You are on page 1of 5

Neuroscience Letters 463 (2009) 93–97

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Age-dependent changes in noradrenergic locus coeruleus system in


wild-type and APP23 transgenic mice
Patricia Szot a,1 , Debby Van Dam b,1 , Sylvia S. White a , Allyn Franklin a ,
Matthias Staufenbiel c , Peter Paul De Deyn b,d,∗
a
Northwest Network for Mental Illness Research, Education, and Clinical Center, Veterans Administration Puget Sound Health Care System,
and Department of Psychiatry and Behavioral Science, University of Washington, Seattle 98195, USA
b
Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp,
Universiteitsplein 1, B-2610 Wilrijk, Belgium
c
Novartis Institutes for Biomedical Research Basel, Basel, Switzerland
d
Department of Neurology – Memory Clinic, Middelheim General Hospital, ZNA, Lindendreef 1, B-2020 Antwerp, Belgium

a r t i c l e i n f o a b s t r a c t

Article history: Alzheimer’s disease (AD), a neurodegenerative disorder, is characterized by the loss of neurons in specific
Received 2 April 2009 regions of the CNS including the locus coeruleus (LC), the major noradrenergic locus in the CNS. Several
Received in revised form 17 July 2009 animal models of AD have been developed that exhibit some of the pathophysiological changes in the
Accepted 17 July 2009
CNS that are observed in AD patients. The purpose of this study was to determine if the integrity of the
LC noradrenergic system is altered in the amyloid precursor protein 23 (APP23) mouse model of AD at
Keywords:
the age of 3, 6 and 12 months through quantification of tyrosine hydroxylase (TH) mRNA expression.
APP23 transgenic
Despite a previous study suggesting alterations in the noradrenergic transmission system of APP23 mice,
Norepinephrine
Locus coeruleus
the current study failed to show altered TH-positive neuronal numbers or expression in LC noradrenergic
Tyrosine hydroxylase neurons of APP23 mice versus wild-type (WT) littermates. However, the present study did demonstrate
Aging an age-dependent effect on TH mRNA expression. Both the number of TH-containing neurons and the
Alzheimer’s disease amount of TH-positive grains/neuron significantly increased between the age of 3 and 6 months with no
difference between 6 and 12 months. These observations indicate that any study comparing the nora-
drenergic system between WT (C57Bl/6) and experimental mice must strictly choose the age to be tested
and limit age differences between control and experimental groups to the absolute minimum. More
importantly, when long-term therapeutic interventions targeting the noradrenergic system are applied
to mouse models, and related parameters are studied longitudinally, care should be taken to distinguish
between potential therapeutic and strain-specific developmental or age-related alterations.
© 2009 Elsevier Ireland Ltd. All rights reserved.

Alzheimer’s disease (AD), a neurodegenerative disorder, is char- important to determine how noradrenergic neuronal loss with
acterized by cognitive impairment and the loss of neurons in compensatory changes affects memory.
the cortex and hippocampus. The locus coeruleus (LC), the major Several different animal models of AD have been developed uti-
noradrenergic locus in the CNS, also demonstrates substan- lizing the genetic information obtained from human AD studies
tial loss of neurons in AD [4,7,13,23,25,40,44]. Interestingly, the [12,50]. These various animal models represent a powerful tool
surviving neurons in the LC demonstrate many compensatory in describing time-linked pathophysiological mechanisms in the
changes in response to the neuronal loss [1,10,11,16,19,24,28,29, CNS that are often not accessible in patients [50], and allow for the
32–34,40,41,43,44]. Because the noradrenergic system has been development of drugs for treating these pathological changes asso-
shown to play a major role in learning and memory [15], it is ciated with dementia. The amyloid precursor protein 23 (APP23)
mouse harbors the Swedish double mutation, which is known to
cause early-onset familial AD in humans [8,26,36]. The neuron-
specific promoter and the integration of the human (APP) carrying
∗ Corresponding author at: Laboratory of Neurochemistry and Behaviour, Institute the Swedish double mutation into the mouse genome lead to a
Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Univer- seven-fold overexpression of the human APP (hAPP) compared to
siteitsplein 1, B-2610 Wilrijk, Belgium. Tel.: +32 3 820 26 20;
the endogenous mRNA level. The first amyloid plaques appear in the
fax: +32 3 820 26 18.
E-mail addresses: peter.dedeyn@ua.ac.be, dedeyn@skynet.be (P.P. De Deyn). frontal cortex and the subiculum at the age of 6 months, to grow in
1
Joint first authorship. size and number with age to occupy the thalamus and amygdala by

0304-3940/$ – see front matter © 2009 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.neulet.2009.07.055
94 P. Szot et al. / Neuroscience Letters 463 (2009) 93–97

the age of 12 months and eventually large parts of the hippocam- the LC that exhibit TH-positively labeled neurons. The density of
pus and neocortex at the age of 24 months [38]. APP23 mice display TH mRNA expression per neuron was performed measuring silver
high levels of face validity with regard to cognitive and behavioral grains over the cell bodies of labeled neurons that were three-
alterations [20,21,31,49,52,54–58], as well as predictive validity fold higher than background under 20× dark-field illumination
[48,53]. Impairment of cholinergic transmission, the prominent using the MicroComputer Imaging Device system (MCID) (Imag-
neurochemical deficit in Alzheimer brain, as indicated by decreased ing Research, St. Catherines, Ontario, Canada). Therefore, all labeled
specific acetylcholinesterase and choline acetyltransferase activity neurons that were counted as positively labeled were also quanti-
levels was observed in basal forebrain nuclei of APP23 transgenic fied for the amount of TH mRNA expression per neuron. The data
mice [51,52]. Recently, Van Dam et al. [51] described changes in are expressed as the average of grains/neuron ± SEM bilaterally over
norepinephrine (NE) concentration in specific regions of the APP23 the 5 consecutive sections for each animal.
transgenic mouse. In most brain regions analyzed, increased NE lev- Two-way analysis of variance with correction for repeated
els compared to wild-type (WT) values were seen. These increased measures (2-way RM-ANOVA) was used to evaluate significant dif-
NE levels reached significance in frontal cortex, mesencephalon and ferences between mean number of TH-positively labeled neurons
medulla oblongata. These data suggest an alteration in the nora- and the amount of TH grains/neuron. Genotype and age were con-
drenergic LC neurons. Therefore, the purpose of this study was to sidered as possible sources of variation. In case of a significant
determine if there is a loss of noradrenergic neurons in the LC of P-value, a post hoc Tukey multiple comparison tests were run. Sta-
APP23 transgenic mouse as compared to WT mice, or if the LC nora- tistical analyses were performed using Sigmastat software (SPSS
drenergic neurons have altered synthesis for NE. Because there is an Inc., Erkrath, Germany) with the level of probability set at 95%.
age-dependent deposition of amyloid plaques in the APP23 trans- The number of TH-positively labeled neurons was summed over
genic mouse, we measured tyrosine hydroxylase (TH) mRNA in the 5 consecutive sections (bilaterally) through the LC (Fig. 1A). The
LC of APP23 mice at various ages before and after the deposition of amount of TH mRNA expressed per positively labeled neuron was
plaques. TH is the rate-limiting enzyme in the synthesis of NE and averaged over the 5 consecutive sections (bilateral) (Fig. 1B). Two-
has been used to assess the loss of noradrenergic neurons in the LC way RM-ANOVA failed to show a significant effect of genotype on
in AD subjects [40]. both parameters (P = 0.343 and P = 0.566, respectively). However, a
Transgenic APP23 mice were genetically engineered in a hybrid significant effect of age, irrespective of genotype, was observed for
C57BL/6 × DBA2 background by Sturchler-Pierrat et al. [38]. The both the number of TH-positive neurons (F2,47 = 16.901, P < 0.001)
neuron-specific murine Thy-1.2 promoter drives human amyloid and the amount of TH grains/neuron (F2,47 = 12.977, P < 0.001).
precursor protein (APP) 751 cDNA harboring the Swedish dou- No significant effect of the interaction genotype × age was noted
ble mutation (K670N/M671L), associated with familial AD. Mice (P = 0.788 and P = 0.320, respectively). Post hoc Tukey multiple
were backcrossed to the C57BL/6J strain for at least 20 gen- comparisons indicated a significant increase in the number of TH-
erations to obtain an isogenic line. Male heterozygous APP23 positive neurons and the amount of TH grains/neuron expressed
mice and WT control littermates were bred and aged within in LC between the age of 3 months and 6 months (P < 0.001; for
the facilities of the Laboratory of Neurochemistry and Behaviour, both parameters) and 3 months and 12 months (P < 0.001; for both
University of Antwerp. Genotypes were confirmed by PCR. Mice parameters) in both WT and APP mice, but not between 6 months
were housed in mixed-genotype groups in standard mouse cages and 12 months (P = 0.289 and P = 0.904). Dark-field images of TH
(38.2 × 22 × 15 cm; length × width × height) with sawdust as bed- mRNA labeling in the LC are shown at midlevel of the LC in 3, 6 and
ding material and under conventional laboratory conditions; 12-month-old WT mice (Fig. 1C).
constant room temperature (22 ± 2 ◦ C), humidity level (55 ± 5%), a The APP23 mouse does not exhibit a loss of noradrenergic neu-
12-h light:12-h dark cycle (lights on at 8 AM) and food (Carfil, Oud- rons in the LC at any age studied or a change in the expression of
Turnhout, Belgium) and water available ad libitum. All experiments TH in each neuron, despite a previous study suggesting alterations
were approved by the Animal Ethics Committee of the University in the noradrenergic transmission system of APP23 mice aged 7–8
of Antwerp and performed in accordance with the European Com- months [51]. It should be kept in mind, however, that this previ-
munities Council Directive (86/609/EEC). ous study used HPLC analysis of NE levels, which was not able to
Heterozygous APP23 mice and WT littermates were killed by distinguish between interstitial and intracellular neurotransmitter
cervical dislocation at the age of 3 months (APP23 n = 8; WT n = 10), levels. In addition, we should state that the presumed compen-
6 months (APP23 n = 9; WT n = 8) or 12 months (APP23 n = 7; WT satory activation of noradrenergic neurons was not substantiated by
n = 11). Brains were quickly removed, frozen on dry ice and stored at the analysis of 3-methoxy-4-hydroxyphenylglycol (MHPG). Since
−80 ◦ C until further manipulations. Serial coronal sections (18 ␮m) MHPG is the predominant metabolite of NE, an increase of the
through the LC were cut on a cryostat, thaw mounted onto Fisher MHPG: NE ratio would suggest activation of the noradrenergic sys-
Superfrost slides, and stored at −70 ◦ C. tem. The lack of TH mRNA alterations in light of the previously
Tissue preparation and labeling of TH oligonucleotide probe published alteration in NE concentrations in APP23 brain could also
was performed as described previously [42]. The TH oligonu- be attributed to a variety of other mechanism, including TH protein
cleotide probe was a 48-base probe complementary to nucleotides stability, NE re-uptake into the terminal and NE uptake into the
1351–1398 of the rat TH mRNA [17]. The oligonucleotide was vesicle. Also, several of the regions where an increase in NE tissue
3 -end-labeled with [33 P]dATP (Perkin Elmer, Boston, MA) using content was measured previously [51], receive only partial LC inner-
terminal deoxyribonculeotidyl transferase (Invitrogen, Piscataway, vation. APP23 mice are considered a valid animal model of AD; they
NJ). The TH probe contained 0.93 × 106 cpm/50 ␮l and was washed display an age-dependent deposition of plaques in the cortex, tha-
as described in detail in previously published work [42]. Slides lamus, amygdala and hippocampus [38], cognitive and behavioral
were coated with NTB Nuclear Track Emulsion (undiluted) (East- impairment [20,21,31,49,52,54–58] and neuronal loss [5,6]. How-
man Kodak, Rochester, NY) and stored at −20 ◦ C for 4 days. Slides ever, this study indicates there is no loss of noradrenergic neurons
were developed as described previously [42]. in the LC of APP23 transgenic mice, even at an age when plaques
Quantification of TH mRNA in the LC expression was similar to are present [38].
that performed by Szot et al. [40]. The number of cells that achieved The link between LC degeneration and AD was previously shown
labeling three-fold higher than background was counted bilaterally by Heneka et al. [18] in the APP23 model. Neurotoxic lesioning of the
over 5 consecutive sections for each animal and totaled as TH- LC augmented amyloid burden paralleled with increased inflam-
positively labeled cell ± SEM. The 5 sections encompass ∼80% of matory reactions, as well as increased memory deficits, reduced
P. Szot et al. / Neuroscience Letters 463 (2009) 93–97 95

Fig. 1. Quantification of tyrosine hydroxylase (TH) mRNA expression in the locus coeruleus (LC) of APP23 mice at 3, 6, and 12 months. (A) Represents the mean summed
number of TH-positively labeled neurons over five consecutive slices through the LC of 3, 6 and 12-month-old APP23 mice (black bars) and wild-type (WT) control littermates
(white bars). (B) Represents the average expression of TH grains/neuron in the same set of mice. (C) Dark-field images of TH mRNA labeling in the LC of 3, 6, and 12-month-old
WT mice. Asterisks indicate a significant effect of age with respect to 3-month-old mice irrespective of genotype by post hoc Tukey multiple comparison: ***P < 0.001. Scale
bar = 100 ␮m.

cerebral glucose metabolism, disturbed neuronal integrity and Concerning LC-related changes with further aging, some contra-
attenuated acetylcholinesterase activity. The integrity of the LC in dictory observations have been published; both increased [37] and
aged hemizygous PDAPP mice (age 23 months) showed no loss of unaltered [9,46,47] TH mRNA levels have been described in aging
TH-positive neurons in the LC, but 24-month-old homozygous mice rat LC. Much less information, however, is available for postnatal LC
exhibited neuronal shrinkage in the portion of the LC with neu- development and TH levels, as well as further age-related changes
rons projecting to the cortex and hippocampus [14]. However, aged in mouse brain [45]. Noradrenergic neurons of the LC maintain their
(16–23 months) double transgenic mice expressing mutated forms capacity to adapt and respond to, e.g. injury well into adulthood, as
of both human APP and presenilin 1 (APP/PS1) showed a reduc- exemplified in lesion studies. Several reports have indicated the
tion of 24% to 50% in the total number of TH-positive neurons in presence of transient or “sleeping” neurons in which TH expression
the LC compared to age-matched WT mice using immunohisto- is inactivated or at a low level of activation [2,3]. Our observations
chemistry [22,27]. Degeneration of noradrenergic neurons in the indicate that both activation of transient cells, as well as increased
LC of APP/PS1 mouse occurs after the deposition of A␤ and loss activation of TH-positive neurons can be presumed.
of noradrenergic terminals in forebrain regions [22]. In addition, These observations indicate that any study comparing the nora-
the APP/PS1 mice were previously shown to exhibit significantly drenergic system between WT (C57Bl/6) and experimental mice
reduced hippocampal NE levels at the age of 12 and 18 months (e.g., lesion model, transgenic model) should strictly choose the
(30% and 40%, respectively), whereas no changes were noted in age to be tested and limit age differences between control and
frontal cortex and brainstem [39]. So it appears the presence of both experimental groups to the absolute minimum. More importantly,
mutation in the APP and PS1 gene are required to observe a loss of when chronic therapeutic interventions targeting the noradrener-
noradrenergic neurons. However, these studies did not determine gic system are applied to a mouse model, and related parameters are
if there is compensation in the expression of TH in the surviving studied longitudinally, care should be taken to distinguish between
neurons, as observed in AD subjects [40]. potential therapeutic and strain-specific developmental or age-
The present study clearly indicates an important increase of related alterations.
the LC-NE system after 3 months in C57BL/6 mice, which seems
independent of genotype in this case. Both the number of TH- Acknowledgements
positive cells and the number of TH-positive grains/cell significantly
increased between the age of 3 and 6 months with no change in This work was supported by the Fund for Scientific Research-
noradrenergic markers between 6 months and 12 months. To our Flanders (FWO grant G.0164.09), Interuniversity Poles of Attraction
knowledge, this is the first study describing such age-dependent (IAP Network P6/43), Methusalem Financing, Stichting Alzheimer
changes in LC of mice, and of C57BL/6 in particular. Several stud- Onderzoek/Fondation pour la Recherche sur la Maladie Alzheimer
ies have shown changes in the number of TH-positive LC neurons (SAO/FRMA), agreement between Institute Born-Bunge and the
and/or LC volume in rat brain around postnatal day 14 [2,30,35]. University of Antwerp, the Medical Research Foundation Antwerp,
96 P. Szot et al. / Neuroscience Letters 463 (2009) 93–97

Neurosearch Antwerp, the Thomas Riellaerts Research Fund and [24] J.J. Mann, M. Stanley, A. Neophytides, M.J. de Leon, S.H. Ferris, S. Gershon, Cen-
Mental Illness Research, Education, Clinical Center, Veterans tral amine metabolism in Alzheimer’s disease: in vivo relationship to cognitive
deficit, Neurobiol. Aging 2 (1981) 57–60.
Administration Puget Sound Health Care System (PS). D.V.D. is a [25] B. Marcyniuk, D.M. Mann, P.O. Yates, The topography of cell loss from locus
Postdoctoral Fellow of the FWO. caeruleus in Alzheimer’s disease, J. Neurol. Sci. 76 (1986) 335–345.
[26] M. Mullan, H. Houlden, M. Windelspecht, L. Fidani, C. Lombardi, P. Diaz, M.
Rossor, R. Crook, J. Hardy, K. Duff, F. Crawford, A locus for familial early-onset
References Alzheimer’s disease on the long arm of chromosome 14, proximal to the ␣1-
antichymotrypsin gene, Nat. Genet. 2 (1992) 340–342.
[1] R. Adolfsson, C.G. Gottfries, B.E. Roos, B. Winblad, Changes in the brain cate- [27] J.N. O’Neil, P.R. Mouton, Y. Tizabi, M.A. Ottinger, D.L. Lei, D.K. Ingram, K.F. Man-
cholamines in patients with dementia of Alzheimer’s type, Br. J. Psychiatry 135 aye, Catecholaminergic neuronal loss in locus coeruleus of aged female dtg
(1979) 216–223. APP/PS1 mice, J. Chem. Neuroanat. 34 (2007) 102–107.
[2] L. Bezin, D. Marcel, S. Desgeorges, J.-F. Pujol, D. Weissmann, Singular subsets [28] A.M. Palmer, P.T. Francis, D.M. Bowen, J.S. Benton, D. Neary, D.M. Mann, J.S.
of locus coeruleus neurons may recover tyrosine hydroxylase phenotype tran- Snowden, Catecholaminergic neurones assessed ante-mortem in Alzheimer’s
siently expressed during development, Mol. Brain Res. 76 (2000) 275–281. disease, Brain Res. 414 (1987) 365–375.
[3] L. Bezin, D. Marcel, C. Rousset, J.F. Pujol, D. Weissmann, Quantitative study of [29] E.K. Perry, G. Blessed, B.E. Tomlinson, R.H. Perry, T.J. Crow, A.J. Cross, G.J. Dock-
tyrosine hydroxylase protein levels within the somatic area of the rat locus ray, R. Dimaline, A. Arregui, Neurochemical activities in human temporal lobe
coeruleus during postnatal development, J. Neurosci. 14 (1994) 7502–7510. related to aging and Alzheimer’s-type changes, Neurobiol. Aging 2 (1981)
[4] W. Bondareff, C.Q. Mountjoy, M. Roth, Loss of neurons of origin of the adren- 251–256.
ergic projection to cerebral cortex (nucleus locus ceruleus) in senile dementia, [30] H. Pinos, P. Collado, M. Rodriguez-Zafra, C. Rodriguez, S. Segovia, A. Guillamon,
Neurology 32 (1982) 164–168. The development of sex differences in the locus coeruleus of the rat, Brain Res.
[5] L. Bondolfi, M. Calhoun, F. Ermini, H.G. Kuhn, K.H. Wiederhold, L. Walker, M. Bull. 56 (2001) 73–78.
Staufenbiel, M. Jucker, Amyloid-associated neuron loss and gliogenesis in the [31] L. Prut, D. Abramowski, T. Krucker, C.L. Levy, A.J. Roberts, M. Staufenbiel, C.
neocortex of amyloid precursor protein transgenic mice, J. Neurosci. 15 (2002) Wiessner, Aged APP23 mice show a delay in switching to the use of a strategy
515–522. in the Barnes maze, Behav. Brain Res. 179 (2007) 107–110.
[6] M.E. Calhoun, K.H. Wiederhold, D. Abramowski, A.L. Phinney, A. Probst, C. [32] M.A. Raskind, E.R. Peskind, J.B. Halter, D.C. Jimerson, Norepinephrine and MHPG
Sturchler-Pierrat, M. Staufenbiel, B. Sommer, M. Jucker, Neuron loss in APP levels in CSF and plasma in Alzheimer’s disease, Arch. Gen. Psychiatry 41 (1984)
transgenic mice, Nature 395 (1998) 755–756. 343–346.
[7] V. Chan-Palay, E. Asan, Alterations in catecholamine neurons of the locus [33] K.J. Reinikainen, L. Paljarvi, M. Huuskonen, H. Soininen, M. Laakso, P. Reikkinen,
coeruleus in senile dementia of the Alzheimer type and in Parkinson’s dis- A post-mortem study of noradrenergic, serotonergic and GABAergic neurons in
ease with and without dementia and depression, J. Comp. Neurol. 287 (1989) Alzheimer’s disease, J. Neurol. Sci. 84 (1988) 101–116.
373–392. [34] A. Russo-Neustadt, T.J. Zomorodian, C.W. Cotman, Preserved cerebellar tyro-
[8] M.-C. Chartier-Harlin, F. Crawford, H. Houlden, A. Warren, D. Hughes, L. Fidani, A. sine hydroxylase-immunoreactive neuronal fibers in a behaviourally aggressive
Goate, M. Rossor, P. Roques, J. Hardy, M. Mullan, Early-onset Alzheimer’s disease subgroup of Alzheimer’s disease patients, Neuroscience 87 (1998) 55–61.
caused by mutations at codon 717 of the beta-amyloid precursor protein gene, [35] K. Saito, M. Shimada, K. Kitahama, T. Maeda, Postnatal development of adren-
Nature 353 (1991) 844–846. ergic terminals in rat locus coeruleus, with special reference to growth of
[9] G. Cizza, K. Pacak, R. Kvetnansky, M. Palkovits, D.S. Goldstein, L.S. Brady, K. noradrenergic neurons, Brain Res. Dev. Brain Res. 96 (1996) 241–248.
Fukuhara, E. Bergamini, I.J. Kopin, M.R. Blackman, G.P. Chrousos, P.W. Gold, [36] G.D. Schellenberg, L. Anderson, S. O’dahl, E.M. Wisjman, A.D. Sadovnick, M.J.
Decreased stress responsivity of central and peripheral catecholaminergic sys- Ball, E.B. Larson, W.A. Kukull, G.M. Martin, A.D. Roses, T.D. Bird, APP717 , APP693 ,
tems in aged 344/N Fischer rats, J. Clin. Invest. 95 (1995) 1217–1224. and PRIG gene mutations are rare in Alzheimer’s disease, Am. J. Genet. 49 (1991)
[10] A.J. Cross, T.J. Crow, E.K. Perry, R.H. Perry, G. Blessed, B.E. Tomlinson, Reduced 511–517.
dopamine-beta-hydroxylase activity in Alzheimer’s disease, Br. Med. J. Clin. Res. [37] M.M. Shores, S.S. White, R.C. Veith, P. Szot, Tyrosine hydroxylase mRNA is
Ed. 282 (1981) 93–94. increased in old age and norepinephrine uptake transporter mRNA is decreased
[11] R. Elrod, E.R. Peskind, L. DiGiacomo, K.I. Brodkin, R.C. Veith, M.A. Raskind, Effects in middle age in locus coeruleus of Brown-Norway rats, Brain Res. 826 (1999)
of Alzheimer’s disease severity on cerebrospinal fluid norepinephrine concen- 143–147.
tration, Am. J. Psychiatry 154 (1997) 25–30. [38] C. Sturchler-Pierrat, D. Abramowski, M. Duke, K.H. Wiederhold, C. Mistl, S.
[12] D.C. German, A.J. Eisch, Mouse models of Alzheimer’s disease: insight into treat- Rothacher, B. Ledermann, K. Burki, P. Frey, P.A. Paganetti, C. Waridel, M.E. Cal-
ment, Rev. Neurosci. 15 (2004) 353–369. houn, M. Jucker, A. Probst, M. Staufenbiel, B. Sommer, Two amyloid precursor
[13] D.C. German, K.F. Manaye, C.L. White 3rd, D.J. Woodward, D.D. McIntire, W.K. protein transgenic mouse models with Alzheimer disease-like pathology, Proc.
Smith, R.N. Kalaria, D.M. Mann, Disease-specific patterns of locus coeruleus cell Natl. Acad. Sci. U.S.A. 94 (1997) 13287–13292.
loss, Ann. Neurol. 32 (1992) 667–676. [39] M.E. Szapacs, A.L. Numis, A.M. Andrews, Late onset loss of hippocampal 5-HT
[14] D.C. German, O. Nelson, F. Liang, C.L. Liang, D. Games, The PDAPP mouse model and NE is accompanied by increases in BDNF protein expression in mice co-
of Alzheimer’s disease: locus coeruleus neuronal shrinkage, J. Comp. Neurol. expressing mutant APP and PS1, Neurobiol. Dis. 16 (2004) 572–580.
492 (2005) 469–476. [40] P. Szot, S.S. White, J.L. Greenup, J.B. Leverenz, E.R. Peskind, M.A. Raskind,
[15] M.E. Gibbs, R.J. Summers, Role of adrenoceptor subtypes in memory consolida- Compensatory changes in the noradrenergic nervous system in the locus
tion, Prog. Neurobiol. 67 (2002) 345–391. coeruleus and hippocampus of postmortem subjects with Alzheimer’s disease
[16] C.G. Gottfries, R. Adolfsson, S.M. Aquilonius, A. Carlsson, S.A. Eckernäs, A. Nord- and dementia with Lewy bodies, J. Neurosci. 26 (2006) 467–478.
berg, L. Oreland, L. Svennerholm, A. Wiberg, B. Winblad, Biochemical changes [41] P. Szot, S.S. White, J.L. Greenup, J.B. Leverenz, E.R. Peskind, M.A. Raskind,
in dementia disorders of Alzheimer type (AD/SDAT), Neurobiol. Aging 4 (1983) Changes in adrenoreceptors in the prefrontal cortex of subjects with dementia:
261–271. evidence of compensatory changes, Neuroscience 146 (2007) 471–480.
[17] B. Grima, A. Lamouroux, F. Blanot, N.F. Biguet, J. Mallet, Complete coding [42] P. Szot, S.S. White, R.C. Veith, Effect of pentylenetetrazol on the expression
sequence of rat tyrosine hydroxylase mRNA, Proc. Natl. Acad. Sci. U.S.A. 82 of tyrosine hydroxylase mRNA and norepinephrine and dopamine transporter
(1985) 617–621. mRNA, Brain Res. Mol. Brain Res. 44 (1997) 46–54.
[18] M.T. Heneka, M. Ramanathan, A.H. Jacobs, L. Dumitrescu-Ozimek, A. Bilkei- [43] H. Tohgi, M. Ueno, T. Abe, S. Takahashi, Y. Nozaki, Concentrations of monoamines
Gorzo, T. Debeir, M. Sastre, N. Galldiks, A. Zimmer, M. Hoehn, W.D. Heiss, T. and their metabolites in the cerebrospinal fluid from patients with senile
Klockgether, M. Staufenbiel, Locus ceruleus degeneration promotes Alzheimer dementia of the Alzheimer type and vascular dementia of the Binswanger type,
pathogenesis in amyloid precursor protein 23 transgenic mice, J. Neurosci. 26 J. Neural. Transm. Parkinson Dis. Dement. Sect. 4 (1992) 69–77.
(2006) 1343–1354. [44] B.E. Tomlinson, D. Irving, G. Blessed, Cell loss in the locus coeruleus in senile
[19] W.J. Hoogendijk, M.G. Feenstra, M.H. Botterblom, J. Gilhuis, I.E. Sommer, W. dementia of Alzheimer type, J. Neurol. Sci. 49 (1981) 419–428.
Kamphorst, P. Eikelenboom, D.F. Swaab, Increased activity of surviving locus [45] M. Touret, J.L. Valatx, M. Jouvet, The locus coeruleus: a quantitative and genetic
ceruleus neurons in Alzheimer’s disease, Ann. Neurol. 45 (1999) 82–91. study in mice, Brain Res. 250 (1982) 353–357.
[20] P.H. Kelly, L. Bondolfi, D. Hunziker, H.P. Schlecht, K. Carver, E. Maguire, D. [46] N. Tümer, H.A. Demirel, L. Serova, E.L. Sabban, C.S. Broxson, S.K. Powers, Gene
Abramowski, K.H. Wiederhold, C. Sturchler-Pierrat, M. Jucker, R. Bergmann, expression of catecholamine biosynthetic enzymes following exercise: modu-
M. Staufenbiel, B. Sommer, Progressive age-related impairment of cognitive lation by age, Neuroscience 103 (2001) 703–711.
behavior in APP23 transgenic mice, Neurobiol. Aging 24 (2003) 365–378. [47] J.R. Unnerstall, M.M. Long, Differential effects of the intraventricular admin-
[21] R. Lalonde, M. Dumont, M. Staufenbiel, C. Sturchler-Pierrat, C. Strazielle, istration of 6-hydroxydopamine on the induction of type II beta-tubulin and
Spatial learning, exploration, anxiety, and motor coordination in female tyrosine hydroxylase mRNA in the locus coeruleus of the aging Fischer 344 rat,
APP23 transgenic mice with the Swedish mutation, Brain Res. 956 (2002) J. Comp. Neurol. 364 (1996) 363–381.
36–44. [48] D. Van Dam, D. Abramowski, M. Staufenbiel, P.P. De Deyn, Symptomatic effect
[22] Y. Liu, M.-J. Yoo, A. Savonenko, W. Stirling, D.L. Price, D.R. Borchelt, L. Mamounas, of donepezil, rivastigmine, galantamine and memantine on cognitive deficits
W.E. Lyons, M. Blue, M.K. Lee, Amyloid pathology is associated with pro- in the APP23 model, Psychopharmacology (Berl.) 180 (2005) 177–190.
gressive monoaminergic neurodegeneration in a transgenic mouse model of [49] D. Van Dam, R. D’Hooge, M. Staufenbiel, C. Van Ginneken, F. Van Meir, P.P. De
Alzheimer’s disease, J. Neurosci. 28 (2008) 13805–13814. Deyn, Age-dependent cognitive decline in the APP23 model precedes amyloid
[23] D.M. Mann, J. Lincoln, P.O. Yates, J.E. Stamp, S. Toper, Changes in the monoamine deposition, Eur. J. Neurosci. 17 (2003) 388–396.
containing neurones of the human CNS in senile dementia, Br. J. Psychiatry 136 [50] D. Van Dam, P.P. De Deyn, Drug discovery in dementia: the role of rodent models,
(1980) 533–541. Nat. Rev. Drug Discov. 5 (2006) 956–970.
P. Szot et al. / Neuroscience Letters 463 (2009) 93–97 97

[51] D. Van Dam, B. Marescau, S. Engelborghs, T. Cremers, J. Mulder, M. Staufenbiel, [55] E. Vloeberghs, D. Van Dam, R. D’Hooge, M. Staufenbiel, P.P. De Deyn, APP23 mice
P.P. De Deyn, Analysis of cholinergic markers, biogenic amines, and amino acids display working memory impairment in the plus-shaped water maze, Neurosci.
in the CNS of two APP overexpression mouse models, Neurochem. Int. 46 (2005) Lett. 407 (2006) 6–10.
409–422. [56] E. Vloeberghs, D. Van Dam, S. Engelborghs, G. Nagels, M. Staufenbiel, P.P. De
[52] D. Van Dam, E. Vloeberghs, D. Abramowski, M. Staufenbiel, P.P. De Deyn, APP23 Deyn, Altered circadian locomotor activity in APP23 mice: a model for BPSD
mice as a model of Alzheimer’s disease: an example of a transgenic approach disturbances, Eur. J. Neurosci. 20 (2004) 2757–2766.
to modeling a CNS disorder, CNS Spectr. 10 (2005) 207–222. [57] E. Vloeberghs, D. Van Dam, F. Franck, J. Serroyen, M. Geert, M. Staufenbiel, P.P.
[53] E. Vloeberghs, K. Coen, D. Van Dam, P.P. De Deyn, Validation of the APP23 trans- De Deyn, Altered ingestive behavior, weight changes, and intact olfactory sense
genic mouse model of Alzheimer’s disease through evaluation of risperidone in an APP overexpression model, Behav. Neurosci. 122 (2008) 491–497.
treatment on aggressive behaviour, Arzneimittelforschung 58 (2008) 265–268. [58] E. Vloeberghs, D. Van Dam, F. Franck, M. Staufenbiel, P.P. De Deyn, Mood and
[54] E. Vloeberghs, D. Van Dam, K. Coen, M. Staufenbiel, P.P. De Deyn, Aggressive male male sexual behaviour in the APP23 model of Alzheimer’s disease, Behav. Brain
APP23 mice modeling behavioral alterations in dementia, Behav. Neurosci. 120 Res. 180 (2007) 146–151.
(2006) 1380–1383.

You might also like