloid 10: 1–6
maging, neuropsychology, genetics. Vieweg, Braunschweig, pp 25–51 Hoshi M, Takashima A, Murayama M, Yasutake K, Yoshida N, Ishiguro K,
Gu Q (2003) Contribution of acetylcholine to visual cortex plasticity. Hoshino T, Imahori K (1997) Nontoxic amyloid b peptide 1–42 sup-
Neurobiol Learn Mem 80: 291–301 presses acetylcholine synthesis. Possible role in cholinergic dysfunc-
Guan ZZ, Miao H, Tian JY, Unger C, Nordberg A, Zhang X (2001) tion in Alzheimer’s disease. J Biol Chem 272: 2038–2041
Suppressed expression of nicotinic acetylcholine receptors by nanomolar Hoyer S, Riederer P (2003) Pathomechanisms and hypothesis-guided
b-amyloid peptides in PC12 cells. J Neural Transm 108: 1417–1433 therapeutic strategies for late-onset Alzheimer’s disease (Germ).
Guan ZZ, Yu WF, Shan KR, Nordman T, Olsson J, Nordberg A (2003) Loss Fortschr Neurol Psychiatr 71 Suppl 1: S16–S26
of nicotinic receptors induced by b-amyloid peptides in PC12 cells: Hoyer S (2004) Glucose metabolism and insulin receptor signal transduc-
possible mechanism involving lipid peroxidation. J Neurosci Res 71: tion in Alzheimer disease. Eur J Pharmacol 490: 115–125
397–406 Hung AY, Haass C, Nitsch RM, Qiu WQ, Citron M, Wurtman RJ, Growdon
Guillozet AL, Smiley JF, Mash DC, Mesulam MM (1997) Butyrylcholin- JH, Selkoe DJ (1993) Activation of protein kinase C inhibits cellular
esterase in the life cycle of amyloid plaques. Ann Neurol 42: 909–918 production of the amyloid beta-protein. J Biol Chem 268: 22959–22962
Haass C, Selkoe DJ (1993) Cellular processing of beta-amyloid precursor Imbimbo BP (2001) Pharmacodynamic-tolerability relationships of cho-
protein and the genesis of amyloid beta-peptide. Cell 75: 1039–1042 lesterase inhibitors for Alzheimer’s disease. CNS Drugs 15: 375–390
Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer’s Inestrosa NC, Alarcon R (1998) Molecular interactions of acetylcholines-
disease: progress and problems on the road to therapeutics. Science terase with senile plaques. J Physiol (Paris) 92: 341–344
297: 353–356 Inestrosa NC, Alvarez A, Perez CA, Moreno RD, Vicente M, Linker C,
Haring R, Gurwitz D, Barg J, Pinkas-Kramarski R, Heldman E, Pittel Z, Casanueva OI, Soto Garrido J (1996) Acetylcholinesterase accelerates
Danenberg HD, Wengier A, Meshulam H, Marciano D et al. (1995) assembly of amyloid-b peptides into Alzheimer’s fibrils: possible role
NGF promotes amyloid precursor protein secretion via muscarinic of the peripheral site of the enzyme. Neuron 16: 881–891
receptor activation. Biochem Biophys Res Commun 213: 15–23 Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT (1997)
Haring R, Fisher A, Marciano D, Pittel Z, Kloog Y, Zuckerman A, Eshhar APPsw transgenic mice develop age-related Ab deposits and neuropil
N, Heldman E (1998) Mitogen-activated protein kinase-dependent abnormalities, but no neuronal loss in CA1. J Neuropathol Exp Neurol
and protein kinase C-dependent pathways link the m1 muscarinic 56: 965–973
1640 R. Schliebs, T. Arendt

Isacson O, Seo H, Lin L, Albeck D, Granholm AC (2002) Alzheimer’s Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M,
disease and Down’s syndrome: roles of APP, trophic factors and Ach. Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C,
Trends Neurosci 25: 79–84 Finch CE, Krafft GA, Klein WL (1998) Diffusible, nonfibrillar ligands
Jankowska A, Madziar B, Tomaszewicz M, Szutowicz A (2000) Acute and derived from Abeta1-42 are potent central nervous system neurotoxins.
chronic effects of aluminum on acetyl-CoA and acetylcholine meta- Proc Natl Acad Sci USA 95: 6448–6453
bolism in differentiated and nondifferentiated SN56 cholinergic cells. Lane RM, Kivipolto M, Greig NH (2004) Acetylcholinesterase and its
J Neurosci Res 62: 615–622 inhibition in Alzheimer disease. Clin Neuropharmacol 27: 141–149
Jaffar S, Counts SE, Ma SY, Dadko E, Gordon MN, Morgan D, Mufson EJ Lane RM, Potkin SG, Enz A (2006) Targeting acetylcholinesterase and
(2001) Neuropathology of mice carrying mutant APPSWE and=or butyrylcholinesterase in dementia. Int J Neuropsychopharmacol 9:
PS1 M146L transgenes: Alterations in the p75NTR cholinergic basal 101–124
forebrain septohippocampal pathway. Exp Neurol 170: 227–243 Law A, Gauthier S, Quirion R (2001) Say NO to Alzheimer’s disease: the
Jellinger KA (1991) Pathology of Parkinson’s disease. Changes other than putative links between nitric oxide and dementia of the Alzheimer’s
the nigrostriatal pathway. Mol Chem Neuropathol 14: 153–197 type. Brain Res Rev 35: 73–96
Jellinger KA (2000) Morphological substrates of mental dysfunctions in Lee DH, Wang HY (2003) Differential physiologic responses of a7
Lewy body disease: an update. J Neural Transm Suppl 59: 185–212 nicotinic acetylcholine receptors to b-amyloid 1–40 and b-amyloid
Julka D, Sandhir R, Gill KD (1995) Altered cholinergicmetabolism in rat 1–42. J Neurobiol 55: 25–30
CNS following aluminum exposure: implications on learning perfor- Lee MG, Hassani OK, Alonso A, Jones BE (2005) Cholinergic basal
mance. J Neurochem 65: 2157–2164 forebrain neuron burst with theta during waking and paradoxical sleep.
Kamal MA, Al-Jafari AA, Yu QS, Greig NH (2006) Kinetic analysis of the J Neurosci 25: 4365–4369
inhibition of human butyrylcholinesterase with cymserine. Biochim Lee VM, Balin BJ, Otvos L Jr, Trojanowski JQ (1991) A68: a major subunit
Biophys Acta 1760: 200–206 of paired helical filaments and derivatized forms of normal Tau.
Kar S, Slowikowski SP, Westaway D, Mount HT (2004) Interactions Science 251: 675–678
between b-amyloid and central cholinergic neurons: implications Lesne S, Koh MT, Kotinilek L, Kayed R, Glabe CG, Yang A, Gallagher M,
for Alzheimer’s disease. J Psychiatry Neurosci 29: 427–441 Ashe KH (2006) A specific amyloid protein assembly in the brain
Katoh-Semba R, Semba R, Takeuchi IK, Kato K (1998) Age-related impairs memory. Nature 440: 352–357
changes in levels of brain-derived neurotrophic factor in selected brain Li B, Stribley JA, Ticu A, Xie W, Schopfer LM, Hammond P, Brimijoin S,
regions of rats, normal mice and senescence-accelerated mice: a Hinrichs SH, Lockridge O (2000) Abundant tissue butyrylcholinester-
comparison to those of nerve growth factor and neurotrophin-3. ase and its possible function in the acetylcholinesterase knockout
Neurosci Res 31: 227–234 mouse. J Neurochem 75: 1320–1331
Kaufer D, Friedman A, Seidman S, Soreq H (1998) Acute stress facilitates Li X, Song L, Jope RS (1996) Cholinergic stimulation of AP-1 and NFkB
long-lasting changes in cholinergic gene expression. Nature 393: transcription factors is differentially sensitive to oxidative stress in SH-
373–377 SY5Y neuroblastoma: relationship to phosphoinositide hydrolysis.
Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, J Neurosci 16: 5914–5922
Glabe CG (2003) Common structure of soluble amyloid oligomers Li Y, Liu L, Kang J, Sheng JG, Barger SW, Mrak RE, Griffin WST (2000)
implies common mechanism of pathogenesis. Science 300: 486–489 Neuronal-glial interactions mediated by interleukin-1 enhance neuro-
Kelly JF, Furukawa K, Barger SW, Rengen MR, Mark RJ, Blanc EM, nal acetylcholinesterase activity and mRNA expression. J Neurosci 20:
Roth GS, Mattson MP (1996) Amyloid b-peptide disrupts carbachol- 149–155
induced muscarinic cholinergic signal transduction in cortical neurons. Lin L, Georgievska B, Mattsson A, Isacson O (1999) Cognitive changes and
Proc Natl Acad Sci USA 93: 6753–6758 modified processing of amyloid precursor protein in the cortical and
Kirkitadze MD, Condron MM, Teplow DB (2001) Identification and hippocampal system after cholinergic synapse loss and muscarinic
characterization of key kinetic intermediates in amyloid b-protein receptor activation. Proc Natl Acad Sci USA 96: 12108–12113
fibrillogenesis. J Mol Biol 312: 1103–1119 Liskowsky W, Schliebs R (2006) Muscarinic acetylcholine receptor inhibi-
Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM (2005) tion in transgenic Alzheimer-like Tg2576 mice by scopolamine
Lipopolysaccharide-induced inflammation exacerbates tau pathology favours the amyloidogenic route of processing of amyloid precursor
by a cyclin-dependent kinase 5-mediated pathway in a transgenic protein. Int J Devl Neurosci 24: 149–156
model of Alzheimer’s disease. J Neurosci 25: 8843–8853 Liu Q, Kawai H, Berg DK (2001) b-amyloid peptide blocks the response of
Klein AM, Kowall NW, Ferrante RJ (1999) Neurotoxicity and oxidative alpha7-containing nicotinic receptors on hippocampal neurons. Proc
damage of b amyloid 1–42 versus b amyloid 1–40 in the mouse Natl Acad Sci USA 98: 4734–4739
cerebral cortex. Ann NY Acad Sci 893: 314–320 Lorenzo A, Yankner BA (1994) b-Amyloid neurotoxicity requires fibril
Kosik KS, Joachim CL, Selkoe DJ (1986) Microtubule-associated protein formation and is inhibited by congo red. Proc Natl Acad Sci USA 91:
tau (tau) is a major antigenic component of paired helical filaments in 12243–12247
Alzheimer disease. Proc Natl Acad Sci USA 83: 4044–4048 Lucas-Meunier E, Fossier P, Baux G, Amar M (2003) Cholinergic
Kuhl DE, Koeppe RA, Snyder SE, Minoshima S, Frey KA, Kilbourn MR modulation of the cortical neuronal network. Pflugers Arch 446:
(2006) In vivo butyrylcholinesterase activity is not increased in 17–29
Alzheimer’s disease synapses. Ann Neurol 59: 13–20 Luth HJ, Ogunlade V, Kuhla B, Kientsch-Engel R, Stahl P, Webster J,
Kuner P, Schubenel R, Hertel C (1998) b-amyloid binds to p57NTR and Arendt T, Munch G (2005) Age- and stage-dependent accumulation of
activates NFkB in human neuroblastoma cells. J Neurosci Res 54: advanced glycation end products in intracellular deposits in normal and
798–804 Alzheimer’s disease brains. Cereb Cortex 15: 211–220
Lahiri DK, Farlow MR, Nurnberger JJ Jr, Greig NH (1997) Effects of Malo M, Diebler MF, Prado de Carvalho L, Meunier FM, Dunant Y, Bloc A,
cholinesterase inhibitors on the secretion of beta-amyloid precursor Stinnakre J, Tomasi M, Tchelingerian J, Couraud PO, Israel M (1999)
protein in cell cultures. Ann NY Acad Sci 826: 416–421 Evoked acetylcholine release by immortalized brain endothelial cells
Lahiri DK, Utsuki T, Chen D, Farlow MR, Shoaib M, Ingram DK, Greig NH genetically modified to express choline acetyltransferase and=or the
(2002) Nicotine reduces the secretion of Alzheimer’s b-amyloid pre- vesicular acetylcholine transporter. J Neurochem 73: 1483–1491
cursor protein containing b-amyloid peptide in the rat without altering Martinez-Murillo R, Rodrigo J (1995) The localization of cholinergic
synaptic proteins. Ann NY Acad Sci 965: 364–372 neurons andmarkers in the CNS. In: Stone TW (ed) CNS Neurotrans-
Cholinergic transmission and Alzheimer’s disease 1641

mitters and neuromodulators: Acetylcholine. CRC Press, Boca Raton, transgenic Tg2576 mice with amyloid plaque pathology. J Neurochem
pp 1–38 86: 283–289
Martinez-Serrano A, Bj€orklund A (1998) Ex vivo growth factor gene M€unch G, Gerlach M, Sian J, Wong A, Riederer P (1998) Advanced
transfer to the basal forebrain in presymptomatic middle-aged rats glycation end products in neurodegeneration: more than early markers
prevents the development of cholinergic neuron atrophy and cognitive of oxidative stress? Ann Neurol 44(3) Suppl 1: S85–S88
impairment during aging. Proc Natl Acad Sci USA 95: 1858–1863 M€unch G, Mayer S, Michaelis J, Hipkiss AR, Riederer P, M€uller R,
Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Neumann A, Schinzel R, Cunningham AM (1997a) Influence of
Beyreuther K (1985) Amyloid plaque core protein in Alzheimer advanced glycation end-products and AGE-inhibitors on nucleation-
disease and Down syndrome. Proc Natl Acad Sci USA 82: 4245–4249 dependent polymerization of amyloid peptide. Biochem et Biophys
McKinney M (2005) Brain cholinergic vulnerability: relevance to behavior Acta 1360: 17–19
and disease. Biochem Pharmacol 70: 1115–1124 M€unch G, Thome J, Foley P, Schinzel R, Riederer P (1997b) Advanced
McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, glycation endproducts in ageing and Alzheimer’s disease. Brain Res
Bush AI, Masters CL (1999) Soluble pool of Ab amyloid as a Rev 23: 134–143
determinant of severity of neurodegeneration in Alzheimer’s disease. M€unch G, Taneli Y, Schraven E, Schindler U, Schinzel R, Palm D, Riederer
Ann Neurol 46: 860–866 P (1994) The cognition-enhancing drug tenilsetam is an inhibitor of
Mehlhorn G, Loffler T, Apelt J, Rossner S, Urabe T, Hattori N, Nagamatsu protein crosslinking by advanced glycosylation. J Neural Transm [P-D
S, Bigl V, Schliebs R (1998) Glucose metabolism in cholinoceptive Sect] 8: 193–208
cortical rat brain regions after basal forebrain cholinergic lesion. Int J Murase K, Nabeshima T, Robitaille Y, Quirion R, Ogawa M, Hayashi K
Dev Neurosci 16: 675–690 (1993) NGF level of is not decreased in the serum, brain-spinal fluid,
Mesulam MM (1986) Alzheimer plaques and cortical cholinergic innerva- hippocampus, or parietal cortex of individuals with Alzheimer’s dis-
tion. Neuroscience 17: 275–276 ease. Biochem Biophys Res Commun 193: 198–203
Mesulam MM (2004) The cholinergic lesion of Alzheimer’s disease: Narisawa-Saito M, Wakabayashi K, Tsuji S, Takahashi H, Nawa H (1996)
pivotal factor or side show. Learn Mem 11: 43–49 Regional specificity of alterations in NGF, BDNF and NT-3 levels in
Mesulam MM, Geula C (1994) Butyrylcholinesterase reactivity differenti- Alzheimer’s disease. Neuroreport 7: 2925–2928
ates the amyloid plaques of aging from those of dementia. Ann Neurol Nitsch RM, Deng M, Tennis M, Schoenfeld D, Growdon JH (2002) The
36: 722–727 selective muscarinic agonist AF102B decreases levels of total Ab in
Mesulam MM, Guillozet A, Shaw P, Levey A, Duysen EG, Lockridge O CSF of patients with Alzheimer’s disease. Ann Neurol 48: 913–918
(2002a) Acetylcholinesterase knockouts establish central cholinergic Nitsch RM, Farber SA, Growdon JH, Wurtman RJ (1993) Release of
pathways and can use butyrylcholinesterase to hydrolyze acetylcho- amyloid beta-protein precursor derivatives by electrical depolarization
line. Neuroscience 110: 627–639 of rat hippocampal slices. Proc Natl Acad Sci USA 90: 5191–5193
Mesulam M, Guillozet A, Shaw P, Quinn B (2002b) Widely spread Nitsch RM, Slack BE, Wurtman RJ, Growdon JH (1992) Release of
butyrylcholinesterase can hydrolyze acetylcholine in the normal and Alzheimer amyloid precursor derivatives stimulated by activation of
Alzheimer brain. Neurobiol Dis 9: 88–93 muscarinic acetylcholine receptors. Science 258: 304–307
Mohs RC, Doody RS, Morris JC, Jeni JR, Rogers SL, Perdomo CA et al. Nordberg A (1992) Neuroreceptor changes in Alzheimer disease. Cere-
(2001) A 1-year, placebo-controlled preservation of function-survival brovasc Brain Met Rev 4: 303–328
study of donezepil in AD patients. Neurology 57: 481–488 Nordberg A (2001) Nicotinic receptor abnormalities of Alzheimer’s dis-
Molchan SE, Martinez RA, Hill JL, Weingartner HJ, Thompson K, Vitiello ease: therapeutic implications. Biol Psychiatry 49: 200–210
B, Sunderland T (1992) Increased cognitive sensitivity to scopolamine Nordberg A (2006) Mechanisms behind the neuroprotective actions of
with age and a perspective on the scopolamine model. Brain Res Rev cholinesterase inhibitors in Alzheimer disease. Alzheimer Dis Assoc
17: 215–226 Disord 20 Suppl 2: S12–S18
Moll G, Gsell W, Wichart I, Jellinger K, Riederer P (1990) Cholinergic and Nordberg A, Hellstrom-Lindahl E, Lee M, Johnson M, Mousavi M, Hall R,
monoaminergic neuromediator systems in DAT. Neuropathological Perry E, Bednar I, Court J (2002) Chronic nicotine treatment reduces
and neurochemical findings. In: Maurer K, Riederer P, Beckmann H b-amyloidosis in the brain of mouse model of Alzheimer’s disease
(eds) Alzheimer’s disease. Epidemiology, neuropathology, neuro- (APPsw). J Neurochem 81: 655–658
chemistry and clinics. Key Topics in Brain Research. Springer Wien Oddo S, LaFerla FM (2006) The role of nicotinic acetylcholine receptors in
New York, pp 235–243 Alzheimer’s disease. J Physiol (Paris) 99: 172–179
Moran MA, Mufson EJ, Gomez-Ramos P (1993) Colocalization of cho- Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R,
linesterases with b-amyloid protein in aged and Alzheimer’s brain. Metherate R, Mattson MP, Akbari Y, LaFerla FM (2003) Triple-
Acta Neuropathol 85: 362–369 transgenic model of Alzheimer’s disease with plaques and tangles:
Mori F, Lai CC, Fusi F, Giacobini E (1995) Cholinesterase inhibitors intracellular Abeta and synaptic dysfunction. Neuron 39: 409–421
increase secretion of APPs in rat brain cortex. Neuroreport 6: 633–636 Oddo S, Caccamo A, Green KN, Liang K, Tran L, Chen Y, Leslie FM,
Mount C, Downtown C (2006) Alzheimer’s disease: progress or profit? Nat LaFerla FM (2005) Chronic nicotine administration exacerbates tau
Med 12: 780–784 pathology in a transgenic model of Alzheimer’s disease. Proc Natl
Mufson EJ, Ma SY, Dills J, Cochran EJ, Leurgans S, Wuu J, Bennett DA, Acad Sci USA 102: 3046–3051
Jaffar S, Gilmor ML, Levey AI, Kordower JH (2002) Loss of basal Olesen OF, Dago L, Mikkelsen JD (1998) Amyloid beta neurotoxicity in
forebrain P75(NTR) immunoreactivity in subjects with mild cognitive cholinergic but not in serotoninergic phenotype of RN46A cells. Brain
impairment and Alzheimer’s disease. J Comp Neurol 443: 136–153 Res Mol Brain Res 57: 266–274
Mufson EJ, Ginsberg SD, Ikonomovic MD, DeKosky ST (2003) Human Ono K, Hasegawa K, Yamada M, Naiki H (2002) Nicotine breaks down
cholinergic basal forebrain: chemoanatomy and neurologic dysfunc- preformed Alzheimer’s b-amyloid fibrils in vitro. Biol Psychiatry 52:
tion. J Chem Neuroanat 26: 233–242 880–886
M€
uller DM, Mendla K, Farber SA, Nitsch RM (1997) Muscarinic M1 Pedersen WA, Kloczewiak MA, Blusztajn JK (1996) Amyloid b-protein
receptor agonists increase the secretion of the amyloid precursor reduces acetylcholine synthesis in a cell line derived from cholinergic
protein ectodomain. Life Sci 60: 985–991 neurons of the basal forebrain. Proc Natl Acad Sci USA 93: 8068–8071
M€
unch G, Apelt J, Kientsch-Engel R, Stahl P, L€uth HJ, Schliebs R (2003) Perini G, Della-Bianca V, Politi V, Della-Valle G, Dal-Pra I, Rossi F,
Advanced glycation endproducts and pro-inflammatory cytokines in Armato U (2002) Role of p75 neurotrophin receptor in the neurotoxi-
1642 R. Schliebs, T. Arendt

city by b-amyloid peptides and synergistic effect of inflammatory cells on cholinergic basal forebrain neurons after partial immunole-
cytokines. J Exp Med 195: 907–918 sion. J Neurosci Res 45: 40–56
Perry EK, Gibson PH, Blessed G, Perry RH, Tomlinson BE (1977a) Roßner S, Ueberham U, Schliebs R, Perez-Polo JR, Bigl V (1998a) The
Neurotransmitter enzyme abnormalities in senile dementia. Choline regulation of amyloid precursor protein metabolism by cholinergic
acetyltransferase and glutamic acid decarboxylase activities in mechanisms and neurotrophin receptor signaling. Prog Neurobiol 56:
necropsy brain tissue. J Neurol Sci 34: 247–265 541–569
Perry EK, Perry RH, Blessed G, Tomlinson BE (1977b) Necropsy evidence Roßner S, Ueberham U, Schliebs R, Perez-Polo JR, Bigl V (1998b)
of central cholinergic deficits in senile dementia. Lancet 1: 189 Neurotrophin binding to the p75 neurotrophin receptor is necessary
Perry EK, Perry RH, Blessed G, Tomlinson BE (1978) Changes in brain but not sufficient to mediate NGF-effects on APP secretion in PC-12
cholinesterases in senile dementia of Alzheimer type. Neuropathol cells. J Neural Transm Suppl 53: 279–285
Appl Neurobiol 4: 273–277 Rubio A, Perez M, Avila J (2006) Acetylcholine receptors and tau phos-
Perry E, McKeith I, Ballard C (2003) Butyrylcholinesterase and progres- phorylation. Curr Mol Med 6: 423–428
sion of cognitive deficits in dementia with Lewy bodies. Neurology 60: Saez-Valero J, Fodero LR, White AR, Barrow CJ, Small DH (2003)
1852–1853 Acetylcholinesterase is increased in mouse neuronal and astrocyte
Pettit DL, Shao Z, Yakel JL (2001) b-amyloid (1–42) peptide directly cultures after treatment with b-amyloid peptides. Brain Res 965:
modulates nicotinic receptors in the rat hippocampal slice. J Neurosci 283–286
21: RC120 Salehi A, Ocampo M, Verhaagen J, Swaab DF (2000) P75 neurotrophin
Pike CJ, Walencewicz AJ, Glabe CG, Cotman CW (1991) In vitro aging of receptor in the nucleus basalis of meynert in relation to age, sex, and
b-amyloid protein causes peptide aggregation and neurotoxicity. Brain Alzheimer’s disease. Exp Neurol 161: 245–258
Res 563: 311–314 Salehi A, Delcroix JD, Swaab DF (2004) Alzheimer’s disease and NGF
Platt B, Fiddler G, Riedel G, Henderson Z (2001) Aluminium toxicity in the signaling. J Neural Transm 111: 323–345
rat brain: histochemical and immunocytochemical evidence. Brain Res Salmond CH, Chatfield DA, Menon DK, Pickard JD, Sahakian BJ (2005)
Bull 55: 257–267 Cognitive sequelae of head injury: involvement of basal forebrain and
Puolivali J, Miettinen R, Pradier L, Riekkinen P Jr (2000) Apolipoprotein E- associated structures. Brain 128: 189–200
deficient mice are not more susceptible to biochemical and memory Salomon AR, Marcinowski KJ, Friedland RP, Zagorski MG (1996) Nico-
deficits induced by nucleus basalis lesion. Neuroscience 96: 291–297 tine inhibits amyloid formation by the b-peptide. Biochemistry 35:
Rabizadeh S, Bitler CM, Butcher LL, Bredesen DE (1994) Expression of 13568–13578
the low-affinity nerve growth factor receptor enhances b-amyloid Sato A, Sato Y, Uchida S (2004) Activation of the intracerebral cholinergic
peptide toxicity. Proc Natl Acad Sci USA 91: 10703–10706 nerve fibers originating in the basal forebrain increases regional
Racchi M, Govoni S (2003) The pharmacology of amyloid precursor cerebral blood flow in the rat’s cortex and hippocampus. Neurosci
protein processing. Exp Gerontol 38: 145–157 Lett 361: 90–93
Racchi M, Mazzucchelli M, Pascale A, Sironi M, Govoni S (2003) Role of Sarter M, Bruno JP (1997) Cognitive functions of cortical acetylcholine:
protein kinase Calpha in the regulated secretion of the amyloid toward a unifying hypothesis. Brain Res Brain Res Rev 23: 28–46
precursor protein. Mol Psychiatry 8: 209–216 Sarter M, Bruno JP (2004) Developmental origins of the age-related decline
Racchi M, Sironi M, Caprera A, Konig G, Govoni S (2001) Short- and long- in cortical cholinergic function and associated cognitive abilities.
term effect of acetylcholinesterase inhibition on the expression and Neurobiol Aging 25: 1127–1139
metabolism of the amyloid precursor protein. Mol Psychiatry 6: 520–528 Sberna G, Saez-Valero J, Beyreuther K, Masters CL, Small DH (1997) The
Rahimi O, Juliano SL (2001) Transplants of NGF-secreting fibroblasts amyloid b-protein of Alzheimer’s disease increases acetylcholinester-
restore stimulus-evoked activity in barrel cortex of basal-forebrain- ase expression by increasing intracellular calcium in embryonal car-
lesioned rats. J Neurophysiol 86: 2081–2096 cinoma P19 cells. J Neurochem 69: 1177–1184
Rees T, Hammond PI, Soreq H, Younkin S, Brimijoin S (2003) Acetyl- Schatz CR, Geula C, Mesulam M (1990) Competitive substrate inhibition in
cholinesterase promotes beta-amyloid plaques in cerebral cortex. the histochemistry of cholinesterase activity in Alzheimer’s disease.
Neurobiol Aging 24: 777–787 Neurosci Lett 117: 56–61
Reyes AE, Chacon MA, Dinamarca MC, Cerpa W, Morgan C, Inestrosa NC Schliebs R, Bigl V, Biesold D (1982) Development of muscarinic choli-
(2004) Acetylcholinesterase-Ab complexes are more toxic than Abeta nergic receptor binding in the visual system of monocularly deprived
fibrils in rat hippocampus: effect on rat beta-amyloid aggregation, and dark reared rats. Neurochem Res 7: 1181–1198
laminin expression, reactive astrocytosis, and neuronal cell loss. Am J Schliebs R, Bigl V (1996) Animal models to produce cortical cholinergic
Pathol 164: 2163–2174 dysfunction. In: Conn PM, Perez-Polo JR (eds) Methods in neuros-
Reyes AE, Perez DR, Alvarez A, Garrido J, Gentry MK, Doctor BP, ciences, Vol. 30. Paradigms of neural injury. Academic Press, San
Inestrosa NC (1997) A monoclonal antibody against acetylcholines- Diego, pp 275–289
terase inhibits the formation of amyloid fibrils induced by the enzyme. Schliebs R, Roßner S, Bigl V (1996) Immunolesion by 192IgG-saporin of
Biochem Biophys Res Commun 232: 652–655 rat basal forebrain cholinergic system – a useful tool to produce
Riederer P, Hoyer S (2005) Disturbances of neurotransmission in dementia cortical cholinergic dysfunction. Progr Brain Res 109: 253–264
(Germ). In: Wallesch CW, F€orstl H (eds) Demenzen. Thieme, Stuttgart, Schliebs R (2005a) Basal forebrain cholinergic lesion by 192IgG-saporin:
pp 27–43 a tool to assess the consequences of cortical cholinergic dysfunc-
Roßner S, Kumar A, Witzemann V, Schliebs R (1994) Development of tion in Alzheimer’s disease. In: Wiley RG, Lappi DA (eds) Molec-
laminar expression of the m2 muscarinic cholinergic receptor gene in ular neurosurgery with targeted toxins. Humana Press, Totowa,
rat visual cortex and the effect of monocular visual deprivation. Dev pp 59–86
Brain Res 77: 55–61. Erratum in: Dev Brain Res 78: 796–797 Schliebs R (2005b) Basal forebrain cholinergic dysfunction in Alzheimer’s
Roßner S, Schliebs R, H€artig W, Bigl V (1995) 192IgG-saporin-induced disease – interrelationship with b-amyloid, inflammation and neuro-
selective lesion of cholinergic basal forebrain system: neurochemical trophin signaling. Neurochem Res 30: 895–908
effects on cholinergic neurotransmission in rat cerebral cortex and Schliebs R, Heidel K, Apelt J, Gniezdzinska M, Kirazov L, Szutowicz A
hippocampus. Brain Res Bull 38: 371–381 (2006) Interaction of interleukin-1b with muscarinic acetylcholine
Roßner S, Yu J, Pizzo K, Werrbach-Perez K, Schliebs R, Bigl V, Perez-Polo receptor-mediated signaling cascade in cholinergically differentiated
JR (1996) Effects of intraventricular transplantation of NGF-secreting SH-SY5Y cells. Brain Res, accepted
Cholinergic transmission and Alzheimer’s disease 1643

Selkoe DJ (2002) Alzheimer’s disease is a synaptic failure. Science 298: secretion of the Alzheimer’s amyloid b-protein precursor that contains
789–791 amyloid b-peptide in rat. J Alzheimers Dis 4: 405–415
Selkoe DJ (2003) Aging, amyloid, and Alzheimer’s disease: a perspective in Villa A, Larasa MJ, Pascual A (2001) Nerve growth factor modulates the
honor of Carl Cotman. Neurochem Res 28: 1705–1713 expression and secretion of b-amyloid precursor protein through
Selkoe DJ, Abraham CR, Podlisny MB, Duffy LK (1986) Isolation of low- different mechanisms in PC12 cells. J Neurochem 77: 1077–1084
molecular-weight proteins from amyloid plaque fibers in Alzheimer’s Walland A, Burkard S, Hammer R, Troger W (1997) In vivo consequences
disease. J Neurochem 46: 1820–1834 of M1-receptor activation by talsaclidine. Life Sci 60: 977–984
Seo H, Ferree AW, Isacson O (2002) Cortico-hippocampal APP and NGF Walch C, Schliebs R, Bigl V (1989) Effect of early visual pattern depriva-
levels are dynamically altered by cholinergic muscarinic antagonist or tion on development and lamina distribution of cholinergic markers in
M1 agonist treatment in normal mice. Eur J Neurosci 15: 498–505 rat visual cortex. EXS 57: 295–304
Seo J, Kim S, Kim H, Park CH, Jeong S, Lee J, Choi SH, Chang K, Rah J, Walsh DM, Hartley DM, Kusumoto Y, Fezoui Y, Condron MM, Lomakin A,
Koo J, Kim E, Suh Y (2001) Effects of nicotine on APP secretion and Benedek GB, Selkoe DJ, Teplow DB (1999) Amyloid b-protein
Ab- or CT(105)-induced toxicity. Biol Psychiatry 49: 240–247 fibrillogenesis. Structure and biological activity of protofibrillar inter-
Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, mediates. J Biol Chem 274: 25945–25952
McKay DM, Tintner R, Frangione B (1992) Production of the Alz- Walsh DM, Klyubin I, Fadeeva JV, Rowan MJ, Selkoe DJ (2002a) Amyloid-
heimer amyloid beta protein by normal proteolytic processing. Science b oligomers: their production, toxicity and therapeutic inhibition.
258: 126–129 Biochem Soc Trans 30: 552–557
Slack BE (2000) The m3 muscarinic acetylcholine receptor is coupled to Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS,
mitogen-activated protein kinase via protein kinase C and epidermal Rowan MJ, Selkoe DJ (2002b) Naturally secreted oligomers of amy-
growth factor receptor kinase. Biochem J 348 (Pt 2): 381–387 loid b protein potently inhibit hippocampal longterm potentiation in
Slack BE, Breu J, Petryniak MA, Srivastava K, Wurtman RJ (1995) vivo. Nature 416: 535–539
Tyrosine phosphorylation-dependent stimulation of amyloid precursor Wang HY, Lee DH, D’Andrea MR, Peterson PA, Shank RP, Reitz AB
protein secretion by the m3 muscarinic acetylcholine receptor. J Biol (2000a) b-Amyloid (1–42) binds to a7 nicotinic acetylcholine receptor
Chem 270: 8337–8344 with high affinity. Implications for Alzheimer’s disease pathology.
Sofic E, Burger R, Riederer P, Beckmann H, Heinsen H, Hebenstreit G, J Biol Chem 275: 5626–5632
Maurer K (1989) The reflection of multiple biochemical deficits in Wang HY, Lee DH, Davis CB, Shank RP (2000b) Amyloid peptide Abeta
nucleus basalis of Meynert of Alzheimer brains to differential scanning (1–42) binds selectively and with picomolar affinity to alpha7 nicotinic
thermograms. J Neural Transm P-D Sect 1(1–2): 134 acetylcholine receptors. J Neurochem 75: 1155–1161
Sofroniew MV, Howe CL, Mobley WC (2001) Nerve growth factor Wang C, Wurtman RJ, Lee RK (2000c) Amyloid precursor protein and
signaling, neuroprotection, and neural repair. Annu Rev Neurosci membrane phospholipids in primary cortical neurons increase with
24: 1217–1281 development, or after exposure to nerve growth factor or Ab (1–40).
Szutowicz A (2001) Aluminium, NO, and nerve growth factor neurotoxicity Brain Res 865: 157–167
in cholinergic neurons. J Neurosci Res 66: 1009–1018 Wang HW, Pasternak JF, Kuo H, Ristic H, Lambert MP, Chromy B,
Szutowicz A, Bielarczyk H, Gul S, Ronowska A, Pawelczyk T, Jankowska- Viola KL, Klein WL, Stine WB, Krafft GA, Trommer BL (2002)
Kulawy A (2006) Phenotype-dependent suceptibility of cholinergic Soluble oligomers of b amyloid (1–42) inhibit long-term potentia-
neuroblastoma cells to neurotoxic inputs. Metab Brain Dis, published tion but not long-term depression in rat dentate gyrus. Brain Res
online 924: 133–140
Tasker A, Perry EK, Ballard CG (2005) Butyrylcholinesterase: impact on Wang HY, Li W, Benedetti NJ, Lee DH (2003) a7 nicotinic acetylcholine
symptoms and progression of cognitive impairment. Expert Rev receptors mediate b-amyloid peptide-induced tau protein phosphor-
Neurother 5: 101–106 ylation. J Biol Chem 278: 31547–31553
Taylor BM, Sarver RW, Fici G, Poorman RA, Lutzke BS, Molinari A, Wenk GL (1993) A primate model of Alzheimer’s disease. Behav Brain Res
Kawabe T, Kappenman K, Buhl AE, Epps DE (2003) Spontaneous 57: 117–122
aggregation and cytotoxicity of the bamyloid Ab1-40: a kinetic model. Wenk GL, McGann K, Mencarelli A, Hauss-Wegrzyniak B, DelSoldato P,
J Protein Chem 22: 31–40 Fiorucci S (2000) Mechanisms to prevent the toxicity of chronic
Teaktong T, Graham AJ, Court JA, Perry RH, Jaros E, Johnson M, Hall R, neuroinflammation on forebrain cholinergic neurons. Eur J Pharmacol
Perry EK (2004) Nicotinic acetylcholine receptor immunohistochem- 402: 77–85
istry in Alzheimer’s disease and dementia with Lewy bodies; differ- Westerman MA, Cooper-Blacketer D, Mariash A, Kotilinek L, Kawara-
ential and astroglial pathology. J Neurol Sci 225: 39–49 bayashi T, Younkin LH, Carlson GA, Younkin SG, Ashe KH (2002)
Teplow DB (1998) Structural and kinetic features of amyloid b-protein The relationship between Ab and memory in the Tg2576 mouse model
fibrillogenesis. Amyloid 5: 121–142 of Alzheimer’s disease. J Neurosci 22: 1858–1867
Terry RD, Buccafusco JJ (2003) The cholinergic hypothesis of age and Whitehouse PJ, Hedreen JC, White CL 3rd, Price DL (1983) Basal
Alzheimer’s disease-realted cognitive deficits: recent challenges and forebrain neurons in the dementia of Parkinson disease. Ann Neurol
their implications for novel drug development. J Pharmacol Exp Ther 13: 243–248
306: 821–827 Wilcox BJ, Applegate MD, Portera-Cailliau C, Koliatsos VE (1995) Nerve
Toth K, Borhegyi Z, Freund TF (1993) Postsynaptic targets of GABAergic growth factor prevents apoptotic cell death in injured central cholin-
hippocampal neurons in the medial septum-diagonal band of broca ergic neurons. J Comp Neurol 359: 573–585
complex. J Neurosci 13: 3712–3724 Wiley RG, Oeltmann TN, Lappi DA (1991) Immunolesioning: selective
Tozaki H, Matsumoto A, KannoT, Nagai K, Nagata T, Yamamoto I, destruction of neurons using immunotoxin to rat NGF receptor. Brain
Nishizaki T (2002) The inhibitory and facilitatory actions of amy- Res 562: 149–153
loid-beta peptides on nicotinic Ach receptors and AMPA receptors. Wilkinson DG (1999) The pharmacology of donezepil: a new treatment of
Biochem Biophys Res Commun 294: 42–45 Alzheimer’s disease. Expert Opin Pharmacother 1: 121–135
Tuszynski MH (2000) Intraparenchymal NGF infusions rescue degenerat- Williams LR, Varon S, Peterson GM, Wictorin K, Fischer W, Bjorklund A,
ing cholinergic neurons. Cell Transplant 9: 629–636 Gage FH (1986) Continuous infusion of nerve growth factor prevents
Utsuki T, Shoaib M, Holloway HW, Ingram DK, Wallace WC, Haroutunian basal forebrain neuronal death after fimbria fornix transection. Proc
V, Sambamurti K, Lahiri DK, Greig NH (2002) Nicotine lowers the Natl Acad Sci USA 83: 9231–9235
1644 R. Schliebs, T. Arendt: Cholinergic transmission and Alzheimer’s disease

Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G et al. (2001) A mice lacking acetylcholinesterase. J Pharmacol Exp Ther 293:
1-year, randomized, placebo-controlled study of donezepil in patients 896–902
with mild to moderate AD. Neurology 57: 489–495 Yaar M, Zhai S, Fine RE, Eisenhauer PB, Arble BL, Stewart KB, Gilchrest
Winkler J, Ramirez GA, Thal LJ, Waite JJ (2000) Nerve growth factor BA (2002) Amyloid b binds to trimers as well as monomers of the 75-
(NGF) augments cortical and hippocampal cholinergic functioning kDa neurotrophin receptor and activates receptor signaling. J Biol
after p75NGF receptor-mediated deafferentation but impairs inhibi- Chem 277: 7720–7725
tory avoidance and induces fear-related behaviors. J Neurosci 20: Yan Z, Feng J (2004) Alzheimer’s disease: interactions between cholinergic
834–844 functions and b-amyloid. Curr Alzheimer Res 1: 241–248
Woolf NJ (1997) A possible role for cholinergic neurons of the basal Zhang X (2004) Cholinergic activity and amyloid precursor protein proces-
forebrain and pontomesencephalon in consciousness. Conscious Cogn sing in aging and Alzheimer’s disease. Curr Drug Targets 3: 137–152
6: 574–596 Zhang Y, Hong Y, Bounhar Y, Blacker M, Roucou X, Tounekti O, Vereker
Wright CI, Geula C, Mesulam MM (1993) Neurological cholinesterases E, Bowers WJ, Federoff HJ, Goodyer CG, LeBlanc A (2003) p75
in the normal brain and in Alzheimer’s disease: relationship to neurotrophin receptor protects primary cultures of human neurons
plaques, tangles, and patterns of selective vulnerability. Ann Neurol against extracellular amyloid beta peptide cytotoxicity. J Neurosci 23:
34: 373–384 7385–7394
Xie W, Stribley JA, Chatonnet A, Wilder PJ, Rizzino A, McComb RD, Z€uchner T, Perez-Polo JR, Schliebs R (2004) b-secretase BACE1 is
Taylor P, Hinrichs SH, Lockridge O (2000) Postnatal developmental differentially controlled through muscarinic acetylcholine receptor
delay and supersensitivity to organophosphate in gene-targeted signaling. J Neurosci Res 77: 250–257

Potrebbero piacerti anche