Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 May 1.
Published in final edited form as: Neurosci Biobehav Rev. 2012 Feb 21;36(5):1357–1375. doi: 10.1016/j.neubiorev.2012.02.011

APP transgenic mice for modelling behavioral and psychological symptoms of dementia (BPSD)

R Lalonde 1,*, K Fukuchi 2, C Strazielle 3
PMCID: PMC3340431  NIHMSID: NIHMS366299  PMID: 22373961

Abstract

The discovery of gene mutations responsible for autosomal dominant Alzheimer's disease has enabled researchers to reproduce in transgenic mice several hallmarks of this disorder, notably Aβ accumulation, though in most cases without neurofibrillary tangles. Mice expressing mutated and wild-type APP as well as C-terminal fragments of APP exhibit variations in exploratory activity reminiscent of behavioral and psychological symptoms of Alzeimer dementia (BPSD). In particular, open-field, spontaneous alternation, and elevated plus-maze tasks as well as aggression are modified in several APP transgenic mice relative to non-transgenic controls. However, depending on the precise murine models, changes in open-field and elevated plus-maze exploration occur in either direction, either increased or decreased relative to controls. It remains to be determined which neurotransmitter changes are responsible for this variability, in particular with respect to GABA, 5HT, and dopamine.

Keywords: amyloid, Alzheimer's disease, open-field, spontaneous alternation, elevated plus-maze, emergence test, GABA, aggression

1. Pathobiology of Alzheimer's disease

1.1 Cognitive symptoms of Alzheimer's disease

Probable Alzheimer's disease is diagnosed when progressive loss in remembering new items (anterograde amnesia) occurs in conjunction with a deficit in language (aphasia), object use (apraxia), form recognition (agnosia), or step-by-step planning (McKhann et al., 1984). In general, memory and verbal fluency deficits are the initial symptoms (Amieva et al., 2005), progressive amnesia sometimes being the only sign over a period of several years (Weintraub and Mesulam, 1993). A loss of factual information (declarative memory) is found for verbal and spatial items (Amieva et al., 2005; Graham et al., 2004; Helmes and Ostbye 2002; Jacobs et al., 1995; Kessels et al., 2005; Lee et al., 2003), whereas procedural memory is relatively spared, evaluated in pursuit-rotor (Eslinger and Damasio, 1986; Jacobs et al., 1999) and either verbal (Karlsson et al., 2002) or spatial (Kessels et al., 2005) priming tasks. However, some procedural tasks are impaired, such as prepulse inhibition of the acoustic startle response, implying a deficit in sensory gating (Ueki et al., 2006), as well as delay or trace conditioning of the eyeblink response (Woodruff-Pak and Papka, 1996; Woodruff-Pak et al., 1996).

In addition to amnesia, patients with Alzheimer's disease are susceptible to constructional apraxia, characterized by a difficulty in copying geometric figures or reproducing them with building blocks (Graham et al., 2004; Helmes and Ostbye 2002; Henderson et al., 1989; Nielson et al., 1996). Impaired flow perception (Kavcic et al., 2006; Tetewsky and Duffy, 1999), visual search (Parasuraman et al., 2000; Tales et al., 2004), abstract reasoning (Helmes and Ostbye 2002), and executive functions (Collette et al., 1999; Graham et al., 2004; Rainville et al., 2002b) may contribute to this symptom. As a result, they are more likely than controls to be disoriented in a maze (Rainville et al., 2002a), a circular arena (Hort et al., 2007), a hospital ward (Monacelli et al., 2003), and in the streets when driving a motored vehicle (Uc et al., 2004).

1.2 Behavioral and psychological symptoms of Alzheimer's disease

Among emotional symptoms, apathy is one of the most common features of Alzheimer dementia, sometimes accompanied by dysphoria, social withdrawal, and depression (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004). But sometimes agitation is the predominant feature, accompanied by disinhibition and inappropriate euphoria. These opposite signs often overlap in the same patient, as apathy scores have been found to be linearly correlated with disinhibition (Holthoff et al., 2005; Lyketsos et al., 2002). Apathy/anxiety scores, on one hand, and disinhibition/euphoria scores, on the other, were both correlated with caregiver non-compliance (Senanarong et al., 2004), underlying their importance in day-to-day activities. Symptoms may differ from one patient to another with respect to inhibition or disinhibition. Alzheimer patients may either be excessively inhibited due to anxiety, or else display a loss in inhibitory control, often accompanied by irritability and hostility (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004). Excessive aggression may be due to degeneration of brainstem 5-hydroxytryptamine (5HT) neurons, known to influence this symptom (Hardy et al., 1985; Lyness et al., 2003; Whitford 1986).

1.3 Behavioral symptoms linked with metabolic markers

Insight into the neurobiological factors underlying dementia symptoms may be obtained by measuring regional brain metabolism. Alzheimer patients characterized by apathy had lower fluorodeoxyglucose (FDG) uptake in medial prefrontal cortex than a subgroup not displaying this sign (Holthoff et al., 2005; Marshall et al., 2007). These results indicate that apathy may be the result of dysfunction at the level of medial prefrontal cortex. In addition, FDG uptake was lower in temporal neocortex of Alzheimer patients suffering the most from anxiety (Hashimoto et al., 2006). Moreover, Alzheimer patients with misidentification delusions had lower glucose utilization in orbitofrontal, medial temporal, and striate cortex but higher utilization in sensory association cortices (superior temporal and inferior parietal) than those without this symptom (Mentis et al., 1995). Likewise, demented patients with delusions had higher glucose utilization in inferior temporal but lower utilization in medial occipital cortex than those without them (Hirono et al., 1998). In addition to glucose, regional brain metabolism may be estimated by brain perfusion on single photon emission computed tomography (SPECT) scans by measuring the uptake of hexamethylpropelenamine oxime (HMPAO). The HMPAO technique dissociated between patient groups on the basis of emotional reactivity (Hirono et al., 2000). In particular, a mixed group of patients with Alzheimer's or vascular dementia displaying marked aggression had lower neocortical HMPAO uptake than a demented group not displaying this symptom.

2. Pathological characteristics in APP mice

2.1 Introduction

Thanks to the discovery of pathologic mutations, several Alzheimer hallmarks have been attained in generating transgenic mice (Cole and Frautschy, 1997; Dodart et al., 2002; Duff, 1999; Hock and Lamb, 2001; Hsiao, 1998a,b; Janus and Westaway, 2001; Seabrook and Rosahl 1999; van Leuven, 2000). But except for the one expressing APP, PS1, and Mapt (Oddo et al., 2003), APP mutants contain no neurofibrillary tangles, though often with hyperphosphorylated tau, thereby most resembling rare cases of Alzheimer dementia with amyloid deposits and hyperphosphorylated tau without neurofibrillary tangles (Tiraboschi et al., 2004a,b). The genetic characteristics of mice expressing mutated or wild-type APP or its C-terminal fragments are presented in Table 1.

Table 1.

Description of transgenic mice expressing human mutated or wild-type (WT) APP genes with or without mutated PS1 or else expressing Aβ species

Name Mutation Promoter Parenchymal
Aβ plaques
References
APP695SWE Tg2576 Swedish hamster Prp yes Hsiao et al., 1996
APP695SWE Tg2123H Swedish hamster Prp no Hsiao et al., 1995
APP695SWE/Thy1 TgTAS10 Swedish murine Thy1 yes Richardson et al., 2003
APP695SWEch Swedish murine PrP no Borchelt et al., 1996, 1997
APP751SWE TgAPP23 Swedish murine Thy1 yes Sturchler-Pierrat et al., 1997
APP770SWE Swedish murine Thy1 yes Moechars et al., 1999b
APP/SWE/Ckm Swedish murine Ckm no Sugarman et al., 2002
APP/IND TgPDAPP Indiana platelet-derived PDGFβ yes Games et al., 1995
APP695LD London murine Thy1 yes Moechars et al., 1999b
APP/LD-PDGF London platelet-derived PDGFβ yes Qin et al., 2000
APP/DU Dutch murine Thy1 no Kumar-Singh et al., 2000
APP/DU/THY1 Dutch human THY1 no Andrä et al.,1996
APP/FL Flemish murine Thy1 no Kumar-Singh et al., 2000
APP695SWE/IND TgCRND8 or Tg19959 Swedish+Indiana hamster Prp yes Chishti et al., 2001
APP/SWE/IND
J9 and J20 lines
Swedish+Indiana platelet-derived PDGFβ yes Mucke et al., 2000;
Palop et al., 2003
APP695SWE/IND/tet Swedish+Indiana pTetSplice yes Jankowski et al., 2005b
APP751SWE/IND TgAPP-Sw,V717F/B6 Swedish+Indiana platelet-derived PDGFβ no Lee et al., 2004
APP/SWE/ARC Swedish+Arctic murine Thy1 yes Lord et al., 2006
APP751SWE/LD Swedish+London murine Thy1 yes Blanchard et al., 2003
APP/SWE/LD Swedish+London murine PRP yes Kitamoto et al., 2002
APP/SWE/DI Swedish+Dutch+Iowa murine Thy1 yes Davis et al., 2004
APP/RK Artificial 684 + 687 positions murine Thy1 no Moechars et al., 1996
APP695TRImyc Tg1130H artificial hamster Prp no Hsiao et al., 1995
APP695WT/Mt2 none Mt2 no Yamaguchi et al., 1991
APP695WT/Thy1 none murine Thy1 no Moechars et al., 1999b
App695WT Tg1874 none hamster Prp no Hsiao et al., 1995
APP695mycWTTg6209 none hamster Prp no Hsiao et al., 1995
APP751WT none rat Nse no Quon et al., 1991
APP751WT none rat Nse no Mucke et al., 1994
APP WT Line I5 none PDGFβ no Mucke et al., 2000
APP/C104 none human NEFL yes Nalbantoglu et al.,1997
APP/C100-Flag none Dmd no Oster-Granite et al.,1996
APP/C99/IND Indiana platelet-derived PDGFβ no Lee et al., 2006
APP/C99 Tg13592 none CMVenhancer/chicken Actb no Fukuchi et al., 1996
APP/C100/IND APP/C100/WT Indiana wild-type human ACTB no Li et al., 2006
pyroglutamate-Aβ none murine Thy1 no Alexandru et al., 2011
40 and Aβ42 fused with BRI none murine Prp yes for Aβ42 McGowan et al., 2005
42 none murine Nefl no LaFerla et al., 1995
APP knockout null non-applicable no Müller et al., 1994
APP695SWE+PS1/M146L APP695SWE+PS1/M146V bigenic hamster Prp + PDGFβ2 yes Duff et al., 1996;
Holcomb et al., 1998
APP695SWE+PS1/A246E bigenic hamster Prp + human THY1 yes Dineley et al., 2002
Qian et al., 1998
APP695SWE+PS1/M146L/Thy1 TgTASTPM bigenic murine Thy1 yes Howlett et al., 2004
APP695SWEch+PS1/A246E bigenic murine PrP yes Borchelt et al., 1996
APP695SWEch+PS1/ΔE9 bigenic murine PrP yes Jankowsky et al., 2005a
APP695SWE/co+PS1/ΔE9 co-injected bigenic murine PrP yes Jankowsky et al., 2004
APP751SW+PS1/G384A bigenic murine ThyI for both yes Busche et al., 2008
APPswe+PS1/L166P co-injected bigenic murine Thy1 yes Radde et al., 2006
APP695LD+PS1/A246E bigenic murine Thy1 for both yes Van Dorpe et al., 2000
APP751SWE/LD+PS1/M146L bigenic murine Thy1 + human HMGCR yes Blanchard et al., 2003
APP751SWE/LD+PS1/M233T+L235P transgenic + knockin murine Thy1 yes Casas et al., 2004
APP/SWE/Ckm+PS1/M146V KI transgenic + knockin murine Ckm no Sugarman et al., 2002
APP751SWE+PS2/VG TgPS2APP bigenic murine Thy1 + murine PrP yes Richards et al., 2003
3xTg-AD trigenic murine Thy1 + endogenous control + murine Thy1+ yes Oddo et al., 2003
5xFAD 5-time mutated murine Thy1 yes Oakley et al., 2006

2.2 APP mutations

2.2.1 Swedish

The most cited murine Alzheimer model appears to be the Tg2576 mouse, containing an APP transgene with the Swedish mutation of the neuron-specific 695-amino acid isoform driven by the hamster Prp promoter (Hsiao et al., 1996). Detergent-insoluble Congo-red positive Aβ plaques were visible in APP695SWE mice as early as 7 months of age and increased with time (Kawarabayashi et al., 2001; Westerman et al., 2002). A second transgene with the same mutation and promoter (Tg2123H) does not harbor Aβ plaques (Hsiao et al., 1995). As seen in Alzheimer brain (Iwatsubo et al., 1994), diffuse plaques in APP695SWE mice contain Aβ42 and mature plaques Aβ42 and Aβ40 (Harigaya et al., 2006; Kawarabayashi et al., 2001; Terai et al., 2001). Unlike Alzheimer brain (Thal et al., 2000), cored Aβ plaques appeared earlier, at 12 months, than diffuse ones (Harigaya et al., 2006; Kawarabayashi et al., 2001), a feature appearing in other murine models such as APP695SWE/Thy1 (see below) (Howlett and Richardson, 2009), and these are accompanied by hyperphosphorylated tau without neurofibrillary tangles (Tomidokoro et al., 2001). Soluble Aβ dimers appeared in lipid rafts at 6 months (Kawarabayashi et al., 2004). As seen in Alzheimer brain, Aβ42 accumulated intracellularly before plaque formation in multivesicular bodies at pre- and postsynaptic sites (Takahashi et al., 2002) and lysosomes (Shie et al., 2003). A third resemblance with Alzheimer brain (Eikelenboom et al., 2002; McLellan et al., 2003) is the presence of astrocytes and microglia (Irizarry et al., 1997; Wegiel et al., 2001) with reactive oxygen species (McLellan et al., 2003) around fibrillar deposits. Gliosis is observed as soon as Aβ plaques appear (Jacobsen et al., 2006). While astrocytes isolate plaques from healthy tissue, microglia cannot phagocytose them (Wegiel et al., 2001, 2003). Glial-mediated inflammation is mostly seen in the vicinity of fibrillar Aβ plaques (Benzing et al., 1999; Quinn et al., 2003). The Aβ plaques also surround vessel walls, causing microhemorrhages (Domnitz et al., 2005; Frackowiak et al., 2001; Fryer et al., 2003).

Although spine density decreased in the CA1 hippocampal subregion of APP695SWE mice (Lanz et al., 2003), neuronal counts were unchanged (Irizarry et al., 1997). The spine density decline in molecular layer of dentate gyrus appeared even before plaque deposition (Jacobsen et al., 2006). White matter tracts appear intact (Harms et al., 2006). Thus, neurodegeneration seems minimal, probably because of enhanced APP-induced gene expression of cell survival pathways (Stein and Johnson, 2003), since chronic infusion of the antibody to the upregulated Aβ-binding protein, transthyretin, caused cell loss in the CA1 pyramidal layer of APP695SWE mice (Stein and Johnson, 2002).

A second highly cited model also contains the APP transgene with the Swedish mutation but this time driven by the endogenous PrP promoter of a chimeric (ch) human/murine 695-amino acid isoform (Borchelt et al., 1996, 1997). Despite increased Aβ42 and Aβ40 levels, few or no neuritic Aβ plaques were seen in APP695SWEch mouse brain (Savonenko et al., 2003). In contrast, the APP695SWE/Thy1 transgene (TgTAS10) driven by the murine neuron-specific Thy1 (thymus cell antigen 1) promoter generated Aβ plaques in cerebral cortex at 12 months of age, acompanied by astrocytosis, microgliosis, and hyperphosphorylated tau (Richardson et al., 2003).

Another highly cited transgene with the Swedish mutation driven by the murine Thy1 promoter was generated on the 751-amino acid isoform (Sturchler-Pierrat et al., 1997). APP751SWE mice (APP23) accumulate Aβ in hippocampus and neocortex, with plaque-associated Aβ peptides augmenting 5-fold from 3 to 6 months of age (Van Dam et al., 2003). As seen in Alzheimer brain, neuritic plaques are surrounded by astrocytes and activated microglia and tau is hyperphosphorylated (Bornemann et al., 2001; Stalder et al., 1999; Sturchler-Pierrat et al., 1997). A further resemblance between human (Lüth et al., 2001, 2002) and murine (Lüth et al., 2001) pathology is the overexpression of induced and endothelial forms of nitric oxide synthase (iNOS and eNOS) accompanying astrocytosis. Rodrigo et al. (2004) found increased iNOS but not nNOS concentrations in APP751SWE brain. A 10% loss of pyramidal neurons in the CA1 hippocampal subregion was documented (Calhoun et al., 1998), though, unlike advanced Alzheimer's disease, without loss in noradrenergic locus coeruleus neurons (Szot et al., 2009). Neocortical neurons increased by 10% and then declined by the same percentage with aging (Bondolfi et al., 2002). The initial increase is probably caused by the neurotrophic action of secreted APP (Stein and Johnson, 2003; Storey and Cappai, 1999). The neuronal loss was accompanied by increased DNA strand breaks (Bondolfi et al., 2002). Amyloid angiopathy led to weakened vessel walls, aneurysm, vasculitis, and hemorrhage (Beckmann et al., 2011; Calhoun et al., 1999; Winkler et al., 2001), as well as a reduction in their vasodilatory properties (Mueggler et al., 2002, 2003). In neocortex, the water diffusion coefficient was reduced (Mueggler et al., 2004) and size of extracellular space increased (Sykova et al., 2005). The chemical composition of Aβ plaques in APP751SWE (Kuo et al., 2001b) as well as the previously APP695SWE mutants (Kalback et al., 2002; Kuo et al., 2001a) resemble those found in Alzheimer's disease, except that the plaques examined in the two mouse models were more soluble (Kuo et al., 2001), characteristic of other murine models as well (Kokjohn and Roher, 2009).

Yet another APP model with the Swedish mutation driven by the murine Thy1 promoter includes the 770-amino acid isoform (Moechars et al., 1999b). Aβ plaques were observed in hippocampus and neocortex of these APP770SWE mice.

In an attempt to model inclusion body myopathy (IBM), a APP/SWE/Ckm transgene with the Swedish mutation driven by the murine Ckm (creatine kinase, muscle) promoter was generated (Sugarman et al., 2002). As found in patients (Askanas and Engel, 1998, 2001, 2002), Aβ accumulated intracellularly in mouse skeletal muscle (Sugarman et al., 2002). As seen in sporadic IBM but not hereditary IBM (Askanas and Engel 1998), inflammation was evident in the mouse model, with neutrophils instead of T cells as the predominant cell type (Sugarman et al., 2002).

2.2.2 Indiana

APP/IND transgenic mice (PDAPP) carry the V717F Indiana mutation of all three major APP isoforms driven by the PDGFβ promoter (Games et al., 1995). APP/IND mouse brain contains Congo-red positive Aβ plaques, appearing as early as 6 to 9 months of age, surrounded by proliferating microglia and astrocytes (Games et al., 1995; Masliah et al., 1996; Schenk et al., 1997) and reactive oxygen species (McLellan et al., 2003). There is also an age-dependent increase in phosphorylated tau (Masliah et al., 2001). As seen in APP695SWE mice (Harigaya et al., 2006; Kawarabayashi et al., 2001), cored Aβ plaques appeared earlier than diffuse ones (Reilly et al., 2003). Cerebral angiopathy is observed with microhemorrhages (Domnitz et al., 2005; Fryer et al., 2003; Kimchi et al., 2001). Although hippocampal volume was reduced (Gonzalez-Lima et al., 2001; Redwine et al., 2003; Weiss et al., 2002), cell counts were unchanged (Irizarry et al., 1997), attributable to dendritic or axonal atrophy, as dendritic length diminished in dentate gyrus granule cells as early as 3 months of age (Wu et al., 2004).

2.2.3 London

Two APP models have been generated with the V717I London mutation, one on the 695-amino acid isoform driven by the murine Thy1 promoter, causing diffuse and neuritic Aβ plaques (Moechars et al., 1999b; Dewachter et al., 2000a,b; Van Dorpe et al., 2000), surrounded by activated astrocytes and microglia, the neurites being linked with hyperphosphorylated tau (Moechars et al., 1999b). The angiopathy in APP695LD mice leads to aneurysms but not hemorrhages (Van Dorpe et al., 2000), potentiated after interbreeding with a PS1/A246E line (Van Dorpe et al., 2000). The second model includes an APP751LD transgene driven by the PDGFβ promoter, which also causes neuritic Aβ plaques (Qin et al., 2000).

2.2.4 Dutch and Flemish

APP/DU/Thy1 and APP/FL/Thy1 mice with Dutch (E693Q) and Flemish (A692G) mutations, respectively, each driven by the murine Thy1 promoter, have cerebral amyloid angiopathy with elevated C-terminal fragments in parenchymal regions but without Aβ plaques (Kumar-Singh et al., 2000), thus serving as animal models of cerebral amyloid angiopathy caused by such mutations. A second APP/DU/THY1 model with the human THY1 promoter also has cerebral amyloid angiopathy without Aβ plaques (Andrä et al., 1996; Gandy et al., 2010).

2.2.5 Swedish/Indiana, Swedish/Arctic, Swedish/London, and Swedish/Dutch/Iowa

Five models characterized by combined Swedish/Indiana mutations are available. APP695SWE/IND mice (TgCRND8 or Tg19959) express the 695-amino acid isoform regulated by the hamster Prp promoter (Chishti et al., 2001; Janus et al., 2000), showing Aβ plaques as early as 3 months of age (Bellucci et al., 2006; Chishti et al., 2001; Janus et al., 2000), together with early onset angiopathy (Domnitz et al., 2005) and activated microglia and astrocytes (Bellucci et al., 2006). As seen in APP695SWE (Harigaya et al., 2006; Kawarabayashi et al., 2001), APP/IND (Reilly et al., 2003), and other murine mutants (Howlett and Richardson, 2009), diffuse Aβ plaques appeared after cored Aβ plaques (Chishti et al., 2001). Two APP/SWE/IND constructs driven by the platelet-derived PDGFβ promoter (J9 and J20 lines) also contain Aβ plaques (Mucke et al., 2000; Palop et al., 2003), as well as mice with a human/mouse chimeric APP695SWE/IND/tet transgene regulated by the tetracycline-responsive promoter of pTetSplice (Jankowsky et al., 2005b). On the contrary, the APP751SWE/IND (APP-Sw,V717F/B6) model driven by the PDGFβ promoter did not harbor plaques up to 14 months of age (Lee et al., 2004). An APP/SWE/ARC line was generated with Swedish and Arctic mutations driven by a murine Thy1 cassette, with Aβ plaques occurring at 10 months of age (Lord et al., 2006, 2009). An APP SWE/LD line with Aβ plaques contains a combined Swedish/London mutation regulated by the murine PRP promoter (Kitamoto et al., 2002).

An APP751SWE/LD line with Swedish and London mutations on a transgene driven by the murine Thy1 promoter was also generated, containing Aβ plaques in parenchyma (Blanchard et al., 2003). In addition, an APP/SDI line was generated with Swedish, Dutch, and Iowa mutations causing Aβ plaque accumulation in parenchyma and cerebral vessels (Davis et al., 2004).

2.2.6 Artificial

APP/RK transgenic mice with an artifical double mutation at the α-secretase site were generated under control of the murine Thy1 promoter (Moechars et al., 1996). These mice harbor no Aβ plaques, but neocortex and hippocampus are marked by astrocytosis and DNA fragmentation (Moechars et al., 1998, 1999a). Likewise, no Aβ plaques were observable in APP695TRImyc mice (Tg1130H), expressing a triple mutation (V717L, V721A, M722V) with a 3'-myc tag driven by the hamster Prp promoter (Hsiao et al., 1995).

2.3 Murine wild-type App and human wild-type APP

The 695 splice variant was used for generating mice expressing human wild-type (WT) APP695under control of the Mt2 (metallothionein IIA) promoter (Yamaguchi et al., 1991). A similar APP695WT construct driven by the murine Thy1 promoter is available (Moechars et al., 1999b). Mice expressing human APP695mycWT with a 3'-myc tag (Tg6209) and murine App695WT (Tg1874) driven by the hamster Prp promoter have also been generated (Hsiao et al., 1995). In addition, two separate mouse lines expressing APP751WT regulated by the rat neuron-specific Nse promoter are available (Mucke et al., 1994; Quon et al., 1991), together with APP WT regulated by the PDGFβ promoter (line I5) (Mucke et al., 2000).

2.4 APP carboxy-terminal fragments and Aβ

In view of the neurotoxic properties of C-terminal fragments in cell cultures (Fukuchi et al., 1993; Kim and Suh, 1996) and their accumulation in Alzheimer brain (McPhie et al., 1997), liable to influence disease onset and progression (Turner et al., 2003), it is of interest to examine their neurobehavioral impact. APP/C104 mice with the human 104-amino acid C-terminal fragment transgene driven by the human NEFL promoter have Aβ plaques surrounded by reactive microglia and astrocytes as well as reduced neuronal counts in the CA1 hippocampal subregion (Nalbantoglu et al., 1997). APP/C99/IND transgenic mice express the human 99-amino acid C-terminal fragment with the V717F Indiana mutation driven by the PDGFβ promoter (Lee et al., 2006), while APP/C100/IND mice and APP/C100/WT mice express the human 100-amino acid C-terminal fragment with the V717F Indiana mutation or wild-type (WT), both driven by the Actb (β-actin) promoter (Li et al., 1999). The APP/C100-Flag transgene driven by the Dmd promoter uses the 100 amino acid C-terminal fragment fused with the hydrophilic 8-amino acid Flag epitope (Oster-Granite et al., 1996). APP/C99 mice (Tg13592) expressing the human 99-amino acid C-terminal fragment in brain and skeletal muscle driven by a CMV enhancer/chicken Actb promoter mimick IBM in that Aβ accumulates in muscle but not in brain (Fukuchi et al., 1996, 1998, 2000).

The direct impact of Aβ at different lengths has been examined. For example, transgenic mice overexpressing endogenous Aβ42 driven by the murine Nefl promoter have increased intracellular levels of the peptide (LaFerla et al., 1995, 1996). These mice are marked by DNA fragmentation, increased p53 levels, and morphological markers of apoptosis in neocortex, hippocampus, amygdala, and thalamus. Moreover, transgenic mice overexpressing either Aβ40 or Aβ42 in the form of fusion proteins with the BRI protein driven by the murine Prp promoter have been generated (McGowan et al., 2005). The influence of truncated Aβ species have also been examined, notably in a transgenic mouse model overexpressing pyroglutamate-Aβ driven by the murine Thy1 promoter, causing hippocampal cell loss, astrocytosis, microgliosis, and behavioral signs such as tremor (Alexandru et al., 2011).

2.5 APP null mutation

The only behaviorally characterized APP knockout model contains a deletion mutation of exon 2, resulting in a shorter protein (Müller et al., 1994).

2.6 APP+PS1 mutations

The above-described APP695SWE mice (Tg2576) were interbred with mutated PS1 lines 5.1 and 6.2, yielding APP695SWE+PS1/M146L and APP695SWE+PS1/M146V, respectively, characterized by earlier Aβ plaque onset than the monogenic (Duff et al., 1996; Holcomb et al., 1998, 1999; Kurt et al., 2003; Westerman et al., 2002), with the same features of astrocytosis and microgliosis (Gordon et al., 2002; Holcomb et al., 1998, 1999), the Aβ plaques surrounding vessel walls (Christie et al., 2001; Domnitz et al. 2005) with reduced vasodilation properties (Christie et al., 2001). The same APP695SWE line was also bred with a PS1/A246E strain on a Ps1 null background (Qian et al., 1998), once more yielding Aβ plaques with earlier onset than the monogenic (Dineley et al., 2002). A similar APP695SWE+PS1/M146L/Thy1 model with Aβ plaques (TgTASTPM) has been described with the mouse Thy1 promoter used for both transgenes, though not on a Ps1 null background (Howlett et al., 2004).

APP695SWEch mice (line C3-3) have also been bred with a PS1/A246E line, each transgene driven by the endogenous PrP promoter (Borchelt et al., 1996, 1997). An age-related increase of soluble and insoluble Aβ42 and Aβ40 levels were documented in the APP695SWEch+PS1/A246E mice (Marutle et al., 2002, Wang et al., 2003). Neuritic Aβ plaques in hippocampus and neocortex were surrounded by astrocytes and microglia (Borchelt et al., 1997). As mentioned above, APP695LD has been interbred with the PS1/A246E line (Van Dorpe et al., 2000). The same APP695SWEch model was bred with PS1/ΔE9 (line S-9), each transgene driven by the endogenous PrP promoter, resulting in Aβ plaques at 7 months of age (Jankowsky et al., 2005a; Perez et al., 2005; Savonenko et al., 2005). Due to Aβ deposits in striatum (Perez et al., 2005), neuronal loss has been reported there (Richner et al., 2009), along with decreased 5HT neurons in raphe and decreased noradrenergic neurons in locus coeruleus (Liu et al., 2008).

A co-injected bigenic model (line 85) contains the same 695-amino acid human/murine APP transgene with the Swedish mutation combined with a PS1/ΔE9 transgene driven by mouse Prp (Jankowsky et al., 2004). APP695SWE/co+PS1/ΔE9 mice harbor Aβ plaques at 6 months of age (Garcia-Alloza et al., 2006; Jankowsky et al., 2004) and possess fewer capillary segments than controls in neocortical white matter (Lee et al., 2005). Analogous to the previous model, 5HT fibers decreased in frontal cortex of the bigenics (Gimbel et al., 2010). In addition, APP751SWE mice were bred with a PS1/G384A line, both under control of the Thy1 promoter (Busche et al., 2008). A second co-injected line contains Swedish-mutated APP combined with a PS1/L166P mutation driven by the mouse Thy1 promoter, containing Aβ plaques and gliosis (Radde et al., 2006).

As mentioned in section 2.2.1, one model with the APP Swedish mutation express APP in skeletal muscle (Sugarman et al., 2002). These mice were crossed with homozygous PS1/M146V knock-in mice, forming a bigenic APP/SWE/Ckm model (Sugarman et al., 2002; Guo et al. 1999; Kitazawa et al. 2006), with higher than normal Aβ42 levels and phosphorylated tau in skeletal muscle with infiltrates of CD8-immunoreactive T cells (Kitazawa et al. 2006).

An APP751SWE/LD line with Swedish and London mutations on a transgene driven by the murine Thy1 promoter was crossed with a PS1/M146L line on a transgene driven by the human HMGCR (hydroxy-methyl-glutaryl-Coenzyme A reductase) promoter (Blanchard et al., 2003). The APP751SWE/LD +PS1/M146L mice have Aβ plaques as early as 2.5 months of age and age-related microglial activation, first with the appearance of a phagocytic followed by a cytotoxic phenotype (Jimenez et al., 2008), and pyramidal cell loss in hippocampus (Schmitz et al., 2004). The same APP751SWE/LD line was crossed with a PS1/M233T+L235P knockin, yielding a mutant also with Aβ plaque onset at 2.5 months of age, glial activation, and pyramidal cell loss in hippocampus (Casas et al., 2004), as well as a loss in dentate gyrus granule cells (Cotel et al., 2008) and CA1–2 thinning (Faure et al., 2011). In addition to hippocampal damage, the APP751SWE/LD +PS1/M233T+L235P model is marked by spinal axonopathy and thoracolumbar kyphosis, i.e. spine curvature (Wirths et al., 2008).

As mentioned above, the only model with Aβ plaques and neurofibrillary tangles is the triple APP/SWE+PS1/M146V+Mapt/P301L mutant (Oddo et al., 2003). The 3xTg-AD mice were generated by co-injecting APP with the Swedish mutation and four-repeat Mapt with the P301L mutation into single-cell embryos of PS1/M146V knockin mice. A five-fold transgenic model with Swedish/Florida/London mutations on APP and M146L and L286V mutations on PS1 has been generated with the murine Thy1 promoter (Oakley et al., 2006). The 5xFAD (Tg6799) transgenic mice have Aβ plaques as early as 2 months of age, accompanied by astrogliosis and microgliosis.

2.7 APP+PS2 mutations

To our knowledge, the only model fitting this category is the PS2 transgenic one carrying the N141I Volga-German mutation driven by the endogenous Prp promoter bred with Swedish mutated APP mice driven by the endogenous Thy1 promoter (Richards et al., 2003), yielding APP751SWE+PS2/VG (TgPS2APP) mice. The brain of the double mutant contains Aβ plaques surrounded by activated microglial cells and astrocytes.

3. Exploratory activity in Alzheimer mice

Several reviews have appeared on murine Alzheimer models, mainly useful in understanding the impact of amyloid formation on learning or neurobiological processes (Ashe, 2001; Ashe and Zahs, 2010; Duyckaerts et al., 2008; German and Eisch, 2004; Gimenez-Llort et al., 2007; Howlett and Richardson, 2009; Kokjohn and Roher, 2009; McGowan et al., 2006; Mineur et al., 2005; Morgan, 2003; Obulesu and Rao, 2010; Sant'Angelo et al., 2003; Scearce-Levie, 2011; Spires and Hyman, 2005; Van Dam et al., 2005; van Leuven, 2000; Wirths and Bayer, 2010), but none of the authors focused their attention on exploratory activity. In the present review, we analyze the effects of genetic modification of APP on open-field, Y- or T-maze, elevated plus-maze, and emergence tests. In view of the tendency of patients with Alzheimer's disease to be either apathetic or agitated (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004), murine models may be expected to be either hypoactive as a result of apathy or hyperactive as a result of agitation. Moreover, because apathy and disinhibition are linearly correlated in Alzheimer patients (Holthoff et al., 2005; Lyketsos et al., 2002), one may perhaps expect murine models to be hypoactive in one apparatus and hyperactive in another. Another possibility is to uncover opposite changes as a function of aging.

3.1 Open-field, Y-maze, and home-cage

The exploration of novel stimuli has long been assessed in the open-field (Walsh and Cummins, 1976) and Y-maze (King and Arendash, 2002). In contrast, the exploration of familiar stimuli can be examined in the home-cage (Ognibene et al., 2005). In all cases, the main measure used is the distance travelled. Alternative measures include time spent walking, sometimes recorded at different motion speeds. An IntelliCage has been developed, like the open-field consisting of an enclosed arena, but instead of being empty with various objects in it, also used for measuring activity over several months as the mouse's home-cage (Codita et al., 2010). As expected, both hypoactivity and hyperactivity have been reported in APP mutant mice relative to age-matched controls (Table 2).

Table 2.

Motor activity in open-field, Y-maze, or home-cage by transgenic mice expressing human mutated or wild-type (WT) APP genes with or without mutated PS1

Mouse name Activity References
APP695SWE (Tg2576) ↑ or normal Arendash et al., 2004;
Chapman et al., 1999;
Comery et al., 2005;
Deacon et al., 2008;
Dineley et al., 2002;
Gil-Bea et al., 2007;
Gorman and Yellon, 2010;
Holcomb et al., 1998;
1999;
King and Arendash 2002;
Lalonde et al., 2003;
Massaad et al., 2009;
Middei et al., 2004, 2006;
Ognibene et al., 2005
Scholtzova et al., 2009;
Tabuchi et al., 2009
APP695SWE (Tg2123H) normal Hsiao et al., 1995
APP695SWEch normal Savonenko et al., 2003
APP751SWE ↓ and ↑ or normal Dumont et al., 2004;
Heneka et al., 2006;
Huang et al., 2006;
Lalonde et al., 2002b;
Senechal et al., 2008
Van Dam et al., 2003;
Vloeberghs et al., 2004
APP695SWE/Thy1 normal Richardson et al., 2003
APP770SWE Moechars et al., 1999b
APP751SWE/LD ↑ or ↓ Blanchard et al., 2009
APP/IND ↑ or ↓ Dodart et al., 1999;
Gerlai et al., 2002
Huitron-Resendiz et al., 2002
APP695SWE/IND (TgCRND8 and Tg19959) ↑ or normal Ambrée et al., 2006;
Dumont et al., 2010;
Görtz et al., 2008
Hyde et al., 2005;
Lewejohann et al., 2009;
Touma et al., 2004;
Walker et al., 2011
APP695SWE/IND/tet Jankowsky et al., 2005b
APP751SWE/IND normal Lee et al., 2004
APP/SWE/IND/J20 normal Galvan et al., 2006
APP/SWE/ARC Codita et al., 2010
APP695LD Moechars et al., 1999b
APP/RK Moechars et al., 1998
APP695WT/Mt2 Yamaguchi et al., 1991
APP695WT/Thy1 Moechars et al., 1999b
App695WT APP695mycWT, APP695TRImyc normal Hsiao et al., 1995
APP751WT normal D'Hooge et al., 1996
APP/C104 normal Nalbantoglu et al., 1997
APP/C100-Flag normal Berger-Sweeney et al., 1999
APP/C100/IND Boon et al., 2010
APP/C100/WT Boon et al., 2010
APP/C99 Lalonde et al., 2002a
APP695SWE+PS1/M146L ↑ or normal Arendash et al., 2001;
Holcomb et al., 1998, 1999;
Sadowski et al., 2004
APP695SWE+PS1/M146L/Thy1 ↓ normal Pugh et al., 2007
Pardon et al., 2009
APP695SWEch+PS1/A246E Liu et al., 2002
APP751SWE/LD+PS1/M146L Le Cudennec et al., 2008
APP695SWE+PS1/A246E normal Dineley et al., 2002
APP695SWE/co+PS1/ΔE9 ↑ or normal Frye and Walf, 2008;
Hartmann et al., 2010;
Hoojimans et al., 2009;
Lalonde et al., 2004, 2005
Timmer et al., 2010
APP751SWE/LD+PS1/M233+L235P Faure et al., 2011
APP751SWE+PS2/VG normal Richards et al., 2003
3X-Tg-AD ↓ ↑ or normal Arsenault et al., 2011;
Gimenez-Llort et al., 2007;
Gulinello et al., 2009;
Halagappa et al., 2007;
Nelson et al., 2007;
Ojha et al., 2011;
Pietropaolo et al., 2008;
Pietropaolo et al., 2009
APP knockout Müller et al., 1994;
Tremml et al., 1998

↑=increase, ↓=decrease, normal=no difference vs controls; WT=wild-type

3.1.1 Hyperactivity

The most often examined Alzheimer model for motor activity is APP695SWE (Tg2576), often found to be more active than non-transgenic controls in the open-field (Deacon et al., 2008, 2009; King and Arendash, 2002; Gil-Bea et al., 2007; Lalonde et al., 2003; Middei et al., 2006; Tabuchi et al., 2009). The same results were reported in the Y-maze (King and Arendash, 2002) and home-cage (Gorman and Yellon, 2010; Ognibene et al., 2005). A positive linear correlation was obtained between open-field and Y-maze values, indicating a common underlying factor (Arendash and King, 2002), one of which may be Aβ-mediated inflammatory responses, since the hyperactivity was reversed after administration of the anti-inflammatory agent, ibuprofen (Lim et al., 2001). However, other experimenters reported no change with this model in the open-field (Arendash et al., 2004; Chapman et al., 1999; Comery et al., 2005; Dineley et al., 2002; Massaad et al., 2009; Scholtzova et al., 2009) or Y-maze (Arendash et al., 2004; Holcomb et al., 1998, 1999; Middei et al., 2004). The age factor is the most likely cause of this discrepancy, since hyperactivity was evident from 14 to 21 months (Deacon et al., 2008, 2009; King and Arendash, 2002; Lalonde et al., 2003; Middei et al., 2006), but generally not at younger ages (Arendash et al., 2004; Chapman et al., 1999; Comery et al., 2005; Holcomb 1998, 1999; Massaad et al., 2009; Middei et al., 2004), except for significant results at 7 (Middei et al., 2006; Tabuchi et al., 2009) and 10 (Tabuchi et al., 2009) months and one negative result at 16 (Scholtzova et al., 2009), implying the additional influence of methodologic factors related to the apparatus, such as lighting conditions, or else animal characteristics, such as different sex distributions or genetic backgrounds, the latter being due to different husbandry practices.

Since the monogenic generally displays age-related hyperactivity, it is no surprise to learn that the APP695SWE+PS1/M146L bigenic shows the same tendency, found in the Y-maze as early as 3 months of age, and also at 6 and 9 months (Holcomb et al., 1998, 1999), later confirmed at 5 and 15 months (Arendash et al., 2001). The latter group was also hyperactive in the open-field (Arendash et al., 2001). However, no change in activity was found with the same model evaluated at 8 and 22 months (Sadowski et al., 2004), reflecting task-mediated factors difficult at the present time to be ascertained.

Hyperactivity in the open-field has been reported in four other bigenic models, including APP751SWE/LD +PS1/M233T+L235P (Faure et al., 2011), APP695SWE/co+PS1/ΔE9 (line 85) (Hoojimans et al., 2009; Lalonde et al., 2005), APP751SWE+PS2/VG (Richards et al., 2003), and 3xTg-AD (Gimenez-Llort et al., 2007; Pietropaolo et al., 2008). In 3xTg-AD mice obtained by co-injecting APP with the Swedish mutation and four-repeat Mapt with the P301L mutation into single-cell embryos of PS1/M146V knockins, hyperactivity was found at 6 (Gimenez-Llort et al., 2007) and 12 (Gimenez-Llort et al., 2007; Pietropaolo et al., 2008) months, but, as described below, the opposite pattern of hypoactivity was prominent at different ages. In the APP695SWE/co+PS1/ΔE9 model, hyperactivity was reported at 12 (Lalonde et al., 2005) and 15 (Hoojimans et al., 2009) months of age, but the bigenics ran for longer distances only in the center region and not in the periphery of the open-field at the younger age (Lalonde et al., 2005). As with the APP695SWE model, variable results are found in younger APP695SWE/co+PS1/ΔE9 mice, with open-field hyperactivity detected at 8 months (Hoojimans et al., 2009), but not at 7 (Lalonde et al., 2004), 9 (Frye and Walf, 2008), or 10 (Hartmann et al., 2010). A negative finding was found even from 5 to 12 months, at least with respect to time spent walking (Timmer et al., 2010).

In view of the close neuropathologic resemblance between APP695SWE (Tg2576) and APP751SWE (APP23) models, it is not surprising to see hyperactivity in both, though once again an age-related pattern is discernable. Indeed, APP751SWE mice were more active than controls in the open-field at 3 (Senechal et al., 2008), 10 (Heneka et al., 2006), and 24 months (Dumont et al., 2004). The same result was found in the home-cage from 3 to 12 months (Van Dam et al., 2003; Vloeberghs et al., 2004). But the reverse pattern of less activity in the open-field was reported at 6 (Van Dam et al., 2003) and 16 (Lalonde et al., 2002b) months, with no change in the Y-maze at 3 months (Huang et al., 2006). It is likely that the change from mid-age hypoactivity (Lalonde et al., 2002b) to old-age hyperactivity (Dumont et al., 2004) in the same laboratory is mediated by amyloid-related action on synaptic activity of neurotransmitters, possibly decreased transmission of gamma-amino butyric acid (GABA) as a function of age, since only the older transgenics exhibited home-cage myoclonus and epileptic seizures associated with this neruotransmitter. Likewise, hypoactivity was converted to hyperactivity as a function of aging in APP751SWE/LD mutants from 2 to 19 months (Blanchard et al., 2009).

The finding of open-field hyperactivity is not limited to transgenic mice with the Swedish mutation, extending to the APP/IND model (PDAPP) with the Indiana mutation, this time at a very early age, either before (3 months of age) or after (6 and 9 months of age) Aβ plaque formation, the former being attributed to the direct action of mutated APP or to intracellular Aβ (Dodart et al., 1999). Three-month-old APP/IND mice were also hyperactive in their home-cage, although this effect disappeared at 20 months (Huitron-Resendiz et al., 2002). However, the reverse pattern of higher immobility time in this transgenic model was found at 11 months of age, though activity levels were not monitored as such (Gerlai et al., 2002).

Hyperactivity was also reported in two models with combined Swedish and Indiana mutations. As with the previous model, APP695SWE/IND mutants (TgCRND8 or Tg19959) were hyperactive in the open-field either before or after Aβ plaque deposition (Dumont et al., 2010; Hyde et al., 2005; Walker et al., 2011) and also in their home-cage (Ambrée et al., 2006). However, no effect was found in the open-field at 3 (Touma et al., 2004) or 4 (Görtz et al., 2008) months of age, a time when plaques are already apparent, perhaps illustrating once again that the phenotype is more apparent in older mice, either because of the long-term action of Aβ in the brain or because of the gradual decline in motor activity as a function of aging in normal mice. Moreover, no change in home-cage activity was discerned in a new procedure with radiofrequency detection (Lewejohann et al., 2009). In any event, hyperactivity was found in a second model with Swedish and Indiana mutations and Aβ plaques, namely APP695SWE/IND/tet mutants measured in their home-cage (Jankowsky et al., 2005b).

The same phenotype was discerned in a mutant with Swedish and Arctic mutations containing Aβ plaques, namely APP/SWE/ARC exposed for the first time to the IntelliCage, amounting to the open-field test (Codita et al., 2010). Moreover, since hyperactivity has been reported prior to Aβ plaque formation in models cited above, it is not surprising to find the same phenotype exhibited in two models which never develop Aβ plaques at all, namely APP/C100/IND and APP/C100/WT (Boon et al., 2010).

Since Aβ deposition or C100 overexpression in the limbic system is a prominent feature of most Alzheimer models, it is reasonable to hypothesize that neuronal dysfunction at this level contributes to hyperactivity. Indeed, hyperactivity has often been demonstrated in rats with lesions in the hippocampus (e.g. Roberts et al., 1962) or septum (Köhler and Srebro, 1980) relative to sham-operated controls. A second factor that should be further investigated is the coincident emergence of hyperactivity with myoclonic movements and seizure activity in APP751SWE mice (Dumont et al., 2004). These results point to a possible contribution of impaired GABAergic transmission in the hyperactive phenotype, since GABAA receptor antagonists increase motor activity in rats, for example picrotoxin injected at subconvulsive doses either in the periphery (Chang et al., 2004) or directly into the hippocampus (Bast et al., 2001), amygdala (Chang et al., 2004), or nucleus accumbens (Morgenstern et al., 1984).

It is probable that the hyperactive phenotype is more clearly discriminable in older mice, since the activity of normal mice declines with aging, as demonstrated in C57BL/6 (Dean et al., 1981; Sprott and Eleftheriou, 1974) and NMRI (Lamberty and Gower, 1990, 1992) strains. The phenotype may be obscured in younger mice because of a ceiling effect, i.e. the mutants cannot be demonstrated to be significantly more active because of the high values seen in non-transgenic controls, especially during brief assessments lasting 5 min, most often the case, and especially in particularly active strains such as C57BL/6.

3.1.2 Hypoactivity

In view of the apathy often displayed by patients with Alzheimer's disease (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004), one expects murine models to show hypoactivity, which may be attributed to Aβ-related changes in specific neurotransmitter systems. Moreover, since the App knockout is hypoactive (Müller et al., 1994; Tremml et al., 1998), different results with respect to hyper- or hypo-activity may be due to changes in the functioning of the endogenous Aβ peptide.

As expected, hypoactivity in the open-field has been described in several models, including two mutations with the Swedish mutation, APP751SWE (as cited in the previous subsection) (Lalonde et al., 2002b; Van Dam et al., 2003) and APP770SWE (Moechars et al., 1999b), one with the London mutation, APP695LD (Moechars et al., 1999b), and one with both Swedish and London mutations, APP751SWE/LD (Blanchard et al., 2009), together with four multigenic models, namely APP695SWEch+PS1/A246E (Liu et al., 2002), APP751SWE/LD+PS1/M146L (Le Cudennec et al., 2008), APP695SWE+PS1/M146L/Thy1 (Pugh et al., 2007), and 3xTg-AD (Arsenault et al., 2011; Gulinello et al., 2009; Halagappa et al., 2007; Nelson et al., 2007; Pietropaolo et al., 2009). Also as expected, in view of findings with the hyperactive phenotype, this pattern appears age-related, at least with respect to APP695SWE+PS1/M146L/Thy1 mutants, found to be hypoactive at 10 months of age (Pugh et al., 2007) but not at 4 (Pardon et al., 2009).

More complicated and difficult to be reconciled results have been obtained with the 3xTg-AD model. As mentioned in the previous subsection, hyperactivity was found at 6 (Gimenez-Llort et al., 2007) and 12 (Gimenez-Llort et al., 2007; Pietropaolo et al., 2008) months of age. Yet the opposite pattern of hypoactivity was noted at 6 (Pietropaolo et al., 2009) and 7 (Nelson et al., 2007) months, as well as 10 and 17 (Halagappa et al., 2007), 12 (Arsenault et al., 2011), or 15 (Gulinello et al., 2009) months, with no effect found at 8 (Ojha et al., 2011). The blatant discrepancies at 6 and 12 months may be due to test duration, as Gimenez-Llort et al. (2007) noted initial hypoactivity followed by a global pattern of hyperactivity. A more interesting neurobiological explanation concerns age-related changes in neurotransmitter systems yet to be uncovered. It is possible that the initial pattern of hyperactivity is converted to hypoactivity as a function of age, since hypoactivity but not hyperactivity has so far been reported in mice above 15 months.

As found with models showing hyperactivity, hypoactivity is also a prominent feature of models without Aβ plaques, notably APP/RK (Moechars et al., 1998), APP695WT/Thy1 (Moechars et al., 1999b), APP695WT/Mt2 (Yamaguchi et al., 1991), and APP/C99 (Lalonde et al., 2002a). These data point towards direct Aβ-mediated changes on neural activity as being reponsible for the hypoactive phenotype.

It remains to be determined which neurobiologic systems are responsible for amyloid-mediated hypoactivity. In view of the presence of Aβ plaques or APP overexpression in neocortex, it is reasonable to suppose a role for the prefrontal cortex in mediating this phenotype, since anomalies in prefronto-cortical-basal ganglia circuits are known to cause apathy (Levy and Dubois, 2006). Future experimentation should be guided by determining the association between hypoactivity and anomalies in prefrontal cortex-mediated functions. Another point to be considered, as mentioned in the previous subsection, is that mouse activity declines as a function of aging (Dean et al., 1981; Lamberty and Gower, 1990, 1992; Sprott and Eleftheriou, 1974). On one hand, the hypoactive phenotype may be more evident in younger mice, because they are more active at this age. On the other hand, it may take time before Aβ-mediated processes occur in prefrontal cortex or other brain regions, mitigating against the chance of finding an effect at a young age.

3.1.3 Unchanged activity

Since either hypo- or hyper-activity has been found in the open-field, sometimes with the same model (APP751, APP751SWE/LD+PS1/M146L, 3xTg-AD), it is not surprising to find no change in activity in several models, some with Aβ plaques, namely APP695SWE/Thy1 (Richardson et al., 2003), APP695SWEch (Savonenko et al., 2003), APP751SWE/IND (Lee et al., 2004), APP/SWE/IND/J20 (Galvan et al., 2006), APP695SWE+PS1/A246E (Dineley et al., 2002)., and APP/C104 (Nalbantoglu et al., 1997), some without Aβ plaques, namely APP751WT (D'Hooge et al., 1996) and APP/C100-Flag (Berger-Sweeney et al., 1999). Likewise, Y-maze activity was normal in APP/SWE/IND/J20 mice with Aβ plaques (Galvan et al., 2006) and APP695mycWT, APP695TRImyc, App695WT, and APP695SWE (Tg2123H) mice without Aβ plaques (Hsiao et al., 1995). In most cases, mice were tested at a single age level, thus mitigating against the chance of uncovering a significant effect.

3.1.4 Conclusions

Changes in activity levels irrespective of the direction are concordant with clinical findings of the behavioral and psychological symptoms of dementia (BPSD) (Lanari et al., 2006), apathy and agitation being prominent features of patients with Alzheimer's disease. Although such gross behavioral changes often occur late in dementia progression, these are correlated with caregiver compliance (Senanarong et al., 2004), and for this reason there is an ethical advantage if not a necessity in evaluating exploratory activity in murine models, including during treatment trials, usually limited to learning abilities.

In two murine models, APP751SWE (Dumont et al., 2004; Lalonde et al., 2002b) and APP751SWE/LD (Blanchard et al., 2009), age-related changes occurred from hypo- to hyper-activity. Similar data seem to apply with 3xTg-AD mice (Arsenault et al., 2011; Gimenez-Llort et al., 2007; Gulinello et al., 2009; Halagappa et al., 2007; Nelson et al., 2007; Pietropaolo et al., 2008, 2009), in need to be re-examined at a wider range within the same study to determine which occurs first. The most likely explanation for these findings is that amyloid exerts different neurochemical changes depending on age. We hypothesize that hyperactivity is the result of decreased transmission of GABAergic pathways, all the more likely in that APP751SWE mutants are susceptible to age-related myoclonus and convulsions linked with this neurotransmitter.

Age-related patterns in normal mice are likely to be relevant in determining the sensitivity of the mutation with respect to motor activity. The motor activity of normal mice decrease with age (Dean et al., 1981; Lamberty and Gower, 1990, 1992; Sprott and Eleftheriou, 1974). Thus, if the predominant phenotype in the transgenic is hyperactivity, it should be more apparent in older mice. But if the predominant phenotype is hypoactivity, it should be more apparent in younger mice.

3.2 Spontaneous alternation

After being placed in the stem of a Y- or T-maze and entering one of its arms, spontaneous alternation occurs on the subsequent trial when a mouse enters the opposite arm (Dember and Fowler, 1958; Lalonde, 2002). A cross-maze version may be used with two stems instead of one, with the mouse placed at either end (Jawhar et al., 2011). The spontaneous alternation test estimates the willingness to explore novel stimuli, or to avoid familiar stimuli. In view of the apathy often exhibited by patients with Alzheimer's disease (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004), one may predict reduced alternation rates in murine models. A second reason alternation rates may decline is the result of behavioral dishinbition or a loss in short-term memory, signs also manifest in Alzheimer dementia. In the former case, mice may perseverate as a result of impulsive behavior, choosing the first available arm without considering its previous choice.

As shown in Table 3, APP695SWE mutants (Tg2576) have often been described as being deficient in the spontaneous alternation test (Arendash et al., 2004; Deacon et al., 2008; Fukumoto et al., 2010; Holcomb et al., 1998, 1999; Hsiao et al., 1996; King and Arendash, 2002; Lalonde et al., 2003; Middei et al., 2004; Ognibene et al., 2005; Ohno et al., 2004). Logically enough, the same result applies to the APP695SWE+PS1/M146L bigenic derived from the monogenic (Holcomb et al., 1998, 1999). However, no such deficit in the bigenic was found in one study (Arendash et al., 2001), possibly due to the use of a different genetic background. Indeed, different normal mouse strains differ quite importantly in terms of alternation rates (Bertholet and Crusio, 1991).

Table 3.

Spontaneous alternation by transgenic mice expressing human mutated or wild-type (WT) APP genes with or without mutated PS1

↓=decrease; normal=no difference vs controls; WT=wild-type

Spontaneous alternation rates declined relative to controls in a second APP model with the Swedish mutation causing an accumulation of Aβ plaques, APP695SWE/Thy1 (Richardson et al., 2003). This occurred at 12 and 18 months of age, but not at 24. The negative result at 24 months is probably due the reduced rate of controls relative to that of younger mice. Indeed, a lower alternation rate as a function of aging is a common finding in normal mouse strains (Lalonde, 2002). A reduction in spontaneous alternation rates appeared in a third APP model with the Swedish mutation causing an accumulation of Aβ plaques, APP751SWE mice (APP23), found at 3 months of age (Huang et al., 2006), as were APP751SWE+PS1/G384A bigenics derived from this monogenic at 6 months of age (Busche et al., 2008). But no deficit was apparent in APP751SWE mice at 16 and 24 months of age, presumably at least in part because of the low values obtained in controls (Dumont et al., 2004; Lalonde et al., 2002b). Likewise, spontaneous alternation rates were lower in APP751SWE+PS2/VG mutants than controls at 4 months of age, but not at 8, 12, and 16 despite increasing plaque load (Richards et al., 2003). Thus, unlike the hyperactive phenotype discussed in section 3.1.1, this assay appears to lose sensitivity as a function of aging, the low rate in controls being liable to cause a floor effect, i.e. a statistically significant result is unlikely because the values of controls are too low.

Nevertheless, four other Aβ plaque-containing models have been shown to be impaired in this test, namely APP/SWE/IND/J20 (Galvan et al., 2006), APP695SWEch+PS1/A246E (Filali and Lalonde, 2009), 3x-Tg-AD (Carroll et al., 2007; Pietropaolo et al., 2009), and 5xFAD (Jawhar et al. 2011; Oakley et al., 2006). However, no such impairment was found in two others carrying Aβ plaques: APP695SWE/IND (Hyde et al., 2005) and APP695SWE/co+PS1/ΔE9 (Bonardi et al., 2011; Frye and Walf, 2008; Harrison et al., 2009a,b; Lalonde et al., 2004, 2005). These negative results appear despite findings of alternation deficits in four models without Aβ plaques, namely APP751WT (Moran et al., 1995), APP/C100/IND (Boon et al., 2010), App695WT (Hsiao et al., 1995), and APP695TRImyc (Hsiao et al., 1995), though not in three others: APP695SWEch (Savonenko et al., 2003), APP/C100/WT (Boon et al., 2010), and APP/C99 (Lalonde et al., 2002a).

The positive findings in mutants with or without Aβ plaques may be explained by amyloid-related dysfunction in brain regions critical for spontaneous alternation, including the hippocampus (Lalonde, 2002). Indeed, hippocampal lesions in rats have often been found to cause spontaneous alternation deficits relative to sham-operated controls (Johnson et al., 1977; Kirkby et al., 1967; Stevens and Cowey, 1973). Identical results have been reported in rats with lesions in the septum (Clody and Carlton, 1969), basal forebrain magnocellular nucleus (Murray and Fibiger, 1986), anterior thalamus (Aggleton et al., 1995), medial prefrontal cortex (Divac et al., 1975), and prelimbic cortex (Delatour and Gisquet-Verrier, 1996). These brain areas either accumulate amyloid in mutants or are liable to be dysfunctional as a result of interrupted brain circuitries secondary to amyloid accumulation, mainly involving the limbic system. The negative findings may be due to insufficient amyloid accumulation in these brain regions, age-related declines in controls, causing a floor effect, or to the use of a genetic background with non-optimal alternation rates.

3.2.2 Conclusions

The spontanenous alternation assay appears sensitive to amyloid loading in mice. These results may be due to an unwillingness to explore novel stimuli or to disinhibitory tendencies, akin to the apathy or disinhibition reported in patients with Alzheimer's disease (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004). Alzheimer patients are liable to perseverate in all sorts of behavior, either because of a lack of initiative in trying out new behaviors, a lack in curiosity, neophobia, or because of a tendency to choose impulsively actions often repeated in the past. Likewise, mice may perseverate as a result of a lack in curiosity, a fear of novel stimuli, or impulsiveness. One way to distinguish between lack in curiosity or impulsiveness as the underlying cause of perseveration is to measure choice latencies, the time taken before mice choose to enter left or right maze arms. One would expect higher choice latencies in apathetic mice and lower latencies in impulsive mice. In one of the rare experiments where this was done, APP695SWE mutants deficient in spontaneous alternation did not differ from controls in choice latencies despite being more active in the open-field (Lalonde et al., 2003). On one hand, this negative result may indicate that another factor was involved, such a loss in short-term memory. On the other hand, it is possible that multiple factors are involved, including a lack in motivation to explore and disinhibition, so that choice latencies might even out. In any event, this measure should be used more often as a way of gauging possible underlying causes.

Amyloid-related dysfunction in the limbic system and medial prefrontal cortex may be responsible for the alternation deficits in mutant mice. However, positive findings appear to occur within a narrow time-frame in the mouse's lifetime. Because deficits have been obtained at an early age in mutants with or without Aβ plaques, it seems that early testing is the optimal stragegy to be used for this assay. Aside from age, the absence of a deficit may be caused by differences in regional distribution of Aβ or to the use of a genetic background alternating at low rates.

3.3 Elevated plus-maze

Patients with Alzheimer's disease are subject to anxiety, but sometimes also to the opposite tendency of disinhibition (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004). One may therefore expect either hypoanxious or hyperanxious phenotypes in murine models of this condition. Anxiety levels may be examined in the elevated plus-maze, containing two wall-enclosed arms opposite two arms without walls (Cole and Rodgers, 1993). Open arm entries and duration reflect exploratory activity in an anxiogenic area, whereas enclosed arm entries and duration reflect exploratory activity in a safer one, the ratio between the two providing a specific measure of anxiety. This test has been pharmacologically validated in mice by comparing benzodiazepines versus placebo (Cole and Rodgers, 1993), though dependent on their genetic background (Rodgers et al., 2002). Table 4 illustrates APP mice that have been evaluated in the elevated plus-maze.

Table 4.

Plus-maze exploration by transgenic mice expressing human mutated or wild-type (WT) APP genes with or without mutated PS1

Mouse Anxiety References
APP695SWE Tg2576 ↓ or normal Arendash et al., 2004;
Gil-Bea et al., 2007;
Lalonde et al., 2003;
Lassalle et al., 2008;
Ognibene et al., 2005;
Tabuchi et al., 2009
APP751SWE mice APP23 ↓ or normal Dumont et al., 2004;
Lalonde et al., 2002b
Vloeberghs et al., 2007
APP695SWEch ↓ or normal Savonenko et al., 2003
APP695SWE/IND ↓ or normal Dumont et al., 2010;
Görtz et al., 2008
Touma et al., 2004
APP/SWE/IND/J20 Chin et al., 2005
APP/SWE/IND/J9 normal Chin et al., 2005
APP751SWE/IND normal Lee et al., 2004
APP751WT normal Moran et al., 1995
APP/C100/IND normal Boon et al., 2010
APP/C100/WT normal Boon et al., 2010
APP/C99/IND Lee et al., 2006
APP/C99 normal Lalonde et al., 2002a
APP695SWE+PS1/M146L normal Arendash et al., 2001
APP695SWE+PS1/M146L/Thy1 Pugh et al., 2007
APP751SWE/LD+PS1/M233+L235P Cotel et al., 2010;
Faure et al., 2011
APP751SWE/LD normal Blanchard et al. 2009;
Le Cudennec et al., 2008
APP695SWEch+PS1/A246E ↑ or normal Puoliväli et al., 2002;
Filali et al., 2011
APP695SWE/co+PS1/ΔE9 Line 85 Lalonde et al., 2004, 2005;
Reiserer et al., 2007
3x-Tg-AD ↑ or normal Pietropaolo et al., 2008, 2009
5xFAD Jawhar et al, 2011;2012

↑=increase, ↓=decrease, normal=no difference vs controls; WT=wild-type

3.3.1 Hypoanxious APP mice

Relative to non-transgenic controls, APP695SWE mutants (Tg2576) had higher open/total arm entries and duration at 17 months of age (Lalonde et al., 2003). The increase in open arm duration occurred in the same mutant at the same age on three different genetic backgrounds (Lassalle et al., 2008). The same increase in open arm exploration occurred at 7 months of age (Gil-Bea et al., 2007; Ognibene et al., 2005; Tabuchi et al., 2009), a time when Aβ plaques begin to form, but not at 5 months (Arendash et al., 2004).

A similar increase in open arm exploration was reported in seven other models with parenchymal Aβ plaques: APP751SWE (APP23) (Dumont et al., 2004), APP695SWE/IND (Dumont et al., 2010), APP/SWE/IND/J20 (Chin et al., 2005), APP695SWE/co+PS1/ΔE9 (Lalonde et al., 2004, 2005; Reiserer et al., 2007), APP695SWE+PS1/M146L/Thy1 (Pugh et al., 2007), APP751SWE/LD+PS1/M233T+L235P (Cotel et al., 2012; Faure et al., 2011), and 5xFAD (Jawhar et al, 2011, 2012). On two of these, age-dependent effects were found. APP695SWE/IND mutants explored the open arms more than controls at 8 months of age (Dumont et al., 2010), but, despite the presence of plaques at this age, not at 3 (Touma et al., 2004) or 4 (Görtz et al., 2008) months. Likewise, the open arm exploration of APP751SWE mice increased at 24 months of age (Dumont et al., 2004), but not at 3, 6, 12 (Vloeberghs et al., 2007), or 16 (Lalonde et al., 2002b) months. These results may be due to insufficient Aβ load in the younger animals, a notion supported by the finding that open arm duration increased in the APP/SWE/IND/J20 strain with high but not in the closely associated APP/SWE/IND/J9 strain with lower amyloid load (Chin et al., 2005).

3.3.2 Hyperanxious APP mice

In contrast to the previous models, APP695SWEch+PS1/A246E mice containing Aβ plaques had lower open arm durations than controls at 14 months of age (Puoliväli et al., 2002), though not at 6 months (Filali et al., 2011). The same pattern of elevated anxiety levels in this paradigm was reported in 3x-Tg-AD mice with Aβ plaques, though this result was found only in a female (Pietropaolo et al., 2009) and not a mixed (Pietropaolo et al., 2008) subgroup. A hyperanxious phenotype was also revealed in two other APP models, whose brains this time do not contain Aβ plaques, as open arm entries and duration diminished in APP/C99/IND mice (Lee et al., 2006), as did open arm entries in APP695SWEch mice, though in the latter only in females on one of two non-congenic lines (Savonenko et al., 2003).

3.3.3 Unchanged anxiety levels in APP mice

Anxiety levels were no different from non-transgenic controls in APP/C99 mutants not harboring Aβ plaques, as only enclosed arm entries decreased (Lalonde et al., 2002a). In a second model without Aβ plaques, APP751SWE/IND, both enclosed and open arm entries decreased relative to controls (Lee et al., 2004). Three other models without Aβ plaques did not differ from controls in in this test, namely APP/C100/IND (Boon et al., 2010), and APP/C100/WT (Boon et al., 2010), and APP751WT (Moran et al., 1995), as well as two models with Aβ plaques, namely APP695SWE+PS1/M146L (Arendash et al., 2001) and APP751SWE/LD (Blanchard et al. 2009; Le Cudennec et al., 2008).

3.3.4 Conclusions

Twelve APP models have been shown to be sensitive to anxiety levels in the elevated plus-maze, concordant with the usefulness of this test in modelling the behavioral and psychological symptoms of dementia (BPSD) (Lanari et al., 2006). As with the open-field test, results were found in either direction, in the form of reduced anxiety in eight cases and the opposite pattern in the other four. Unlike the open-field test, no single mutant has yet been reported to display both increases and decreases in exploratory behaviors as a function of age.

Changes in anxiety levels in the elevated plus-maze may be ascribed to alterations in the neurotransmission of GABA. Indeed, benzodiazepines increased open arm exploration in normal mice (Cole and Rodgers, 1993; Griebel et al., 2000; Lalonde and Strazielle, 2010), as did muscimol, the GABAA receptor agonist, injected either peripherally (Dalvi and Rodgers, 1996) or at the level of the central nucleus of the amygdala (Moreira et al., 2007). Moreover, a reduction in anxiety levels was found in rats injected with a benzodiazepine directly into the hippocampus (Menard and Treit, 2001). 5HT-related pathways are also involved on the basis of findings of reduced anxiety levels in mice exposed to the elevated plus-maze following injections of either 5HT1A (Cao and Rodgers, 1997; Lalonde and Strazielle, 2010; Nunes-de-Souza et al., 2000) or 5HT2 (Nic Dhonnchadha et al., 2003) receptor agonists. In particular, a reduction in anxiety levels was found in rats injected with a 5HT1A receptor agonist in the hippocampus (Menard and Treit, 1998). Moreover, lesion studies in rats have determined the importance of the septum in this test, as lesions of this brain region increased open arm exploration relative to sham-operated controls (Treit and Menard, 1997). Thus, altered circuitries in the limbic system, comprising hippocampus, septum, and amygdala, liable to Aβ accumulation in most Alzheimer mutants, are likely to be involved in changes of elevated plus-maze behavior. In addition, the influence of the neocortex in these behaviors is underlined by the finding of increased open arm exploration in rats with lesions of the medial prefrontal cortex (Gonzalez et al., 2000).

3.4 Emergence

The emergence test is a second method to screen anxiety in rodents (Holmes, 2001). Mice are placed inside a small enclosure and latencies before emerging into a larger anxiogenic one are measured. As with the elevated plus-maze, this test has been pharmacologically validated in mice injected with a benzodiazepine (Lalonde and Strazielle, 2010). The assay is also sensitive to gene inactivation of corticotropin releasing factor (Smith et al., 1998), 5HT (Tecott et al., 1998), and dopamine (Dulawa et al., 1999) receptors, as well as the GT2 subtype of GTPase activating protein (Schmalzigaug et al., 2009).

To our knowledge, this test has only been used once among APP models. APP751SWE mice had equivalent emergence latencies relative to non-transgenic controls despite differing from them in the elevated plus-maze, indicating that the two tests reflect different facets of anxiety (Dumont et al., 2004; Lalonde et al., 2002b).

3.5 Aggression

Increased hostility and aggression are part of the BPSD described in Alzheimer dementia (Lanari et al., 2006). Aggression in male mice may be evaluated by the resident-intruder test, whereby a mouse isolated for several weeks is exposed to an intruder in its home cage (File, 1980). Female mice cannot be used in this test because they do not attack intruders in their territory, defending only their young. Male APP695SWE mice (Tg2576) had shorter latencies before the first attack towards an intruder and initiated more attacks than age-matched male controls (Alexander et al., 2011). Likewise, APP/DU/Thy1 and APP/FL/Thy1 residents with amyloid angiopathy but without Aβ plaques in parenchymal regions attacked more often and more quickly intruders than non-transgenic controls (Kumar-Singh et al., 2000). Aggression levels also increased in APP695SWE+PS1/M146L/Thy1 mice (TgTASTPM) with Aβ plaques (Pugh et al., 2007) and APP/RK mice without Aβ plaques (Moechars et al., 1996) exposed to the same test. Thus, this test appears useful in the development of anti-aggressive drugs in dementia syndromes, in mice with or without Aβ plaques as well as those with amyloid angiopathy.

4. General conclusions

Willingness to initiate various behaviors, curiosity, anxiety, mood, and aggression are liable to be altered in dementia syndromes including Alzheimer's disease (Chung and Cummings, 2000; Daffner et al., 1992; Frisoni et al., 1999; Hart et al., 2003; Lanari et al., 2006; Lyketsos et al., 2002; Petry et al., 1988; Senanarong et al., 2004). Changes in the behavioral and psychological symptoms of dementia (BPSD) seen in Alzheimer patients (Lanari et al., 2006) can be mimicked in murine models. In particular, open-field activity, Y- or T-maze spontaneous alternation, exploration in the elevated plus-maze, and aggression are susceptible to be modified in APP mice relative to controls. However, changes in the open-field and elevated plus-maze occur in either direction. In some models, sometimes depending on age, open-field activity is increased, in others decreased. Likewise, open arm exploration in the elevated plus-maze may increase or descrease. The underlying reasons have only begun to be evaluated. There is a lack of in-depth analysis of neurotransmitter changes in association with those in behavior as a result of amyloid accumulation, in particular for GABA, 5HT, and dopamine.

Highlights.

/APP mutants display BPSD/

/APP mutants are impaired in spontaneous alternation/

/APP mutants display hyperactivity or hypoactivity/

/APP mutants display hyperanxiety or hypoanxiety/

/APPmutants show increased aggression/

Acknowledgements

This study was supported in part by the National Institutes of Health (AG030399) to KF and RL and Alzheimer's Association of America to KF. We thank Karen Minter, Debbie McCollum, and Linda Walter for assistance in preparing the text.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aggleton JP, Neave N, Nagle S, Sahgal A. A comparison of the effects of medial prefrontal, cingulate cortex, and cingulum bundle lesions on tests of spatial memory: evidence of a double dissociation between frontal and cingulum bundle contributions. J. Neurosci. 1995;15:7270–7281. doi: 10.1523/JNEUROSCI.15-11-07270.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alexander G, Hanna A, Serna V, Younkin L, Younkin S, Janus C. Increased aggression in males in transgenic Tg2576 mouse model of Alzheimer's disease. Behav. Brain Res. 2011;216:77–83. doi: 10.1016/j.bbr.2010.07.016. [DOI] [PubMed] [Google Scholar]
  3. Alexandru A, Jagla W, Graubner S, Becker A, Bäuscher C, Kohlmann S, Sedlmeier R, Raber KA, Cynis H, Rönicke R, Reymann KG, Petrasch-Parwez E, Hartlage-Rübsamen M, Waniek A, Rossner S, Schilling S, Osmand AP, Demuth HU, von Hörsten S. Selective hippocampal neurodegeneration in transgenic mice expressing small amounts of truncated Aβ is induced by pyroglutamate-Aβ formation. J. Neurosci. 2011;31:12790–12801. doi: 10.1523/JNEUROSCI.1794-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ambrée O, Touma C, Gortz N, Keyvani K, Paulius W, Palme R, Sachser N. Activity changes and marked stereotyped behavior precede Aβ pathology in TgCRND8 Alzheimer mice. Neurobiol. Aging. 2006;27:955–964. doi: 10.1016/j.neurobiolaging.2005.05.009. [DOI] [PubMed] [Google Scholar]
  5. Amieva H, Jacqmin-Gadda H, Orgogozo JM, Le Carret N, Helmer C, Letenneur L, Barberger-Gateau P, Fabrigoule C, Dartigues JF. The 9 year cognitive decline before dementia of the Alzheimer type: a prospective population-based study. Brain. 2005;128:1093–1111. doi: 10.1093/brain/awh451. [DOI] [PubMed] [Google Scholar]
  6. Andrä K, Abramowski D, Duke M, Probst A, Wiederhold KH, Bürki K, Goedert M, Sommer B, Staufenbiel M. Expression of APP in transgenic mice: a comparison of neuron-specific promoters. Neurobiol. Aging. 1996;17:183–190. doi: 10.1016/0197-4580(95)02066-7. [DOI] [PubMed] [Google Scholar]
  7. Arendash GW, King DL. Intra- and intertask relationships in a behavioral test battery given to Tg2576 transgenic mice and controls. Physiol. Behav. 2002;75:643–652. doi: 10.1016/s0031-9384(02)00640-6. [DOI] [PubMed] [Google Scholar]
  8. Arendash GW, King DL, Gordon MN, Morgan D, Hatcher JM, Hope CE, Diamond DM. Progressive, age-related behavioral impairments in transgenic mice carrying both mutant amyloid precursor protein and presenilin-1 transgenes. Brain Res. 2001;891:42–53. doi: 10.1016/s0006-8993(00)03186-3. [DOI] [PubMed] [Google Scholar]
  9. Arendash GW, Lewis J, Leighty RE, McGowan E, Cracchiolo JR, Hutton M, Garcia MF. Multi-metric behavioral comparison of APPsw and P301L models for Alzheimer's disease: linkage of poorer cognitive performance to tau pathology in forebrain. Brain Res. 2004;1012:29–41. doi: 10.1016/j.brainres.2004.02.081. [DOI] [PubMed] [Google Scholar]
  10. Arsenault D, Julien C, Tremblay C, Calon F. DHA improves cognition and prevents dysfunction of entorhinal cortex neurons in 3xTg-AD mice. PLoS One. 2011;6:e17397. doi: 10.1371/journal.pone.0017397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Ashe KH. Learning and memory in transgenic mice modeling Alzheimer's disease. Learn Mem. 2001;8:301–308. doi: 10.1101/lm.43701. [DOI] [PubMed] [Google Scholar]
  12. Ashe KH, Zahs KR. Probing the biology of Alzheimer's disease in mice. Neuron. 2010;66:631–645. doi: 10.1016/j.neuron.2010.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Askanas V, Engel WK. Sporadic inclusion-body myositis and hereditary inclusion-body myopathies: current concepts of diagnosis and pathogenesis. Curr. Opin. Rheumatol. 1998;10:530–542. doi: 10.1097/00002281-199811000-00005. [DOI] [PubMed] [Google Scholar]
  14. Askanas V, Engel WK. Inclusion-body myositis: newest concepts of pathogenesis and relation to aging and Alzheimer's disease. J. Neuropathol. Exp. Neurol. 2001;60:1–14. doi: 10.1093/jnen/60.1.1. [DOI] [PubMed] [Google Scholar]
  15. Askanas V, Engel WK. Inclusion-body myositis and myopathies: different etiologies, possibly similar pathogenic mechanisms. Curr. Opin. Neurol. 2002;15:525–531. doi: 10.1097/00019052-200210000-00002. [DOI] [PubMed] [Google Scholar]
  16. Bast T, Zhang WN, Feldon J. Hyperactivity, decreased startle reactivity, and disrupted prepulse inhibition following disinhibition of the rat ventral hippocampus by the GABAA receptor antagonist picrotoxin. Psychopharmacology. 2001;156:225–233. doi: 10.1007/s002130100775. [DOI] [PubMed] [Google Scholar]
  17. Beckmann N, Gérard C, Abramowski D, Cannet C, Staufenbiel M. Noninvasive magnetic resonance imaging detection of cerebral amyloid angiopathy-related microvascular alterations using superparamagnetic iron oxide particles in APP transgenic mouse models of Alzheimer's disease: application to passive Aβ immunotherapy. J. Neurosci. 2011;31:1023–1031. doi: 10.1523/JNEUROSCI.4936-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bellucci A, Luccarini I, Scali C, Prosperi C, Giovannini MG, Pepeu G, Casamenti F. Cholinergic dysfunction, neuronal damage and axonal loss in TgCRND8 mice. Neurobiol. Dis. 2006;23:260–272. doi: 10.1016/j.nbd.2006.03.012. [DOI] [PubMed] [Google Scholar]
  19. Benzing WC, Wujek JR, Ward EK, Shaffer D, Ashe KH, Younkin SG, Brunden KR. Evidence for glial-mediated inflammation in aged APPSW transgenic mice. Neurobiol. Aging. 1999;20:581–589. doi: 10.1016/s0197-4580(99)00065-2. [DOI] [PubMed] [Google Scholar]
  20. Berger-Sweeney J, McPhie DL, Arters JA, Greenan J, Oster-Granite ML, Neve RL. Impairments in learning and memory accompanied by neurodegeneration in mice transgenic for the carboxyl-terminus of the amyloid precursor protein. Mol. Brain Res. 1999;66:150–162. doi: 10.1016/s0169-328x(99)00014-5. [DOI] [PubMed] [Google Scholar]
  21. Bertholet JY, Crusio WE. Spatial and non-spatial spontaneous alternation and hippocampal mossy fibre distribution in nine inbred mouse strains. Behav. Brain Res. 1991;43:197–202. doi: 10.1016/s0166-4328(05)80071-3. [DOI] [PubMed] [Google Scholar]
  22. Blanchard V, Decorte L, Noguès X, Micheau J. Characterization of cognition alteration across the course of the disease in APP751SL mice with parallel estimation of cerebral Abeta deposition. Behav. Brain Res. 2009;201:147–157. doi: 10.1016/j.bbr.2009.02.005. [DOI] [PubMed] [Google Scholar]
  23. Blanchard V, Moussaoui S, Czech C, Touchet N, Bonici B, Planche M, Canton T, Jedidi I, Gohin M, Wirths O, Bayer TA, Langui D, Duyckaerts C, Tremp G, Pradier L. Time sequence of maturation of dystrophic neurites associated with Aβ deposits in APP/PS1 transgenic mice. Exp. Neurol. 2003;184:247–263. doi: 10.1016/s0014-4886(03)00252-8. [DOI] [PubMed] [Google Scholar]
  24. Bonardi C, de Pulford F, Jennings D, Pardon M-C. A detailed analysis of the early context extinction deficits seen in APPswe/PS1dE9 female mice and their relevance to preclinical Alzheimer's disease. Behav. Brain Res. 2011;222:89–97. doi: 10.1016/j.bbr.2011.03.041. [DOI] [PubMed] [Google Scholar]
  25. Bondolfi L, Calhoun M, Ermini F, Kuhn HG, Wiederhold K-H, Walker L, Staufenbiel M, Jucker M. Amyloid-associated neuron loss and gliogenesis in the neocortex of amyloid precursor protein transgenic mice. J. Neurosci. 2002;22:515–522. doi: 10.1523/JNEUROSCI.22-02-00515.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Boon WC, van den Buuse M, Wegener N, Martin S, Chua HK, Bush AI, Masters CL, Adlard PA, Li QX. Behavioural phenotype of APPC100.V717F transgenic mice over-expressing a mutant Aβ-bearing fragment is associated with reduced NMDA receptor density. Behav Brain Res. 2010;209:27–35. doi: 10.1016/j.bbr.2010.01.013. [DOI] [PubMed] [Google Scholar]
  27. Borchelt DR, Ratovitski T, van Lare J, Lee MK, Gonzales V, Jenkins NA, Copeland NG, Price DL, Sisodia SS. Accelerated amyloid deposition in the brains of transgenic mice coexpressing mutant presenilin 1 and amyloid precursor proteins. Neuron. 1997;19:939–945. doi: 10.1016/s0896-6273(00)80974-5. [DOI] [PubMed] [Google Scholar]
  28. Borchelt DR, Thinakaran G, Eckman CB, Lee MK, Davenport F, Ratovitsky T, Prada CM, Kim G, Seekins S, Yager D, Slunt HH, Wang R, Seeger M, Levey AI, Gandy SE, Copeland NG, Jenkins NA, Price DL, Younkin SG, Sisodia SS. Familial Alzheimer's disease-linked presenilin 1 variants elevate Aβ1–42/1–40 ratio in vitro and in vivo. Neuron. 1996;17:1005–1013. doi: 10.1016/s0896-6273(00)80230-5. [DOI] [PubMed] [Google Scholar]
  29. Bornemann KD, Wiederhold K-H, Pauli C, Ermini F, Stalder M, Schnell L, Sommer B, Jucker M, Staufenbiel M. Aβ-induced inflammatory processes in microglia cells of APP23 transgenic mice. Am. J. Pathol. 2001;158:63–73. doi: 10.1016/s0002-9440(10)63945-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Busche MA, Eichhoff G, Adelsberger H, Abramowski D, Wiederhold KH, Haass C, Staufenbiel M, Konnerth A, Garaschuk O. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer's disease. Science. 2008;321:1686–1699. doi: 10.1126/science.1162844. [DOI] [PubMed] [Google Scholar]
  31. Calhoun ME, Wiederhold K-H, Abramowski D, Phinney AL, Probst A, Sturchler-Pierrat C, Staufenbiel M, Sommer B, Jucker M. Neuron loss in APP transgenic mice. Nature. 1999;395:755–756. doi: 10.1038/27351. [DOI] [PubMed] [Google Scholar]
  32. Calhoun ME, Burgermeister P, Phinney AL, Stalder M, Tolnay M, Wiederhold K-H, Abramaowski D, Sturchler-Pierrat C, Sommer B, Staufenbiel M, Jucker M. Neuronal overexpression of mutant amyloid precursor protein in prominent deposition of cerebrovascular amyloid. Proc. Natl. Acad. Sci. U.S.A. 1998;96:14088–14093. doi: 10.1073/pnas.96.24.14088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Cao BJ, Rodgers RJ. Comparative behavioural profiles of buspirone and its metabolite 1-(2-pyrimidinyl)-piperazine (1-PP) in the murine elevated plus-maze. Neuropharmacology. 1997;36:1089–1097. doi: 10.1016/s0028-3908(97)00094-4. [DOI] [PubMed] [Google Scholar]
  34. Carroll JC, Rosario ER, Chang L, Stanczyk FZ, Oddo S, LaFerla FM, Pike CJ. Progesterone and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice. J. Neurosci. 2007;27:13357–13365. doi: 10.1523/JNEUROSCI.2718-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Casas C, Sergeant N, Itier JM, Blanchard V, Wirths O, van der Kolk N, Vingtdeux V, van de Steeg E, Ret G, Canton T, Drobecq H, Clark A, Bonici B, Delacourte A, Benavides J, Schmitz C, Tremp G, Bayer TA, Benoit P, Pradier L. Massive CA1/2 neuronal loss with intraneuronal and N-terminal truncated Aβ42 accumulation in a novel Alzheimer transgenic model. Am. J. Pathol. 2004;165:1289–1300. doi: 10.1016/s0002-9440(10)63388-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Chang CK, Wang NL, Lin MT. Inhibition of the dopamine system in rat amygdala attenuates the picrotoxin-induced locomotor hyperactivity and hypertension. Clin. Exp. Pharmacol. Physiol. 2004;31:284–288. doi: 10.1111/j.1440-1681.2004.03994.x. [DOI] [PubMed] [Google Scholar]
  37. Chapman PF, White GL, Jones MW, Cooper-Blacketer D, Marshall VJ, Irizarry M, Younkin L, Good MA, Bliss TVP, Hyman BT, Younkin SG, Hsiao KK. Impaired synaptic plasticity and learning in aged amyloid precursor protein transgenic mice. Nature Neurosci. 1999;2:271–276. doi: 10.1038/6374. [DOI] [PubMed] [Google Scholar]
  38. Chin J, Palop JJ, Puolivali J, Massaro C, Bien-Ly N, Gerstein H, Scearce-Levie K, Masliah E, Mucke L. Fyn kinase induces synaptic and cognitive impairments in a transgenic mouse model of Alzheimer's disease. J. Neurosci. 2005;25:9694–9703. doi: 10.1523/JNEUROSCI.2980-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Chishti MA, Yang D-S, Janus C, Phinney AL, Horne P, Pearson J, Strome R, Zuker N, Loukides J, French J, Turner S, Lozza G, Grilli M, Kunicki S, Morissette C, Paquette J, Gervais F, Bergeron C, Fraser PE, Carlson GA, St. George-Hyslop P, Westaway D. Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid protein 695. J. Biol. Chem. 2001;276:21562–21570. doi: 10.1074/jbc.M100710200. [DOI] [PubMed] [Google Scholar]
  40. Christie R, Yamada M, Moskowitz M, Hyman B. Structural and functional disruption of vascular smooth cells in a transgenic mouse model of amyloid angiopathy. Am. J. Pathol. 2001;158:1065–1071. doi: 10.1016/S0002-9440(10)64053-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Chung JA, Cummings JL. Neurobehavioral and neuropsychiatric symptoms in Alzheimer's disease. Neurol. Clin. 2000;18:829–846. doi: 10.1016/s0733-8619(05)70228-0. [DOI] [PubMed] [Google Scholar]
  42. Clody DE, Carlton PL. Behavioral effects of lesions of the medial septum of rats. J. Comp. Physiol. Psychol. 1969;67:344–351. doi: 10.1037/h0026756. [DOI] [PubMed] [Google Scholar]
  43. Codita A, Gumucio A, Lannfelt L, Gellerfors P, Winblad B, Mohammed AH, Nilsson LN. Impaired behavior of female tg-ArcSwe APP mice in the IntelliCage: A longitudinal study. Behav. Brain Res. 2010;215:83–94. doi: 10.1016/j.bbr.2010.06.034. [DOI] [PubMed] [Google Scholar]
  44. Cole JC, Rodgers RJ. An ethological analysis of the effects of chlordiazepoxide and bretazenil (Ro 16-6028) in the murine elevated plus-maze. Behav. Pharmacol. 1993;4:573–580. [PubMed] [Google Scholar]
  45. Cole GM, Frautschy SA. Animal models for Alzheimer's disease. Alz. Dis. Rev. 1997;2:2–10. [Google Scholar]
  46. Collette F, Van der Linden M, Salmon E. Executive dysfunction in Alzheimer's disease. Cortex. 1999;35:57–72. doi: 10.1016/s0010-9452(08)70785-8. [DOI] [PubMed] [Google Scholar]
  47. Comery TA, Martone RL, Aschmies S, Atchison KP, Diamantidis G, Gong X, Zhou H, Kreft AF, Pangalos MN, Sonnenberg-Reines J, Jacobsen JS, Marquis KL. Acute γ-secretase inhibition improves contextual fear conditioning in the Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 2005;25:8898–8902. doi: 10.1523/JNEUROSCI.2693-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Cotel MC, Bayer TA, Wirths O. Age-dependent loss of dentate gyrus granule cells in APP/PS1KI mice. Brain Res. 2008;1222:207–213. doi: 10.1016/j.brainres.2008.05.052. [DOI] [PubMed] [Google Scholar]
  49. Cotel MC, Jawhar S, Christensen DZ, Bayer TA, Wirths O. Environmental enrichment fails to rescue working memory deficits, neuron loss, and neurogenesis in APP/PS1KI mice. Neurobiol. Aging. 2012;33:96–107. doi: 10.1016/j.neurobiolaging.2010.02.012. [DOI] [PubMed] [Google Scholar]
  50. Daffner KR, Scinto LFM, Weintraub S, Guinessey JE, Mesulam MM. Diminished curiosity in patients with probable Alzheimer's disease as measured by exploratory eye movements. Neurology. 1992;42:320–328. doi: 10.1212/wnl.42.2.320. [DOI] [PubMed] [Google Scholar]
  51. Dalvi A, Rodgers RJ. GABAergic influences on plus-maze behaviour in mice. Psychopharmacology. 1996;128:380–397. doi: 10.1007/s002130050148. [DOI] [PubMed] [Google Scholar]
  52. Davis J, Xu F, Deane R, Romanov G, Previti ML, Zeigler K, Zlokovic BV, Van Nostrand WE. Early-onset and robust cerebral microvascular accumulation of amyloid β-protein in transgenic mice expressing low levels of a vasculotropic Dutch/Iowa mutant form of β-amyloid precursor. J. Biol. Chem. 2004;279:20296–20306. doi: 10.1074/jbc.M312946200. [DOI] [PubMed] [Google Scholar]
  53. Deacon RMJ, Koros E, Bornemann KD, Rawlins JN. Aged Tg2576 mice are impaired on social memory and open field habituation tests. Behav. Brain Res. 2009;197:466–468. doi: 10.1016/j.bbr.2008.09.042. [DOI] [PubMed] [Google Scholar]
  54. Deacon RMJ, Cholerton LL, Talbot K, Nair-Roberts RG, Sanderson DJ, Romberg C, Koros E, Bornemann KD, Rawlins JNP. Age-dependent and -independent behavioral deficits in Tg2576 mice. Behav. Brain Res. 2008;189:126–138. doi: 10.1016/j.bbr.2007.12.024. [DOI] [PubMed] [Google Scholar]
  55. Dean RL, III, Scozzafava J, Goas JA, Regan B, Beer B, Bartus RT. Age-related differences in behavior across the life span of the C57BL/6J mouse. Exp. Aging Res. 1981;7:427–451. doi: 10.1080/03610738108259823. [DOI] [PubMed] [Google Scholar]
  56. Delatour B, Gisquet-Verrier P. Prelimbic cortex specific lesions disrupt delayed-variable response tasks in the rat. Behav. Neurosci. 1996;110:1282–1298. doi: 10.1037//0735-7044.110.6.1282. [DOI] [PubMed] [Google Scholar]
  57. Dember WN, Fowler H. Spontaneous alternation behavior. Psychol. Bull. 1958;55:412–428. doi: 10.1037/h0045446. [DOI] [PubMed] [Google Scholar]
  58. Dewachter I, van Dorpe J, Spittaels K, Tesseur I, Van Den Haute C, Moechars D, Van Leuven F. Modeling Alzheimer's disease in transgenic mice: effect of age and of presenilin on amyloid biochemistry and pathology in APP/London mice. Exp. Gerontol. 2000a;35:831–841. doi: 10.1016/s0531-5565(00)00149-2. [DOI] [PubMed] [Google Scholar]
  59. Dewachter I, Van Dorpe J, Smeijers L, Gilis M, Kuipéri C, Laenen I, Caluwaerts D, Checler F, Vanderstichele H, Van Leuven F. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. Eur. J. Neurosci. 2000b;20:6452–6458. doi: 10.1523/JNEUROSCI.20-17-06452.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. D'Hooge R, Nagels G, Mucke L, De Deyn PP. Spatial learning deficit in mice expressing human 751-amino acid β-amyloid precursor protein. NeuroReport. 1996;7:2807–2811. doi: 10.1097/00001756-199611040-00080. [DOI] [PubMed] [Google Scholar]
  61. Dineley KT, Xia X, Bui D, Sweatt JD, Zheng H. Accelerated plaque accumulation, associative learning deficits, and up-regulation of α7 nicotinic receptor protein in transgenic mice co-expressing mutant human presenilin 1 and amyloid precursor proteins. J. Biol. Chem. 2002;277:22768–22780. doi: 10.1074/jbc.M200164200. [DOI] [PubMed] [Google Scholar]
  62. Divac I, Wikmark RG, Gade A. Spontaneous alternation in rats with lesions in the frontal lobes: an extension of the frontal lobe syndrome. Physiol. Psychol. 1975;3:29–42. [Google Scholar]
  63. Dodart J-C, Mathis C, Bales KR, Paul SM. Does my mouse have Alzheimer's disease? Genes Brain Behav. 2002;1:142–155. doi: 10.1034/j.1601-183x.2002.10302.x. [DOI] [PubMed] [Google Scholar]
  64. Dodart J-C, Meziane H, Mathis C, Bales KR, Paul SM, Ungerer A. Behavioral disturbances in transgenic mice overexpressing the V717F β-amyloid precursor protein. Behav. Neurosci. 1999;113:982–990. doi: 10.1037//0735-7044.113.5.982. [DOI] [PubMed] [Google Scholar]
  65. Domnitz SB, Robbins EM, Hoang AW, Garcia-Alloza M, Hyman BT, Rebeck GW, Greenberg SM, Bacskai BJ, Frosch MP. Progression of cerebral amyloid angiopathy in transgenic mouse models of Alzheimer disease. J. Neuropathol. Exp. Neurol. 2005;64:588–594. doi: 10.1097/01.jnen.0000171644.00180.fc. [DOI] [PubMed] [Google Scholar]
  66. Duff K. Transgenic mouse models of Alzheimer's disease. In: Crusio W, Gerlai R, editors. Handbook of Molecular-genetic Techniques for Brain and Behavior Research (Techniques in the Behavioral and Neural Sciences, vol 13) Amsterdam: Elsevier; 1999. pp. 888–894. [Google Scholar]
  67. Duff K, Eckman C, Zehr C, Yu X, Prada C-M, Perez-tur J, Hutton J, Buee L, Harigaya Y, Yager D, Morgan D, Gordon MN, Holcomb L, Refolo L, Zenk B, Hardy J, Younkin S. Increased β-amyloid42(43) in brains of mice expressing mutant presenilin 1. Nature. 1996;383:710–713. doi: 10.1038/383710a0. [DOI] [PubMed] [Google Scholar]
  68. Dulawa SC, Grandy DK, Low MJ, Paulus MP, Geyer MA. Dopamine D4 receptor-knockout mice exhibit reduced exploration of novel stimuli. J. Neurosci. 1999;19:9550–9556. doi: 10.1523/JNEUROSCI.19-21-09550.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Dumont M, Strazielle C, Staufenbiel M, Lalonde R. Spatial learning and exploration of environmental stimuli in 24-month-old female APP23 transgenic mice with the Swedish mutation. Brain Res. 2004;1024:113–121. doi: 10.1016/j.brainres.2004.07.052. [DOI] [PubMed] [Google Scholar]
  70. Dumont M, Wille E, Calingasan NY, Nathan C, Beal MF, Lin MT. N-iminoethyl-L-lysine improves memory and reduces amyloid pathology in a transgenic mouse model of amyloid deposition. Neurochem. Int. 2010;56:345–351. doi: 10.1016/j.neuint.2009.11.006. [DOI] [PubMed] [Google Scholar]
  71. Duyckaerts C, Potier MC, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta Neuropathol. 2008;115:5–38. doi: 10.1007/s00401-007-0312-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJM, Rozemuller JM, Veerhuis R, Williams A. Neuroinflammation in Alzheimer's disease and prion disease. Glia. 2002;40:232–239. doi: 10.1002/glia.10146. [DOI] [PubMed] [Google Scholar]
  73. Eslinger PJ, Damasio AR. Preserved motor learning in Alzheimer's disease: implications for anatomy and behavior. J. Neurosci. 1986;6:3006–3009. doi: 10.1523/JNEUROSCI.06-10-03006.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Faure A, Verret L, Bozon B, El Tannir El Tayara N, Ly M, Kober F, Dhenain M, Rampon C, Delatour B. Impaired neurogenesis, neuronal loss, and brain functional deficits in the APPxPS1-Ki mouse model of Alzheimer's disease. Neurobiol. Aging. 2011;32:407–418. doi: 10.1016/j.neurobiolaging.2009.03.009. [DOI] [PubMed] [Google Scholar]
  75. Filali M, Lalonde R. Age-related cognitive decline and nesting behavior in an APPswe/PS1 bigenic model of Alzheimer's disease. Brain Res. 2009;1292:93–99. doi: 10.1016/j.brainres.2009.07.066. [DOI] [PubMed] [Google Scholar]
  76. Filali M, Lalonde R, Rivest S. Anomalies in social behaviors and exploratory activities in an APPswe/PS1 mouse model of Alzheimer's disease. Physiol. Behav. 2011;104:880–885. doi: 10.1016/j.physbeh.2011.05.023. [DOI] [PubMed] [Google Scholar]
  77. File SE. The use of social interaction as a method for detecting anxiolytic activity of chlordiazepoxide-like drugs. J. Neurosci. Meth. 1980;2:239–252. doi: 10.1016/0165-0270(80)90012-6. [DOI] [PubMed] [Google Scholar]
  78. Frackowiak J, Mazur-Kolecka B, Kaczmarski W, Dickson D. Deposition of Alzheimer's disease vascular amyloid-β is associated with decreased expression of brain L-3-hydroxyacyl-coenzyme A dehydrogenase (ERAB) Brain Res. 2001;907:44–53. doi: 10.1016/s0006-8993(01)02497-0. [DOI] [PubMed] [Google Scholar]
  79. Frisoni GB, Rozzini L, Gozetti A, Binetti G, Zanetti O, Bianchetti A, Trabucchi M, Cummings JL. Behavioral syndromes in Alzheimer’s disease: description and correlates. Dement. Geriatr. Cogn. Disord. 1999;10:130–138. doi: 10.1159/000017113. [DOI] [PubMed] [Google Scholar]
  80. Frye CA, Walf AA. Effects of progesterone administration and APPswe+PSEN1ΔE9 mutation for cognitive performance of mid-aged mice. Neurobiol. Learn. Mem. 2008;89:17–26. doi: 10.1016/j.nlm.2007.09.008. [DOI] [PubMed] [Google Scholar]
  81. Fryer JD, Taylor JW, DeMattos RB, Bales KR, Paul SM, Parsadanian M, Holtzman DM. Apolipoprotein E markedly facilitates age-dependent cerebral amyloid angiopathy and spontaneous hemorrhage in amyloid precursor protein transgenic mice. J. Neurosci. 2003;23:7889–7896. doi: 10.1523/JNEUROSCI.23-21-07889.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Fukumoto H, Takahashi H, Tarui N, Matsui J, Tomita T, Hirode M, Sagayama M, Maeda R, Kawamoto M, Hirai K, Terauchi J, Sakura Y, Kakihana M, Kato K, Iwatsubo T, Miyamoto M. A noncompetitive BACE1 inhibitor TAK-070 ameliorates Abeta pathology and behavioral deficits in a mouse model of Alzheimer's disease. J. Neurosci. 2010;30:11157–11166. doi: 10.1523/JNEUROSCI.2884-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Fukuchi K, Li L, Hart M, Lindsey JR. Accumulation of amyloid-β protein in exocrine glands of transgenic mice overexpressing a carboxyl terminal portion of amyloid protein precursor. Int. J. Exp. Pathol. 2000;81:231–239. doi: 10.1046/j.1365-2613.2000.00156.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Fukuchi K, Pham D, Hart M, Li L, Lindsey JR. Amyloid-β deposition in skeletal muscle of transgenic mice: possible model of inclusion body myopathy. Am. J. Pathol. 1998;153:1687–1693. doi: 10.1016/s0002-9440(10)65682-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Fukuchi K, Sopher B, Furlong CE, Smith AC, Dang N, Martin GM. Selective neurotoxicity of COOH-terminal fragments of the β-amyloid-precursor protein. Neurosci. Lett. 1993;154:145–148. doi: 10.1016/0304-3940(93)90192-n. [DOI] [PubMed] [Google Scholar]
  86. Fukuchi K, Ho L, Younkin SG, Kunkel DD, Ogburn CE, LeBoeuf RC, Furlong CE, Deeb SS, Nochlin D, Wegiel J, Wisniewski HM, Martin GM. High levels of circulating β-amyloid peptide do not cause cerebral β-amyloidosis in transgenic mice. Am. J. Pathol. 1996;149:219–227. [PMC free article] [PubMed] [Google Scholar]
  87. Galvan V, Gorostiza OF, Banwait S, Ataie M, Logvinova AV, Sitaraman S, Carlson E, Sagi SA, Chevallier N, Jin K, Greenberg DA, Bredesen DE. Reversal of Alzheimer's-like pathology and behavior in human APP transgenic mice by mutation of Asp664. Proc. Natl. Acad. Sci. U.S.A. 2006;103:7130–7135. doi: 10.1073/pnas.0509695103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Games D, Adams D, Alessandri R, Barbour R, Berthelette P, Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F, Guido T, Hagopian S, Johnson-Wood K, Khan K, Lee M, Leibowitz P, Lieberburg I, Little S, Masliah E, McConlogue L, Montoya-Zavala M, Mucke L, Paganini L, Penniman E, Power M, Schenk D, Seubert P, Snyder B, Soriano F, Tan H, Vitale J, Wadworth S, Wolozin B, Zhao J. Alzheimer-type neuropathology in transgenic mice overexpressing V717F β-amyloid precursor protein. Nature. 1995;373:523–527. doi: 10.1038/373523a0. [DOI] [PubMed] [Google Scholar]
  89. Gandy S, Simon AJ, Steele JW, Lublin AL, Lah JJ, Walker LC, Levey AI, Krafft GA, Levy E, Checler F, Glabe C, Bilker WB, Abel T, Schmeidler J, Ehrlich ME. Days to criterion as an indicator of toxicity associated with human Alzheimer amyloid-β oligomers. Ann. Neurol. 2010;68:220–230. doi: 10.1002/ana.22052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Garcia-Alloza M, Robbins EM, Zhang-Nunes SX, Purcell SM, Betensky RA, Raju S, Prada C, Greenberg SM, Bacskai BJ, Frosch MP. Characterization of amyloid deposition in the APPswe/PS1dE9 mouse model of Alzheimer disease. Neurobiol. Dis. 2006;24:516–524. doi: 10.1016/j.nbd.2006.08.017. [DOI] [PubMed] [Google Scholar]
  91. Gerlai R, Fitch T, Bales KR, Gitter BD. Behavioral impairment of APPV717F mice in fear conditioning: is it only cognition? Behav. Brain Res. 2002;136:503–509. doi: 10.1016/s0166-4328(02)00198-5. [DOI] [PubMed] [Google Scholar]
  92. German DC, Eisch AJ. Mouse models of Alzheimer's disease: insight into treatment. Rev. Neurosci. 2004;15:353–369. doi: 10.1515/revneuro.2004.15.5.353. [DOI] [PubMed] [Google Scholar]
  93. Gil-Bea FJ, Aisa B, Schliebs R, Ramirez MJ. Increase of locomotor activity underlying the behavioral dishinbition in Tg2576 mice. Behav. Neurosci. 2007;121:340–344. doi: 10.1037/0735-7044.121.2.340. [DOI] [PubMed] [Google Scholar]
  94. Gimbel DA, Nygaard HB, Coffey EE, Gunther EC, Lauren J, Gimbel ZA, Strittmatter SM. Memory impairment in transgenic Alzheimer mice requires cellular prion protein. J. Neurosci. 2010;30:6367–6374. doi: 10.1523/JNEUROSCI.0395-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Gimenez-Llort L, Blasquez G, Canete T, Johansson B, Oddo S, Tobena A, LaFerla FM, Fernandez-Teruel A. Modeling behavioral and neuronal symptoms of Alzheimer's disease in mice: a role for intraneuronal amyloid. Neurosci. Biobehav. Rev. 2007;31:125–147. doi: 10.1016/j.neubiorev.2006.07.007. [DOI] [PubMed] [Google Scholar]
  96. Gonzalez LE, Rujano M, Tucci S, Paredes D, Silva E, Alba G, Hernandez L. Medial prefrontal transection enhances social interaction. I: Behavioral studies. Brain Res. 2000;887:7–15. doi: 10.1016/s0006-8993(00)02931-0. [DOI] [PubMed] [Google Scholar]
  97. Gonzalez-Lima F, Berndt JD, Valla J, Games D, Reiman EM. Reduced corpus callocum, fornix and hippocampus in PDAPP transgenic mouse model of Alzheimer's disease. NeuroReport. 2001;12:2375–2379. doi: 10.1097/00001756-200108080-00018. [DOI] [PubMed] [Google Scholar]
  98. Gordon MN, Holcomb LA, Jantzen PT, DiCarlo G, Wilcock D, Boyett KW, Connor K, Melachrino J, O'Callaghan JP, Morgan D. Time course of the development of Alzheimer-like pathology in the doubly transgenic mice PS1 + APP mouse. Exp. Neurol. 2002;173:183–195. doi: 10.1006/exnr.2001.7754. [DOI] [PubMed] [Google Scholar]
  99. Gorman MR, Yellon S. Lifespan daily locomotor activity rhythms in a mouse model of amyloid-induced neuropathology. Chronobiol. Int. 2010;27:1159–1177. doi: 10.3109/07420528.2010.485711. [DOI] [PubMed] [Google Scholar]
  100. Görtz N, Lewejohann L, Tomm M, Ambrée O, Keyvani K, Paulus W, Sachser N. Effects of environmental enrichment on exploration, anxiety, and memory in female TgCRND8 Alzheimer mice. Behav. Brain Res. 2008;191:43–48. doi: 10.1016/j.bbr.2008.03.006. [DOI] [PubMed] [Google Scholar]
  101. Graham NL, Emery T, Hodges JR. Distinctive cognitive profiles in Alzheimer's disease and subcortical vascular dementia. J. Neurol. Neurosurg. Psychiatry. 2004;75:61–71. [PMC free article] [PubMed] [Google Scholar]
  102. Griebel G, Belzung C, Perrault G, Sanger DJ. Differences in anxiety-related behaviours and in sensitivity to diazepam in inbred and outbred strains of mice. Psychopharmacology. 2000;148:164–170. doi: 10.1007/s002130050038. [DOI] [PubMed] [Google Scholar]
  103. Gulinello M, Gertner M, Mendoza G, Schoenfeld BP, Oddo S, LaFerla F, Choi CH, McBride SM, Faber DS. Validation of a 2-day water maze protocol in mice. Behav. Brain Res. 2009;196:220–227. doi: 10.1016/j.bbr.2008.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Guo Q, Fu W, Sopher BL, Miller MW, Ware CB, Martin GM, Mattson MP. Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nature Med. 1999;5:101–106. doi: 10.1038/4789. [DOI] [PubMed] [Google Scholar]
  105. Halagappa VK, Guo Z, Pearson M, Matsuoka Y, Cutler RG, Laferla FM, Mattson MP. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol. Dis. 2007;26:212–220. doi: 10.1016/j.nbd.2006.12.019. [DOI] [PubMed] [Google Scholar]
  106. Hardy J, Adolfsson R, Alafuzoff I, Bucht G, Marcusson J, Nyberg P, Perdahl E, Wester P, Winblad B. Transmitter deficits in Alzheimer's disease. Neurochem. Int. 1985;7:545–563. doi: 10.1016/0197-0186(85)90050-6. [DOI] [PubMed] [Google Scholar]
  107. Harigaya Y, Tomidokoro Y, Ikeda M, Sasaki A, Kawarabayashi T, Matsubara E, Kanai M, Saido TC, Younkin SG, Shoji M. Type-specific evolution of amyloid plaque and angiopathy in APPsw mice. Neurosci. Lett. 2006;395:37–41. doi: 10.1016/j.neulet.2005.10.087. [DOI] [PubMed] [Google Scholar]
  108. Harms MP, Kotyk JJ, Merchant KM. Evaluation of white matter integrity in ex vivo brains of amyloid plaque-bearing APPsw transgenic mice using magnetic resonance diffusion tensor imaging. Exp. Neurol. 2006;199:408–415. doi: 10.1016/j.expneurol.2006.01.002. [DOI] [PubMed] [Google Scholar]
  109. Harrison FE, Hosseini AH, McDonald MP, May JM. Vitamin C reduces spatial learning deficits in middle-aged and very old APP/PSEN1 transgenic and wild-type mice. Pharmacol. Biochem. Behav. 2009a;93:443–450. doi: 10.1016/j.pbb.2009.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Harrison FE, Allard J, Bixler R, Usoh C, Li L, May JM, McDonald MP. Antioxidants and cognitive training interact to affect oxidative stress and memory in APP/PSEN1 mice. Nutr. Neurosci. 2009b;12:203–218. doi: 10.1179/147683009X423364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Hart DJ, Craig D, Compton SA, Critchlow S, Kerrigan BM, McIlroy SP, Passmore AP. A retrospective study of the behavioural and psychological symptoms of mid and late phase Alzheimer's disease. Int. J. Geriatr. Psychiatry. 2003;18:1037–1042. doi: 10.1002/gps.1013. [DOI] [PubMed] [Google Scholar]
  112. Hartmann J, Kiewert C, Klein J. Neurotransmitters and energy metabolites in amyloid-bearing APPSWExPSEN1dE9 mouse brain. J. Pharmacol. Exp. Ther. 2010;332:364–370. doi: 10.1124/jpet.109.161091. [DOI] [PubMed] [Google Scholar]
  113. Hashimoto H, Monserratt L, Nguyen P, Feil D, Harwood D, Mandelkern MA, Sultzer DL. Anxiety and regional cortical glucose metabolism in patients with Alzheimer's disease. J. Neuropsychiatry Clin. Neurosci. 2006;18:521–528. doi: 10.1176/jnp.2006.18.4.521. [DOI] [PubMed] [Google Scholar]
  114. Helmes E, Ostbye T. Beyond memory impairment: cognitive changes in Alzheimer's disease. Arch. Clin. Neuropsychol. 2002;17:179–193. doi: 10.1016/s0887-6177(00)00109-8. [DOI] [PubMed] [Google Scholar]
  115. Henderson VW, Mack W, Williams BW. Spatial disorientation in Alzheimer's disease. Arch. Neurol. 1989;46:391–394. doi: 10.1001/archneur.1989.00520400045018. [DOI] [PubMed] [Google Scholar]
  116. Heneka MT, Ramanathan M, Jacobs AH, Dumitrescu-Ozimek L, Bilkei-Gorzo A, Debeir T, Sastre M, Galldiks N, Zimmer A, Hoehn M, Heiss WD, Klockgether T, Staufenbiel M. Locus ceruleus degeneration promotes Alzheimer pathogenesis in amyloid precursor protein 23 transgenic mice. J. Neurosci. 2006;26:1343–1354. doi: 10.1523/JNEUROSCI.4236-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Hirono N, Mega MS, Dinov ID, Mishkin F, Cummings JL. Left frontotemporal hypoperfusion is associated with aggression in patients with dementia. Arch. Neurol. 2000;57:861–866. doi: 10.1001/archneur.57.6.861. [DOI] [PubMed] [Google Scholar]
  118. Hirono N, Mori E, Ishii K, Kitagaki H, Sasaki M, Ikejiri Y, Imamura T, Shimomura T, Ikeda M, Yamashita H. Alteration of regional cerebral glucose utilization with delusions in Alzheimer's disease. J. Neuropsychiat. Clin. Neurosci. 1998;10:433–439. doi: 10.1176/jnp.10.4.433. [DOI] [PubMed] [Google Scholar]
  119. Hock C, Lamb BT. Transgenic mouse models of Alzheimer's disease. Trends Genet. 2001;17:S7–S12. doi: 10.1016/s0168-9525(01)02449-0. [DOI] [PubMed] [Google Scholar]
  120. Holcomb LA, Gordon MN, Jantzen P, Hsiao K, Duff K, Morgan D. Behavioral changes in transgenic mice expressing both amyloid precursor protein and presenilin-1 mutations: lack of association with amyloid deposits. Behav. Genet. 1999;29:177–185. doi: 10.1023/a:1021691918517. [DOI] [PubMed] [Google Scholar]
  121. Holcomb L, Gordon MN, McGowan E, Yu X, Benkowic S, Jantzen P, Wright K, Saad I, Mueller R, Morgan D, Sanders S, Zehr C, O'Campo K, Hardy J, Prada C-M, Eckman C, Younkin S, Hsiao K, Duff K. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin transgenes. Nature Med. 1998;4:97–100. doi: 10.1038/nm0198-097. [DOI] [PubMed] [Google Scholar]
  122. Holmes A. Targeted gene mutation approaches to the study of anxiety-like behavior in mice. Neurosci. Biobehav. Rev. 2001;25:261–273. doi: 10.1016/s0149-7634(01)00012-4. [DOI] [PubMed] [Google Scholar]
  123. Holthoff VA, Beuthien-Baumann B, Kalbe E, Ludecke S, Lenz O, Zundorf G, Spirling S, Schierz K, Winiecki P, Sorbi S, Herholz K. Regional cerebral metabolism in early Alzheimer's disease with clinically significant apathy or depression. Biol. Psychiatry. 2005;57:412–421. doi: 10.1016/j.biopsych.2004.11.035. [DOI] [PubMed] [Google Scholar]
  124. Hoojimans CR, Van der Zee CE, Dederen PJ, Brouwer KM, Reijmer YD, van Groen T, Broersen LM, Lütjohann D, Heerschap A, Kiliaan AJ. DHA and cholesterol containing diets influence Alzheimer-like pathology, cognition and cerebral vasculature in APPswe/PS1dE9 mice. Neurobiol. Dis. 2009;33:482–498. doi: 10.1016/j.nbd.2008.12.002. [DOI] [PubMed] [Google Scholar]
  125. Hort J, Laczo J, Vyhnalek M, Bojar M, Bures J, Vlcek K. Spatial navigation deficit in amnestic mild cognitive impairment. Proc. Natl. Acad. Sci. U.S.A. 2007;104:4042–4047. doi: 10.1073/pnas.0611314104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Howlett DR, Richardson JC. The pathology of APP transgenic mice: a model of Alzheimer's disease or simply overexpression of APP? Histol. Histopathol. 2009;24:83–100. doi: 10.14670/HH-24.83. [DOI] [PubMed] [Google Scholar]
  127. Howlett DR, Richardson JC, Austin A, Parsons AA, Bate ST, Davies DC, Gonzalez MI. Cognitive correlates of Aβ deposition in male and female mice bearing amyloid precursor protein and presenilin-1 mutant transgenes. Brain Res. 2004;1017:130–136. doi: 10.1016/j.brainres.2004.05.029. [DOI] [PubMed] [Google Scholar]
  128. Hsiao K. Transgenic mice expressing Alzheimer amyloid precursor proteins. Exp. Gerontol. 1998a;33:883–889. doi: 10.1016/s0531-5565(98)00045-x. [DOI] [PubMed] [Google Scholar]
  129. Hsiao K. Strain dependent and invariant features of transgenic mice expressing Alzheimer amyloid precursor proteins. Prog. Brain Res. 1998b;117:335–341. doi: 10.1016/s0079-6123(08)64026-1. [DOI] [PubMed] [Google Scholar]
  130. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
  131. Hsiao K, Borchelt DR, Olson K, Johannsdottir R, Kitt C, Yunis W, Xu S, Eckman C, Younkin S, Price D, Iadecola C, Clark HB, Carlson G. Age-related CNS disorder and early death in transgenic FVB-N mice overexpressing Alzheimer amyloid precursor proteins. Neuron. 1995;15:1203–1218. doi: 10.1016/0896-6273(95)90107-8. [DOI] [PubMed] [Google Scholar]
  132. Huang SM, Mouri A, Kokubo H, Nakajima R, Suemoto T, Higuchi M, Staufenbiel M, Noda Y, Yamaguchi H, Nabeshima T, Saido TC, Iwata N. Neprilysin-sensitive synapse-associated amyloid-beta peptide oligomers impair neuronal plasticity and cognitive function. J. Biol. Chem. 2006;281:17941–17951. doi: 10.1074/jbc.M601372200. [DOI] [PubMed] [Google Scholar]
  133. Huitron-Resendiz S, Sanchez-Alavez M, Gallegos R, Berg G, Crawford E, Giacchino JL, Games D, Henriksen SJ, Criado JR. Age-independent and age-related deficits in visuospatial learning, sleep-wake states, thermoregulation and motor activity in PDAPP mice. Brain Res. 2002;928:126–137. doi: 10.1016/s0006-8993(01)03373-x. [DOI] [PubMed] [Google Scholar]
  134. Hyde LA, Kazdoba TM, Grilli M, Lozza G, Brussa R, Zhang Q, Wong GT, McCool MF, Zhang L, Parker EM, Higgins GA. Age-progressing cognitive impairments and neuropathology in transgenic CRND8 mice. Behav. Brain Res. 2005;160:344–355. doi: 10.1016/j.bbr.2004.12.017. [DOI] [PubMed] [Google Scholar]
  135. Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT. APPβSw transgenic mice develop age-related Aβ deposits and neuropil abnormalities but no neuronal loss in CA1. J. Neuropathol. Exp. Neurol. 1997;56:965–973. doi: 10.1097/00005072-199709000-00002. [DOI] [PubMed] [Google Scholar]
  136. Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of Aβ42(43) and Aβ40 in senile plaques and end-specific Aβ monoclonals: evidence that an initially deposited species is Aβ42(43) Neuron. 1994;13:45–53. doi: 10.1016/0896-6273(94)90458-8. [DOI] [PubMed] [Google Scholar]
  137. Jacobsen JS, Wu CC, Redwine JM, Comery TA, Arias R, Bowlby M, Martone R, Morrison JH, Pangalos MN, Reinhart PH, Bloom FE. Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U.S.A. 2006;103:5161–5166. doi: 10.1073/pnas.0600948103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Jacobs DH, Adair JC, Williamson DJ, Na DL, Gold M, Foundas AL, Shuren JE, Cibula JE, Heilman KM. Apraxia and motor-skill acquisition in Alzheimer's disease are dissociable. Neuropsychologia. 1999;37:875–880. doi: 10.1016/s0028-3932(98)00139-0. [DOI] [PubMed] [Google Scholar]
  139. Jacobs DM, Sano M, Dooneief G, Marder K, Bell KL, Stern Y. Neuropsychological detection and characterization of preclinical Alzheimer’s disease. Neurology. 1995;45:957–962. doi: 10.1212/wnl.45.5.957. [DOI] [PubMed] [Google Scholar]
  140. Jankowsky JL, Melnikova T, Fadale DJ, Xu GM, Slunt HH, Gonzales V, Younkin LH, Younkin SG, Borchelt DR, Savonenko AV. Environmental enrichment mitigates cognitive deficits in a mouse model of Alzheimer's disease. J. Neurosci. 2005a;25:5217–5224. doi: 10.1523/JNEUROSCI.5080-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Jankowsky JL, Slunt HH, Gonzales V, Savonenko AV, Wen JC, Jenkins NA, Copeland NG, Younkin LH, Lester HA, Younkin SG, Borchelt DR. Persistent amyloidosis following suppression of Abeta production in a transgenic model of Alzheimer disease. PLoS Med. 2005b;2:e355. doi: 10.1371/journal.pmed.0020355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, Copeland NG, Lee MK, Younkin LH, Wagner SL, Younkin SG, Borchelt DR. Mutant presenilins specifically elevate the levels of the 42 residue β-amyloid peptide in vivo: evidence for augmentation of a 42-specific γ secretase. Hum. Mol. Genet. 2004;13:159–170. doi: 10.1093/hmg/ddh019. [DOI] [PubMed] [Google Scholar]
  143. Janus C, Westaway D. Transgenic mouse models of Alzheimer's disease. Physiol. Behav. 2001;73:873–886. doi: 10.1016/s0031-9384(01)00524-8. [DOI] [PubMed] [Google Scholar]
  144. Janus C, Pearson J, McLaurin J, Matthews PM, Jiang Y, Schmidt SD, Chishti MA, Horne P, Heslin D, French J, Mount HTJ, Nixon RA, Mercken M, Bergeron C, Fraser PE, St George-Hyslop P, Westaway D. Aβ peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease. Nature. 2000;408:979–982. doi: 10.1038/35050110. [DOI] [PubMed] [Google Scholar]
  145. Jawhar S, Trawicka A, Jenneckens C, Bayer TA, Wirths O. Motor deficits, neuron loss, and reduced anxiety coinciding with axonal degeneration and intraneuronal Abeta aggregation in the 5XFAD mouse model of Alzheimer's disease. Neurobiol. Aging. 2012;33(1):196.e29, 196.e40. doi: 10.1016/j.neurobiolaging.2010.05.027. [DOI] [PubMed] [Google Scholar]
  146. Jawhar S, Wirths O, Schilling S, Graubner S, Demuth HU, Bayer TA. Overexpression of glutaminyl cyclase, the enzyme responsible for pyroglutamate Aβ formation, induces behavioral deficits, and glutaminyl cyclase knock-out rescues the behavioral phenotype in 5XFAD mice. J. Biol. Chem. 2011;286:4454–4460. doi: 10.1074/jbc.M110.185819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J. Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch to the microglial phenotype from alternative to classic. J. Neurosci. 2008;28:11650–11661. doi: 10.1523/JNEUROSCI.3024-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Johnson CT, Olton DS, Gage FH, III, Jenko PG. Damage to hippocampus and hippocampal connections: effects on DRL and spontaneous alternation. J. Comp. Physiol. Psychol. 1977;91:508–522. doi: 10.1037/h0077346. [DOI] [PubMed] [Google Scholar]
  149. Kalback W, Watson MD, Kokjohn TA, Kuo Y-M, Weiss N, Luehrs DC, Lopez J, Brune D, Sisodia SS, Staufenbiel M, Emmerling M, Roher AE. APP transgenic mice Tg2576 accumulate Aβ peptides that are distinct from the chemically modified and insoluble peptides deposited in Alzheimer's disease senile plaques. Biochemistry. 2002;41:922–928. doi: 10.1021/bi015685+. [DOI] [PubMed] [Google Scholar]
  150. Karlsson T, Börjesson A, Adolfsson R, Nilsson L-G. Successive memory test performance and priming in Alzheimer's disease: evidence from the word-fragment completion task. Cortex. 2002;38:341–355. doi: 10.1016/s0010-9452(08)70664-6. [DOI] [PubMed] [Google Scholar]
  151. Kavcic V, Fernandez R, Logan D, Duffy CJ. Neurophysiological and perceptual correlates of navigational impairment in Alzheimer's disease. Brain. 2006;129:736–746. doi: 10.1093/brain/awh727. [DOI] [PubMed] [Google Scholar]
  152. Kawarabayashi T, Younkin LH, Saido TC, Shoji M, Ashe KH, Younkin SG. Age-dependent changes in brain, CSF,and amyloid β protein in the Tg2576 transgenic mouse model of Alzheimer's disease. J. Neurosci. 2001;21:372–381. doi: 10.1523/JNEUROSCI.21-02-00372.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Kawarabayashi T, Shoji M, Younkin LH, Wen-Lang L, Dickson DW, Murakami T, Matsubara E, Abe K, Ashe KH, Younkin SG. Dimeric amyloid β protein rapidly accumulates in lipid rafts followed by apolipoprotein E and phosphorylated tau accumulation in the Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 2004;24:3801–3809. doi: 10.1523/JNEUROSCI.5543-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Kessels RP, Feijen J, Postma A. Implicit and explicit memory for spatial information in Alzheimer's disease. Dement. Geriatr. Cogn. Disord. 2005;20:184–191. doi: 10.1159/000087233. [DOI] [PubMed] [Google Scholar]
  155. Kim H-S, Suh Y-H. Neurotoxicity of a carboxyl-terminal fragment of the Alzheimer's amyloid precursor protein. J. Neurochem. 1996;67:1172–1182. doi: 10.1046/j.1471-4159.1996.67031172.x. [DOI] [PubMed] [Google Scholar]
  156. Kimchi EY, Kajdasz S, Bacskai BJ, Hyman BT. Analysis of cerebral amyloid angiopathy in a transgenic mouse model of Alzheimer's disease using in vivo multiphoton microscopy. J. Neuropathol. Exp. Neurol. 2001;60:274–279. doi: 10.1093/jnen/60.3.274. [DOI] [PubMed] [Google Scholar]
  157. King DL, Arendash GW. Behavioral characterization of the Tg2576 transgenic model of Alzheimer's disease through 19 months. Physiol. Behav. 2002;75:627–642. doi: 10.1016/s0031-9384(02)00639-x. [DOI] [PubMed] [Google Scholar]
  158. Kirkby RJ, Stein DG, Kimble RJ, Kimble DP. Effects of hippocampal lesions and duration of sensory input on spontaneous alternation. J. Comp. Physiol. Psychol. 1967;64:342–345. doi: 10.1037/h0088012. [DOI] [PubMed] [Google Scholar]
  159. Kitamoto T, Mohri S, Ironside JW, Miyoshi I, Tanaka T, Kitamoto N, Itohara S, Kasai N, Katsuki M, Higuchi J, Muramoto T, Shin RW. Follicular dendritic cell of the knock-in mouse provides a new bioassay for human prions. Biochem. Biophys. Res. Commun. 2002;294:280–286. doi: 10.1016/S0006-291X(02)00476-X. [DOI] [PubMed] [Google Scholar]
  160. Kitazawa M, Green KN, Caccamo A, LaFerla FM. Genetically augmenting Aβ42 levels in skeletal muscle exacerbates inclusion body myositis-like pathology and motor deficits in transgenic mice. Am. J. Pathol. 2006;168:1986–1997. doi: 10.2353/ajpath.2006.051232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Köhler C, Srebro B. Effects of lateral and medial septal lesions on exploratory behavior in the albino rat. Brain Res. 1980;182:423–440. doi: 10.1016/0006-8993(80)91199-3. [DOI] [PubMed] [Google Scholar]
  162. Kokjohn TA, Roher AE. Amyloid precursor protein transgenic mouse models and Alzheimer's disease: understanding the paradigms, limitations, and contributions. Alzheimers Dement. 2009;5:340–347. doi: 10.1016/j.jalz.2009.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Kumar-Singh S, Dewachter I, Moechars D, Lübke U, De Jonghe C, Ceuterick C, Checler F, Naidu A, Cordell B, Cras P, Van Broeckhoven C, Van Leuven F. Behavioral disturbances without amyloid deposits in mice overexpressing human amyloid precursor protein with Flemish (A692G) or Dutch (E693Q) mutation. Neurobiol. Dis. 2000;7:9–22. doi: 10.1006/nbdi.1999.0272. [DOI] [PubMed] [Google Scholar]
  164. Kuo Y-M, Kokjohn TA, Beach TG, Sue LI, Brune D, Lopez JC, Kalback WM, Abramowski D, Sturchler-Pierrat C, Staufenbiel M, Roher AE. Comparative analysis of amyloid-beta chemical structure and amyloid plaque morphology of transgenic mouse and Alzheimer's disease brains. J. Biol. Chem. 2001a;276:12991–12998. doi: 10.1074/jbc.M007859200. [DOI] [PubMed] [Google Scholar]
  165. Kuo YM, Beach TG, Sue LI, Scott S, Layne KJ, Kokjohn TA, Kalback WM, Luehrs DC, Vishnivetskaya TA, Abramowski D, Sturchler-Pierrat C, Staufenbiel M, Weller RO, Roher AE. The evolution of A beta peptide burden in the APP23 transgenic mice: implications for A beta deposition in Alzheimer disease. Mol. Med. 2001b;7:609–618. [PMC free article] [PubMed] [Google Scholar]
  166. Kurt MA, Davies DC, Kidd M, Duff K, Howlett DR. Hyperphosphorylated tau and paired helical filament-like structures in the brains of mice carrying mutant amyloid precursor protein and mutant presenilin-1 transgenes. Neurobiol. Dis. 2003;14:89–97. doi: 10.1016/s0969-9961(03)00084-6. [DOI] [PubMed] [Google Scholar]
  167. LaFerla FM, Hall CK, Ngo L, Jay G. Extracellular deposition of β-amyloid upon p53-dependent neuronal cell death in transgenic mice. J. Clin. Invest. 1996;98:1626–1632. doi: 10.1172/JCI118957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. LaFerla FM, Tinkle BT, Bieberich CJ, Haudenschild CC, Jay G. The Alzheimer's Aβ peptide induces neurodegeneration and apoptotic cell death in transgenic mice. Nature Genet. 1995;9:21–30. doi: 10.1038/ng0195-21. [DOI] [PubMed] [Google Scholar]
  169. Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci. Biobehav. Rev. 2002;26:91–104. doi: 10.1016/s0149-7634(01)00041-0. [DOI] [PubMed] [Google Scholar]
  170. Lalonde R, Strazielle C. Relations between open-field, elevated plus-maze, and emergence tests in C57BL/6J and BALB/c mice injected with GABA- and 5HT-anxiolytic agents. Fundam. Clin. Pharmacol. 2010;24:365–376. doi: 10.1111/j.1472-8206.2009.00772.x. [DOI] [PubMed] [Google Scholar]
  171. Lalonde R, Kim H-D, Fukuchi K. Exploratory activity, anxiety, and motor coordination in bigenic APPswe+PS1/ΔE9 mice. Neurosci. Lett. 2004;369:156–161. doi: 10.1016/j.neulet.2004.07.069. [DOI] [PubMed] [Google Scholar]
  172. Lalonde R, Kim H-D, Maxwell JA, Fukuchi K. Exploratory activity and spatial learning in 12-month-old APP695SWE/co+PS1/ΔE9 mice with amyloid plaques. Neurosci. Lett. 2005;390:87–92. doi: 10.1016/j.neulet.2005.08.028. [DOI] [PubMed] [Google Scholar]
  173. Lalonde R, Dumont M, Fukuchi K, Strazielle C. Transgenic mice expressing the human C99 terminal fragment of βAPP: effects on spatial learning, exploration, anxiety, and motor coordination. Exp. Gerontol. 2002a;37:1399–1410. doi: 10.1016/s0531-5565(02)00123-7. [DOI] [PubMed] [Google Scholar]
  174. Lalonde R, Dumont M, Staufenbiel M, Sturchler-Pierrat C, Strazielle C. Spatial learning, exploration, anxiety, and motor coordination in female APP23 transgenic mice with the Swedish mutation. Brain Res. 2002b;956:36–44. doi: 10.1016/s0006-8993(02)03476-5. [DOI] [PubMed] [Google Scholar]
  175. Lalonde R, Lewis TL, Strazielle C, Kim H, Fukuchi K. Neurobehavioral testing of transgenic βAPP695SWE (Tg2576) mice with the Swedish mutation. Brain Res. 2003;977:38–45. doi: 10.1016/s0006-8993(03)02694-5. [DOI] [PubMed] [Google Scholar]
  176. Lamberty Y, Gower AJ. Age-related changes in spontaneous behavior and learning in NMRI mice from maturity to middle age. Physiol. Behav. 1990;47:1137–1144. doi: 10.1016/0031-9384(90)90364-a. [DOI] [PubMed] [Google Scholar]
  177. Lamberty Y, Gower AJ. Age-related changes in spontaneous behavior and learning in NMRI mice from middle to old age. Physiol. Behav. 1992;51:81–88. doi: 10.1016/0031-9384(92)90206-h. [DOI] [PubMed] [Google Scholar]
  178. Lanari A, Amenta F, Silvestrelli G, Tomassoni D, Parnetti L. Neurotransmitter deficits in behavioural and psychological symptoms of Alzheimer's disease. Mech. Ageing Dev. 2006;127:158–165. doi: 10.1016/j.mad.2005.09.016. [DOI] [PubMed] [Google Scholar]
  179. Lanz TA, Carter DB, Merchant KM. Dendritic spine loss in the hippocampus of young PDAPP and Tg2576 mice and its prevention by the ApoE2 genotype. Neurobiol. Dis. 2003;13:246–253. doi: 10.1016/s0969-9961(03)00079-2. [DOI] [PubMed] [Google Scholar]
  180. Lassalle JM, Halley H, Daumas S, Verret L, Francés B. Effects of the genetic background on cognitive performances of TG2576 mice. Behav. Brain Res. 2008;191:104–110. doi: 10.1016/j.bbr.2008.03.017. [DOI] [PubMed] [Google Scholar]
  181. Le Cudennec C, Faure A, Ly M, Delatour B. One-year longitudinal evaluation of sensorimotor functions in APP751SL transgenic mice. Genes Brain Behav. 2008;7(Suppl 1):83–91. doi: 10.1111/j.1601-183X.2007.00374.x. [DOI] [PubMed] [Google Scholar]
  182. Lee AC, Rahman S, Hodges JR, Sahakian BJ, Graham KS. Associative and recognition memory for novel objects in dementia: implications for diagnosis. Eur. J. Neurosci. 2003;18:1660–1670. doi: 10.1046/j.1460-9568.2003.02883.x. [DOI] [PubMed] [Google Scholar]
  183. Lee GD, Aruna JH, Barrett PM, Lei DL, Ingram DK, Mouton PR. Stereological analysis of microvascular parameters in a double transgenic model of Alzheimer's disease. Brain Res Bull. 2005;65:317–322. doi: 10.1016/j.brainresbull.2004.11.024. 2005. [DOI] [PubMed] [Google Scholar]
  184. Lee K-W, Lee SH, Kim H, Song JS, Yang SD, Paik SG, Han PL. Progressive cognitive impairment and anxiety induction in the absence of plaque deposition in C57BL/6 inbred mice expressing transgenic amyloid precursor protein. J. Neurosci. Res. 2004;76:572–580. doi: 10.1002/jnr.20127. [DOI] [PubMed] [Google Scholar]
  185. Lee K-W, Im JY, Song JS, Lee SH, Lee HJ, Ha HY, Koh JY, Gwag BJ, Yang SD, Paik SG, Han PL. Progressive neuronal loss and behavioral impairments of transgenic C57BL/6 inbred mice expressing the carboxy terminus of amyloid precursor protein. Neurobiol. Dis. 2006;22:10–24. doi: 10.1016/j.nbd.2005.09.011. [DOI] [PubMed] [Google Scholar]
  186. Levy R, Dubois B. Apathy and the functional anatomy of the prefrontal cortex-basal ganglia circuits. Cereb. Cortex. 2006;16:916–928. doi: 10.1093/cercor/bhj043. [DOI] [PubMed] [Google Scholar]
  187. Lewejohann L, Hoppmann AM, Kegel P, Kritzler M, Krüger A, Sachser N. Behavioral phenotyping of a murine model of Alzheimer's disease in a seminaturalistic environment using RFID tracking. Behav. Res. Methods. 2009;41:850–856. doi: 10.3758/BRM.41.3.850. [DOI] [PubMed] [Google Scholar]
  188. Li QX, Maynard C, Cappai R, McLean CA, Cherny RA, Lynch T, Culvenor JG, Trevaskis J, Tanner JE, Bailey KA, Czech C, Bush AI, Beyreuther K, Masters CL. Intracellular accumulation of detergent-soluble amyloidogenic A beta fragment of Alzheimer's disease precursor protein in the hippocampus of aged transgenic mice. J. Neurochem. 1999;72:2479–2487. doi: 10.1046/j.1471-4159.1999.0722479.x. [DOI] [PubMed] [Google Scholar]
  189. Lim GP, Yang F, Chu T, Gahtan E, Ubeda O, Beech W, Overmier JB, Hsiao-Ashe K, Frautsky SA, Cole GM. Ibuprofen effects on Alzheimer pathology and open field activity in APPSW transgenic mice. Neurobiol. Aging. 2001;22:983–991. doi: 10.1016/s0197-4580(01)00299-8. [DOI] [PubMed] [Google Scholar]
  190. Liu L, Ikonen S, Heikkinen T, Tapiola T, van Groen T, Tanila H. The effects of long-term treatment with metrifonate, a cholinesterase inhibitor, on cholinergic activity, amyloid pathology, and cognitive function in APP and PS1 doubly transgenic mice. Exp. Neurol. 2002;173:196–204. doi: 10.1006/exnr.2001.7819. [DOI] [PubMed] [Google Scholar]
  191. Liu Y, Yoo MJ, Savonenko A, Stirling W, Price DL, Borchelt DR, Mamounas L, Lyons WE, Blue ME, Lee MK. Amyloid pathology is associated with progressive monoaminergic neurodegeneration in a transgenic mouse model of Alzheimer's disease. J. Neurosci. 2008;28:13805–13814. doi: 10.1523/JNEUROSCI.4218-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Lord A, Kalimo H, Eckman C, Zhang X-Q, Lannfelt L, Nilsson LNG. The Arctic Alzheimer mutation facilitates early intraneuronal Aβ aggregation and senile plaque formation in transgenic mice. Neurobiol. Aging. 2006;27:67–77. doi: 10.1016/j.neurobiolaging.2004.12.007. [DOI] [PubMed] [Google Scholar]
  193. Lord A, Englund H, Söderberg L, Tucker S, Clausen F, Hillered L, Gordon M, Morgan D, Lannfelt L, Pettersson FE, Nilsson LN. Amyloid-β protofibril levels correlate with spatial learning in Arctic Alzheimer's disease transgenic mice. FEBS. J. 2009;276:995–1006. doi: 10.1111/j.1742-4658.2008.06836.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Lüth H-J, Münch G, Arendt T. Aberrant expression of NOS isoforms in Alzheimer's disease is structurally related to nitrotyrosine formation. Brain Res. 2002;953:135–143. doi: 10.1016/s0006-8993(02)03280-8. [DOI] [PubMed] [Google Scholar]
  195. Lüth H-J, Holzer M, Gärtner U, Staufenbiel M, Arendt T. Expression of endothelial and induced NOS-isoforms is increased in Alzheimer's disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Res. 2001;913:57–67. doi: 10.1016/s0006-8993(01)02758-5. [DOI] [PubMed] [Google Scholar]
  196. Lyketsos CG, Lopez O, Jones B, Fitzpatrick AL, Breitner J, DeKosky S. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the cardiovascular health study. JAMA. 2002;288:1475–1483. doi: 10.1001/jama.288.12.1475. [DOI] [PubMed] [Google Scholar]
  197. Lyness SA, Zarow C, Chui HC. Neuron loss in key cholinergic and aminergic nuclei in Alzheimer's disease: a meta-analysis. Neurobiol. Aging. 2003;24:1–23. doi: 10.1016/s0197-4580(02)00057-x. [DOI] [PubMed] [Google Scholar]
  198. Marshall GA, Monserratt L, Harwood D, Mandelkern M, Cummings JL, Sultzer DL. Positron emission tomography metabolic correlates of apathy in Alzheimer disease. Arch. Neurol. 2007;64:1015–1020. doi: 10.1001/archneur.64.7.1015. [DOI] [PubMed] [Google Scholar]
  199. Marutle A, Unger C, Hellström-Lindahl E, Wang J, Puoliväli J, Tanila H, Nordberg A, Zhang X. Elevated levels of Aβ1–40 and Aβ1–42 do not alter the binding sites of nicotinic recepeptor subtypes in the brain of APPswe and PS1 double transgenic mice. Neurosci. Lett. 2002;328:269–272. doi: 10.1016/s0304-3940(02)00546-3. [DOI] [PubMed] [Google Scholar]
  200. Masliah E, Sisk A, Mallory M, Games D. Neurofibrillary pathology in transgenic mice overexpressing V717F β-amyloid precursor protein. J. Neuropathol. Exp. Neurol. 2001;60:357–368. doi: 10.1093/jnen/60.4.357. [DOI] [PubMed] [Google Scholar]
  201. Masliah E, Sisk A, Mallory M, Mucke L, Schenk D, Games D. Comparison of neurodegenerative pathology in transgenic mice overexpressing V717F β-amyloid precursor protein and Alzheimer's disease. J. Neurosci. 1996;16:5795–5811. doi: 10.1523/JNEUROSCI.16-18-05795.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Massaad CA, Washington TM, Pautler RG, Klann E. Overexpression of SOD-2 reduces hippocampal superoxide and prevents memory deficits in a mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U.S.A. 2009;106:13576–13581. doi: 10.1073/pnas.0902714106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. McGowan E, Eriksen J, Hutton M. A decade of modeling Alzheimer's disease in transgenic mice. Trends Genet. 2006;22:281–289. doi: 10.1016/j.tig.2006.03.007. [DOI] [PubMed] [Google Scholar]
  204. McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, Yu C, Skipper L, Murphy MP, Beard J, Das P, Jansen K, Delucia M, Lin WL, Dolios G, Wang R, Eckman CB, Dickson DW, Hutton M, Hardy J, Golde T. Aβ42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron. 2005;47:191–199. doi: 10.1016/j.neuron.2005.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRA work group under the Department of Health and Human Services task force on Alzheimer's disease. Neurology. 1984;34:939–944. doi: 10.1212/wnl.34.7.939. [DOI] [PubMed] [Google Scholar]
  206. McLellan ME, Kajdasz ST, Hyman BT, Bacskai BJ. In vivo imaging of reactive oxygen species specifically associated with thioflavine S-positive amyloid plaques by multiphoton microscopy. J. Neurosci. 2003;23:2212–2217. doi: 10.1523/JNEUROSCI.23-06-02212.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. McPhie DL, Lee RKK, Eckman CB, Olstein DH, Durham SP, Yager D, Younkin SG, Wurtman RJ, Neve RL. Neuronal expression of β-amyloid-precursor protein Alzheimer mutations causes intracellular accumulation of a C-terminal fragment containing both the amyloid β and cytoplasmic domains. J. Biol. Chem. 1997;272:24743–24746. doi: 10.1074/jbc.272.40.24743. [DOI] [PubMed] [Google Scholar]
  208. Menard J, Treit D. The septum and the hippocampus differentially mediate anxiolytic effects of R(+)-8-OH-DPAT. Behav. Pharmacol. 1998;9:93–101. [PubMed] [Google Scholar]
  209. Menard J, Treit D. The anxiolytic effects of intra-hippocampal midazolam are antagonized by intra-septal L-glutamate. Brain Res. 2001;888:163–166. doi: 10.1016/s0006-8993(00)03046-8. [DOI] [PubMed] [Google Scholar]
  210. Mentis MJ, Weinstein EA, Horwitz B, McIntosh AR, Pietrini P, Alexander GE, Furey M, Murphy DGM. Abnormal brain glucose metabolism in the delusional misidentification syndromes: a positron emission tomography study in Alzheimer disease. Biol. Psychiat. 1995;38:438–449. doi: 10.1016/0006-3223(94)00326-x. [DOI] [PubMed] [Google Scholar]
  211. Middei S, Geracitano R, Caprioli A, Mercuri N, Ammassari-Teule M. Preserved frontostriatal plasticity and enhanced procedural learning in a transgenic mouse model of Alzheimer's disease overexpressing mutant hAPPswe. Learn. Mem. 2004;11:447–452. doi: 10.1101/lm.80604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Middei S, Daniele S, Caprioli A, Ghirardi O, Ammassari-Teule M. Progressive cognitive decline in a transgenic mouse model of Alzheimer's disease overexpressing mutant hAPPswe. Genes Brain Behav. 2006;5:249–256. doi: 10.1111/j.1601-183X.2005.00160.x. [DOI] [PubMed] [Google Scholar]
  213. Mineur YS, McGoughlin D, Crusio WE, Sluyter F. Genetic models of Alzheimer's disease. Neural Plast. 2005;12:299–310. doi: 10.1155/NP.2005.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Moechars D, Lorent K, Van Leuven F. Premature death in transgenic mice that overexpress a mutant amyloid precursor protein is preceded by severe neurodegeneration and apoptosis. Neuroscience. 1999a;91:819–830. doi: 10.1016/s0306-4522(98)00599-5. [DOI] [PubMed] [Google Scholar]
  215. Moechars D, Lorent K, Dewachter I, Baekelandt V, De Strooper B, Van Leuven F. Transgenic mice expressing an alpha-secretion mutant of the amyloid precursor protein in the brain develop a progressive CNS disorder. Behav. Brain Res. 1998;95:55–64. doi: 10.1016/s0166-4328(97)00210-6. [DOI] [PubMed] [Google Scholar]
  216. Moechars D, Lorent K, De Strooper B, Dewachter I, Van Leuven F. Expression in brain of amyloid precursor protein mutated in the α-secretase site causes disturbed behavior, neuronal degeneration and premature death in transgenic mice. EMBO. J. 1996;15:1265–1274. [PMC free article] [PubMed] [Google Scholar]
  217. Moechars D, Dewachter I, Lorent K, Reversé D, Baekelandt V, Naidu A, Tesseur I, Spittaels K, Van Den Haute C, Checler F, Godaux E, Cordell B, Van Leuven F. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J. Biol. Chem. 1999b;274:6483–6492. doi: 10.1074/jbc.274.10.6483. [DOI] [PubMed] [Google Scholar]
  218. Monacelli AM, Cushman LA, Kavcic V, Duffy CJ. Spatial disorientation in Alzheimer's disease: the remembrance of things passed. Neurology. 2003;61:1491–1497. doi: 10.1212/wnl.61.11.1491. [DOI] [PubMed] [Google Scholar]
  219. Moran PM, Higgins LS, Cordell B, Moser PC. Age-related learning deficits in transgenic mice expressing the 751-amino acid isoform of human β-amyloid protein precursor protein. Proc. Natl. Acad. Sci. U.S.A. 1995;92:5341–5345. doi: 10.1073/pnas.92.12.5341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Moreira CM, Masson S, Carvalho MC, Brandão ML. Exploratory behaviour of rats in the elevated plus-maze is differentially sensitive to inactivation of the basolateral and central amygdaloid nuclei. Brain Res. Bull. 2007;71:466–474. doi: 10.1016/j.brainresbull.2006.10.004. [DOI] [PubMed] [Google Scholar]
  221. Morgan D. Learning and memory deficits in APP transgenic mouse models of amyloid deposition. Neurochem. Res. 2003;28:1029–1034. doi: 10.1023/a:1023255106106. [DOI] [PubMed] [Google Scholar]
  222. Morgenstern R, Mende T, Gold R, Lemme P, Oelssner W. Drug-induced modulation of locomotor hyperactivity induced by picrotoxin in nucleus accumbens. Pharmacol. Biochem. Behav. 1984;21:501–506. doi: 10.1016/s0091-3057(84)80030-1. [DOI] [PubMed] [Google Scholar]
  223. Mucke L, Masliah E, Johnson WB, Ruppe MD, Alford M, Rockenstein EM, Forss-Petter S, Pietropaolo M, Mallory M, Abraham CR. Synaptotrophic effects of human amyloid beta protein precursors in the cortex of transgenic mice. Brain Res. 1994;666:151–167. doi: 10.1016/0006-8993(94)90767-6. [DOI] [PubMed] [Google Scholar]
  224. Mucke L, Masliah E, Yu G-Q, Mallory M, Rockenstein EM, Tatsuno G, Hu K, Kholodenko D, Johnson-Wood K, McConlogue L. High-level neuronal expression of Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 2000;20:4050–4058. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Mueggler T, Baumann D, Rausch M, Staufenbiel M, Rudin M. Age-dependent impairment of somatosensory response in the amyloid precursor protein 23 transgenic mouse model of Alzheimer's disease. J. Neurosci. 2003;23:8231–8236. doi: 10.1523/JNEUROSCI.23-23-08231.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Mueggler T, Meyer-Luehmann M, Rausch M, Staufenbiel M, Jucker M, Rudin M. Restricted diffusion in the brain of transgenic mice with cerebral amyloidosis. Eur. J. Neurosci. 2004;20:811–817. doi: 10.1111/j.1460-9568.2004.03534.x. [DOI] [PubMed] [Google Scholar]
  227. Mueggler T, Sturchler-Pierrat C, Baumann D, Rausch M, Staufenbiel M, Rudin M. Compromised hemodynamic response in amyloid precursor transgenic mice. J. Neurosci. 2002;22:7218–7224. doi: 10.1523/JNEUROSCI.22-16-07218.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Murray CL, Fibiger HC. Pilocarpine and physostigmine attenuate spatial memory impairments produced by lesions of the nucleus basalis magnocellularis. Behav. Neurosci. 1986;100:23–32. doi: 10.1037//0735-7044.100.1.23. [DOI] [PubMed] [Google Scholar]
  229. Müller U, Cristina N, Li Z-W, Wolfer DP, Lipp H-P, Rülicke T, Brandner S, Aguzzi A, Weissmann C. Behavioral and anatomical deficits in mice homozygous for a modified β-amyloid precursor protein gene. Cell. 1994;79:755–765. doi: 10.1016/0092-8674(94)90066-3. [DOI] [PubMed] [Google Scholar]
  230. Nalbantoglu J, Tirado-Santiago G, Lahsaïni A, Poirier J, Goncalves O, Verge G, Momoli F, Welner SA, Massicotte G, Julien J-P, Shapiro ML. Impaired learning and LTP in mice expressing the carboxy terminus of the Alzheimer amyloid precursor protein. Nature. 1997;387:500–505. doi: 10.1038/387500a0. [DOI] [PubMed] [Google Scholar]
  231. Nelson RL, Guo Z, Halagappa VM, Pearson M, Gray AJ, Matsuoka Y, Brown M, Martin B, Iyun T, Maudsley S, Clark RF, Mattson MP. Prophylactic treatment with paroxetine ameliorates behavioral deficits and retards the development of amyloid and tau pathologies in 3xTgAD mice. Exp. Neurol. 2007;205:166–176. doi: 10.1016/j.expneurol.2007.01.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Nic Dhonnchadha BA, Bourin M, Hascoët M. Anxiolytic-like effects of 5-HT2 ligands on three mouse models of anxiety. Behav. Brain Res. 2003;140:203–214. doi: 10.1016/s0166-4328(02)00311-x. [DOI] [PubMed] [Google Scholar]
  233. Nielson KA, Cummings BJ, Cotman CW. Constructional apraxia in Alzheimer's disease correlates with neuritic neuropathology in occipital cortex. Brain Res. 1996;741:284–293. doi: 10.1016/s0006-8993(96)00983-3. [DOI] [PubMed] [Google Scholar]
  234. Nunes-de-Souza RL, Canto-de-Souza A, da-Costa M, Fornari RV, Graeff FG, Pelá IR. Anxiety-induced antinociception in mice: effects of systemic and intra-amygdala administration of 8-OH-DPAT and midazolam. Psychopharmacology. 2000;150:300–310. doi: 10.1007/s002130000428. [DOI] [PubMed] [Google Scholar]
  235. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer's disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 2006;26:10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Obulesu M, Rao DM. Animal models of Alzheimer's disease: an understanding of pathology and therapeutic avenues. Int. J. Neurosci. 2010;120:531–537. doi: 10.3109/00207451003760080. [DOI] [PubMed] [Google Scholar]
  237. Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, Metherate R, Mattson MP, Akbari Y, LaFerla FM. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron. 2003;39:409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
  238. Ognibene E, Middei S, Daniele S, Adriani W, Ghirardi O, Caprioli A, Laviola G. Aspects of spatial memory and behavioral disinhibition in Tg2576 transgenic mice as a model of Alzheimer's disease. Behav. Brain Res. 2005;156:225–232. doi: 10.1016/j.bbr.2004.05.028. [DOI] [PubMed] [Google Scholar]
  239. Ohno M, Sametsky EA, Younkin LH, Oakley H, Younkin SG, Citron M, Vassar R, Disterhoft JF. BACE1 deficiency rescues memory deficits and cholinergic dysfunction in a mouse model of Alzheimer's disease. Neuron. 2004;41:27–33. doi: 10.1016/s0896-6273(03)00810-9. [DOI] [PubMed] [Google Scholar]
  240. Ojha J, Karmegam RV, Masilamoni JG, Terry AV, Cashikar AG. Behavioral defects in chaperone-deficient Alzheimer's disease model mice. PLoS One. 2011;6:e16550. doi: 10.1371/journal.pone.0016550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Oster-Granite ML, McPhie DL, Greenan J, Neve RL. Age-dependent neuronal and synaptic degeneration in mice transgenic for the C terminus of the amyloid precursor protein. J. Neurosci. 1996;16:6732–6741. doi: 10.1523/JNEUROSCI.16-21-06732.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Palop JJ, Jones B, Kekonius L, Chin J, Yu GQ, Raber J, Masliah E, Mucke L. Neuronal depletion of calcium-dependent proteins in the dentate gyrus is tightly linked to Alzheimer's disease-related cognitive deficits. Proc. Natl. Acad. Sci. U.S.A. 2003;100:9572–9577. doi: 10.1073/pnas.1133381100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Parasuraman R, Greenwood PM, Alexander GE. Alzheimer disease constricts the dynamic range of spatial attention in visual search. Neuropsychologia. 2000;38:1126–1135. doi: 10.1016/s0028-3932(00)00024-5. [DOI] [PubMed] [Google Scholar]
  244. Pardon MC, Sarmad S, Rattray I, Bates TE, Scullion GA, Marsden CA, Barrett DA, Lowe J, Kendall DA. Repeated novel cage exposure-induced improvement of early Alzheimer's-like cognitive and amyloid changes in TASTPM mice is unrelated to changes in brain endocannabinoids levels. Neurobiol. Aging. 2009;30:1099–1113. doi: 10.1016/j.neurobiolaging.2007.10.002. [DOI] [PubMed] [Google Scholar]
  245. Perez SE, Lazarov O, Koprich JB, Chen EY, Rodriguez-Menendez V, Lipton JW, Sisodia SS, Mufson EJ. Nigrostriatal dysfunction in familial Alzheimer's disease-linked APPswe/PS1ΔE9 transgenic mice. J. Neurosci. 2005;25:10220–1029. doi: 10.1523/JNEUROSCI.2773-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Petry S, Cummings JL, Hill MA, Shapira J. Personality alterations in dementia of the Alzheimer type. Arch. Neurol. 1988;45:1187–1190. doi: 10.1001/archneur.1988.00520350025009. [DOI] [PubMed] [Google Scholar]
  247. Pietropaolo S, Feldon J, Yee BK. Age-dependent phenotypic characteristics of a triple transgenic mouse model of Alzheimer's disease. Behav. Neurosci. 2008;122:733–747. doi: 10.1037/a0012520. [DOI] [PubMed] [Google Scholar]
  248. Pietropaolo S, Sun Y, Li R, Brana C, Feldon J, Yee BK. Limited impact of social isolation on Alzheimer-like symptoms in a triple transgenic model. Behav. Neurosci. 2009;123:181–195. doi: 10.1037/a0013607. [DOI] [PubMed] [Google Scholar]
  249. Pugh PL, Richardson JC, Bate ST, Upton N, Sunter D. Non-cognitive behaviours in an APP/PS1 transgenic model of Alzheimer's disease. Behav. Brain Res. 2007;178:18–28. doi: 10.1016/j.bbr.2006.11.044. [DOI] [PubMed] [Google Scholar]
  250. Puoliväli J, Wang J, Heikkinen T, Heikkilä M, Tapiola T, van Groen T, Tanila H. Hippocampal Aβ42 levels correlate with spatial memory deficit in APP and PS1 double transgenic mice. Neurobiol. Dis. 2002;9:339–347. doi: 10.1006/nbdi.2002.0481. [DOI] [PubMed] [Google Scholar]
  251. Qian S, Jiang P, Guan X-M, Singh G, Trumbauer ME, Yu H, Chen HY, Van der Ploeg LHT, Zheng H. Mutant human presenilin-1 protects presenilin 1 null mouse against embryonic lethality and elevates Aβ1–42/43 production. Neuron. 1998;20:611–617. doi: 10.1016/s0896-6273(00)80999-x. [DOI] [PubMed] [Google Scholar]
  252. Qin C, Zhu H, Zhang B, Chang Y, Yin H, Liu Y. Patho-morphological and immunohistochemical findings in brains of APP transgenic model. Acta Lab. Sci. Sinica. 2000;8:213–217. [Google Scholar]
  253. Quinn J, Montine T, Morrow J, Woodward WR, Kulhanek D, Eckenstein F. Inflammation and cerebral amyloidosis are disconnected in an animal model of Alzheimer's disease. J. Neuroimmunol. 2003;137:32–41. doi: 10.1016/s0165-5728(03)00037-7. [DOI] [PubMed] [Google Scholar]
  254. Quon D, Wang Y, Catalano R, Scardina JM, Murakami K, Cordell B. Formation of β-amyloid protein deposits in brains of transgenic mice. Nature. 1991;352:239–241. doi: 10.1038/352239a0. [DOI] [PubMed] [Google Scholar]
  255. Radde R, Bolmont T, Kaeser SA, Coomaraswamy J, Lindau D, Stoltze L, Calhoun ME, Jäggi F, Wolburg H, Gengler S, Haass C, Ghetti B, Czech C, Hölscher C, Mathews PM, Jucker M. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006;7:940–946. doi: 10.1038/sj.embor.7400784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Rainville C, Marchand N, Passini R. Performances of patients with a dementia of the Alzheimer type in the Standardized Road-Map test of Direction Sense. Neuropsychologia. 2002a;40:567–573. doi: 10.1016/s0028-3932(01)00133-6. [DOI] [PubMed] [Google Scholar]
  257. Rainville C, Amieva H, Lafont S, Dartigues JF, Orgogozo JM, Fabrigoule C. Executive function deficits in patients with dementia of the Alzheimer's type: a study with a Tower of London task. Arch. Clin. Neuropsychol. 2002b;17:513–530. [PubMed] [Google Scholar]
  258. Redwine JM, Kosofsky B, Jacobs RE, Games D, Reilly JF, Morrison JH, Young WG, Bloom FE. Dentate gyrus volume is reduced before onset of plaque formation in PDAPP mice: a magnetic resonance microscopy and stereologic analysis. Proc. Natl. Acad. Sci. U.S.A. 2003;100:1381–1386. doi: 10.1073/pnas.242746599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Reilly JF, Games D, Rydel RE, Freedman S, Schenk D, Young WG, Morrison JH, Bloom FE. Amyloid deposition in the hippocampus and entorhinal cortex: quantitative analysis of a transgenic mouse model. Proc. Natl. Acad. Sci. U.S.A. 2003;100:4837–4842. doi: 10.1073/pnas.0330745100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Reiserer RS, Harrison FE, Syverud DC, McDonald MP. Impaired spatial learning in the APPSwe+PSEN1deltaE9 bigenic mouse model of Alzheimer's disease. Genes Brain Behav. 2007;6:54–65. doi: 10.1111/j.1601-183X.2006.00221.x. [DOI] [PubMed] [Google Scholar]
  261. Richards JG, Higgins GA, Ouagazzal A-M, Ozmen L, Kew JNC, Bohrmann B, Malherbe P, Brockhaus M, Loetscher H, Czech C, Huber G, Bluethmann H, Jacobsen H, Kemp JA. PS2APP transgenic mice, coexpressing hPS2mut and hAPPswe, show age-related cognitive deficits associated with discrete brain amyloid deposition and inflammation. J. Neurosci. 2003;23:8989–9003. doi: 10.1523/JNEUROSCI.23-26-08989.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Richardson JC, Kendal CE, Anderson R, Priest F, Gower E, Soden P, Gray R, Topps S, Howlett DR, Lavender D, Clarke NJ, Barnes JC, Haworth R, Stewart MG, Rupniak HTR. Ultrastructural and behavioural changes precede amyloid deposition in a transgenic model of Alzheimer's disease. Neuroscience. 2003;122:213–228. doi: 10.1016/s0306-4522(03)00389-0. [DOI] [PubMed] [Google Scholar]
  263. Richner M, Bach G, West M. Over expression of amyloid beta-protein reduces the number of neurons in the striatum of APPswe/PS1/ΔE9. Brain Res. 2009;1266:87–92. doi: 10.1016/j.brainres.2009.02.025. [DOI] [PubMed] [Google Scholar]
  264. Roberts WW, Dember WN, Brodwick M. Alternation and exploration in rats with hippocampal lesions. J. Comp. Physiol. Psychol. 1962;55:695–700. doi: 10.1037/h0045168. [DOI] [PubMed] [Google Scholar]
  265. Rodgers RJ, Davies B, Shore R. Absence of anxiolytic response to chlordiazepoxide in two common background strains exposed to the elevated plus-maze: importance and implications of behavioural baseline. Genes Brain Behav. 2002;1:242–251. doi: 10.1034/j.1601-183x.2002.10406.x. [DOI] [PubMed] [Google Scholar]
  266. Rodrigo J, Fernández-Vizarra P, Castro-Blanco S, Bentura ML, Nieto M, Gómez-Isla T, Martinez-Murillo R, Martinez A, Serrano J, Fernández AP. Nitric oxide in the cerebral cortex of amyloid-precursor protein (SW) Tg2576 transgenic mice. Neuroscience. 2004;128:73–89. doi: 10.1016/j.neuroscience.2004.06.030. [DOI] [PubMed] [Google Scholar]
  267. Sadowski M, Pankiewicz J, Scholtzova H, Ji Y, Quartermain D, Jensen CH, Duff K, Nixon RA, Gruen RJ, Wisniewski T. Amyloid-β deposition is associated with decreased hippocampal glucose metabolism and spatial memory impairment in APP/PS1 mice. J. Neuropathol. Exp. Neurol. 2004;63:418–428. doi: 10.1093/jnen/63.5.418. [DOI] [PubMed] [Google Scholar]
  268. Sant'Angelo A, Trinchese F, Arancio O. Usefulness of behavioral and electrophysiological studies in transgenic models of Alzheimer disease. Neurochem. Res. 2003;28:1009–1015. doi: 10.1023/a:1023251005197. [DOI] [PubMed] [Google Scholar]
  269. Savonenko AV, Xu GM, Price DL, Borchelt DR, Markowska AL. Normal cognitive behavior in two distinct congenic lines of transgenic mice hyperexpresing APPSWE. Neurobiol. Dis. 2003;12:194–211. doi: 10.1016/s0969-9961(02)00012-8. [DOI] [PubMed] [Google Scholar]
  270. Savonenko A, Xu GM, Melnikova T, Morton JL, Gonzales V, Wong MP, Price DL, Tang F, Markowska AL, Borchelt DR. Episodic-like memory deficits in the APPswe/PS1dE9 mouse model of Alzheimer's disease: relationships to β-amyloid deposition and neurotransmitter abnormalities. Neurobiol. Dis. 2005;18:602–617. doi: 10.1016/j.nbd.2004.10.022. [DOI] [PubMed] [Google Scholar]
  271. Scearce-Levie K. Monitoring spatial learning and memory in Alzheimer's disease mouse models using the Morris water maze. In: Roberson ED, editor. Alzheimer's Disease and Frontotemporal Dementia (Methods Mol Biol 670) Berlin: Springer; 2011. pp. 191–205. [DOI] [PubMed] [Google Scholar]
  272. Schenk D, Masliah E, Lee M, Johnson-Wood K, Seubert P, Games D. The PDAPP transgenic mouse as an animal model for Aβ-induced amyloidosis and neuropathology. Alzheimer Dis. Rev. 1997;2:20–27. [Google Scholar]
  273. Schmalzigaug R, Rodriguiz RM, Phillips LE, Davidson CE, Wetsel WC, Premont RT. Anxiety-like behaviors in mice lacking GIT2. Neurosci. Lett. 2009;452:156–161. doi: 10.1016/j.neulet.2008.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Schmitz C, Rutten BP, Pielen A, Schafer S, Wirths O, Tremp G, Czech C, Blanchard V, Multhaup G, Rezaie P, Korr H, Steinbusch HW, Pradier L, Bayer TA. Hippocampal neuron loss exceeds amyloid plaque load in a transgenic mouse model of Alzheimer's disease. Am. J. Pathol. 2004;164:1495–1502. doi: 10.1016/S0002-9440(10)63235-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Scholtzova H, Kascsak RJ, Bates KA, Boutajangout A, Kerr DJ, Meeker HC, Mehta PD, Spinner DS, Wisniewski T. Induction of toll-like receptor 9 signaling as a method for ameliorating Alzheimer's disease-related pathology. J. Neurosci. 2009;29:1846–1854. doi: 10.1523/JNEUROSCI.5715-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Seabrook GR, Rosahl TW. Transgenic animals relevant to Alzheimer's disease. Neuropharmacology. 1999;8:1–17. doi: 10.1016/s0028-3908(98)00170-1. [DOI] [PubMed] [Google Scholar]
  277. Senanarong V, Cummings JL, Fairbanks L, Mega M, Masterman DM, O'Connor SM, Strickland TL. Agitation in Alzheimer's disease is a manifestation of frontal lobe dysfunction. Dement. Geriatr. Cogn. Disord. 2004;17:14–20. doi: 10.1159/000074080. [DOI] [PubMed] [Google Scholar]
  278. Senechal Y, Prut L, Kelly PH, Staufenbiel M, Natt F, Hoyer D, Wiessner C, Dev KK. Increased exploratory activity of APP23 mice in a novel environment is reversed by siRNA. Brain Res. 2008;1243:124–133. doi: 10.1016/j.brainres.2008.09.024. [DOI] [PubMed] [Google Scholar]
  279. Shie F-S, LeBoeur RC, Jin L-W. Early intraneuronal Aβ deposition in the hippocampus of APP transgenic mice. NeuroReport. 2003;14:123–129. doi: 10.1097/01.wnr.0000051151.87269.7d. [DOI] [PubMed] [Google Scholar]
  280. Smith GW, Aubry J-M, Dellu F, Contarino A, Bilezikjian L, Gold LH, Chen R, Marchuk Y, Hauser C, Bentley CA, Sawchenko PE, Koob GF, Vale W, Lee K-F. Corticotropin releasing factor receptor 1-deficient mice display decreased anxiety, impaired stress response, and aberrant neurendocrine development. Neuron. 1998;20:1093–1102. doi: 10.1016/s0896-6273(00)80491-2. [DOI] [PubMed] [Google Scholar]
  281. Spires TL, Hyman BT. Transgenic models of Alzheimer's disease: learning from animals. NeuroRx. 2005;2:423–437. doi: 10.1602/neurorx.2.3.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Sprott RL, Eleftheriou BE. Open-field behavior in aging inbred mice. Gerontologia. 1974;20:155–162. doi: 10.1159/000212009. [DOI] [PubMed] [Google Scholar]
  283. Stalder M, Phinney A, Probst A, Sommer B, Staufenbiel M, Jucker M. Association of microglia with amyloid plaques in brains of APP23 transgenic mice. Am. J. Pathol. 1999;154:1673–1684. doi: 10.1016/S0002-9440(10)65423-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Stein TD, Johnson JA. Lack of neurodegeneration in transgenic mice overexpressing mutant amyloid precursor protein is associated with increased levels of transthyretin and the activation of cell survival pathways. J. Neurosci. 2002;22:7380–7388. doi: 10.1523/JNEUROSCI.22-17-07380.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Stein TD, Johnson JA. Genetic programming by the proteolytic fragments of the amyloid precursor protein: somewhere between confusion and clarity. Rev. Neurosci. 2003;14:317–341. doi: 10.1515/revneuro.2003.14.4.317. [DOI] [PubMed] [Google Scholar]
  286. Stevens R, Cowey A. Effects of dorsal and ventral hippocampal lesions on spontaneous alternation, learned alternation and probability learning in rats. Brain Res. 1973;52:203–224. doi: 10.1016/0006-8993(73)90659-8. [DOI] [PubMed] [Google Scholar]
  287. Storey E, Cappai R. The amyloid precursor protein of Alzheimer's disease and the Aβ peptide. Neuropathol. Appl. Neurobiol. 1999;25:81–97. doi: 10.1046/j.1365-2990.1999.00164.x. [DOI] [PubMed] [Google Scholar]
  288. Sturchler-Pierrat C, Abramowski D, Duke M, Wiederhold K-H, Mislt C, Rothacher S, Ledermann B, Bürki K, Frey P, Paganetti PA, Waridel C, Calhoun ME, Jucker M, Probst A, Staufenbiel M, Sommer B. Two precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc. Natl. Acad. Sci. U.S.A. 1997;94:13287–13292. doi: 10.1073/pnas.94.24.13287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Sugarman MC, Yamasaki TR, Oddo S, Echegoyen JC, Murphy MP, Golde TE, Jannatipour M, Leissring MA, LaFerla FM. Inclusion body myositis-like phenotype induced by transgenic overexpression of βAPP in skeletal muscle. Proc. Natl. Acad. Sci. U.S.A. 2002;99:6334–6339. doi: 10.1073/pnas.082545599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Sykova E, Vorisek I, Antonova T, Mazel T, Meyer-Luehmann M, Jucker M, Hajek M, Or M, Bures J. Changes in extracellular space size and geometry in APP23 transgenic mice: a model of Alzheimer's disease. Proc. Natl. Acad. Sci. U.S.A. 2005;102:479–484. doi: 10.1073/pnas.0408235102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Szot P, Van Dam D, White SS, Franklin A, Staufenbiel M, De Deyn PP. Age-dependent changes in noradrenergic locus coeruleus system in wild-type and APP23 transgenic mice. Neurosci. Lett. 2009;463:93–97. doi: 10.1016/j.neulet.2009.07.055. [DOI] [PubMed] [Google Scholar]
  292. Tabuchi M, Yamaguchi T, Iizuka S, Imamura S, Ikarashi Y, Kase Y. Ameliorative effects of yokukansan, a traditional Japanese medicine, on learning and non-cognitive disturbances in the Tg2576 mouse model of Alzheimer's disease. J. Ethnopharmacol. 2009;122:157–162. doi: 10.1016/j.jep.2008.12.010. [DOI] [PubMed] [Google Scholar]
  293. Takahashi RH, Milner TA, Li F, Nam EE, Edgar MA, Yamaguchi H, Beal MF, Xu H, Greengard P, Gouras GK. Intraneuronal Alzheimer Aβ42 accumulates in multivesicular bodies and is associated with synaptic pathology. Am. J. Pathol. 2002;161:1869–1879. doi: 10.1016/s0002-9440(10)64463-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Tales A, Muir J, Jones R, Bayer A, Snowden RJ. The effects of saliency and task difficulty on visual search performance in ageing and Alzheimer's disease. Neuropsychologia. 2004;42:335–345. doi: 10.1016/j.neuropsychologia.2003.08.002. [DOI] [PubMed] [Google Scholar]
  295. Tecott LH, Logue SF, Wehner JM, Kauer JA. Perturbed dentate gyrus function in 5-HT2c receptor mutant mice. Proc. Natl. Acad. Sci. U.S.A. 1998;95:15026–15031. doi: 10.1073/pnas.95.25.15026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Terai K, Iwai A, Kawabata S, Tasaki Y, Watanabe T, Miyata K, Yamaguchi T. β-Amyloid deposits in transgenic mice expressing human βamyloid precursor protein have the same characteristics as those in Alzheimer's disease. Neuroscience. 2001;104:299–310. doi: 10.1016/s0306-4522(01)00095-1. [DOI] [PubMed] [Google Scholar]
  297. Tetewsky SJ, Duffy CJ. Visual loss and getting lost in Alzheimer's disease. Neurology. 1999;52:958–965. doi: 10.1212/wnl.52.5.958. [DOI] [PubMed] [Google Scholar]
  298. Thal DR, Rüb U, Schultz C, Sassin I, Ghebremedhin E, Tredici KD, Braak E, Braak H. Sequence of Aβ-protein deposition in the human medial temporal lobe. J. Neuropathol. Exp. Neurol. 2000;59:733–748. doi: 10.1093/jnen/59.8.733. [DOI] [PubMed] [Google Scholar]
  299. Timmer NM, van Dijk L, van der Zee CE, Kiliaan A, de Waal RM, Verbeek MM. Enoxaparin treatment administered at both early and late stages of Aβ deposition improves cognition of APPswe/PS1dE9 mice with differential effects on brain Aβ levels. Neurobiol. Dis. 2010;40:340–347. doi: 10.1016/j.nbd.2010.06.008. [DOI] [PubMed] [Google Scholar]
  300. Tiraboschi P, Hansen LA, Thal LJ, Corey-Bloom J. The importance of neuritic plaques and tangles to the development and evolution of AD. Neurology. 2004a;62:1984–1989. doi: 10.1212/01.wnl.0000129697.01779.0a. [DOI] [PubMed] [Google Scholar]
  301. Tiraboschi P, Sabbagh MN, Hansen LA, Salmon DP, Merdes A, Gamst A, Masliah E, Alford M, Thal LJ, Corey-Bloom J. Alzheimer disease without neocortical neurofibrillary tangles: "a second look". Neurology. 2004b;62:1141–1147. doi: 10.1212/01.wnl.0000118212.41542.e7. [DOI] [PubMed] [Google Scholar]
  302. Tomidokoro Y, Harigaya Y, Matsubara E, Ikeda M, Kawarabayashi T, Shirao T, Ishiguro K, Okamoto K, Younkin SG, Shoji M. Brain Aβ amyloidosis in APPSW mice induces accumulation of presenilin-1 and tau. J. Pathol. 2001;193:500–506. doi: 10.1002/path.897. [DOI] [PubMed] [Google Scholar]
  303. Touma C, Ambrée O, Görtz N, Keyvani K, Lewejohann L, Palme R, Paulus W, Schwarze-Eicker K, Sachser N. Age- and sex-dependent development of adrenocortical hyperactivity in a transgenic mouse model of Alzheimer's disease. Neurobiol. Aging. 2004;25:893–904. doi: 10.1016/j.neurobiolaging.2003.09.004. [DOI] [PubMed] [Google Scholar]
  304. Treit D, Menard J. Dissociations among the anxiolytic effects of septal, hippocampal, and amygdaloid lesions. Behav. Neurosci. 1997;111:653–658. doi: 10.1037//0735-7044.111.3.653. [DOI] [PubMed] [Google Scholar]
  305. Tremml P, Lipp H-P, Müller U, Ricceri L, Wolfer DP. Neurobehavioral development, adult openfield exploration and swimming navigation learning in mice with a modified β-amyloid-precursor protein gene. Behav. Brain Res. 1998;95:65–76. doi: 10.1016/s0166-4328(97)00211-8. [DOI] [PubMed] [Google Scholar]
  306. Turner PR, O'Connor K, Tate WP, Abraham WC. Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog. Neurobiol. 2003;70:1–32. doi: 10.1016/s0301-0082(03)00089-3. [DOI] [PubMed] [Google Scholar]
  307. Uc EY, Rizzo M, Anderson SW, Shi Q, Dawson JD. Driver route-following and safety errors in early Alzheimer disease. Neurology. 2004;63:832–837. doi: 10.1212/01.wnl.0000139301.01177.35. [DOI] [PubMed] [Google Scholar]
  308. Ueki A, Goto K, Sato N, Iso H, Morita Y. Prepulse inhibition of acoustic startle response in mild cognitive impairment and mild dementia of Alzheimer type. Psychiatry Clin. Neurosci. 2006;60:55–62. doi: 10.1111/j.1440-1819.2006.01460.x. [DOI] [PubMed] [Google Scholar]
  309. Van Dam D, D'Hooge R, Staufenbiel M, Van Ginneken C, Van Meir F, De Deyn PP. Age-dependent cognitive decline in the APP23 model precedes amyloid deposition. Eur. J. Neurosci. 2003;17:388–396. doi: 10.1046/j.1460-9568.2003.02444.x. [DOI] [PubMed] [Google Scholar]
  310. Van Dam D, Vloeberghs E, Abramowski D, Staufenbiel M, De Deyn PP. APP23 Mice as a model of Alzheimer's disease: an example of a transgenic approach to modeling a CNS disorder. CNS Spectr. 2005;10:207–222. doi: 10.1017/s1092852900010051. [DOI] [PubMed] [Google Scholar]
  311. Van Dorpe J, Smeijers L, Wewachter I, Nuens D, Spittaels K, Van den Haute C, Mercken M, Moechars D, Laenen I, Kuiperi C, Bruyseels K, Tesseur I, Loos R, Vanderstichele H, Checler F, Sciot R, Van Leuven F. Prominent cerebral amyloid angiopathy in transgenic mice overexpressing the London mutant of human APP in neurons. Am. J. Pathol. 2000;157:1283–1298. doi: 10.1016/S0002-9440(10)64644-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. van Leuven FF. Single and multiple transgenic mice as models for Alzheimer's disease. Prog. Neurobiol. 2000;61:305–312. doi: 10.1016/s0301-0082(99)00055-6. [DOI] [PubMed] [Google Scholar]
  313. Vloeberghs E, Van Dam D, Franck F, Staufenbiel M, De Deyn PP. Mood and male sexual behaviour in the APP23 model of Alzheimer's disease. Behav. Brain Res. 2007;180:146–151. doi: 10.1016/j.bbr.2007.03.002. [DOI] [PubMed] [Google Scholar]
  314. Vloeberghs E, Van Dam D, Engelborghs S, Nagels G, Staufenbiel M, De Deyn PP. Altered circadian locomotor activity in APP23 mice: a model for BPSD disturbances. Eur. J. Neurosci. 2004;20:2757–2766. doi: 10.1111/j.1460-9568.2004.03755.x. [DOI] [PubMed] [Google Scholar]
  315. Walker JM, Fowler SW, Miller DK, Sun AY, Weisman GA, Wood WG, Sun GY, Simonyi A, Schachtman TR. Spatial learning and memory impairment and increased locomotion in a transgenic amyloid precursor protein mouse model of Alzheimer's disease. Behav. Brain Res. 2011;222:169–175. doi: 10.1016/j.bbr.2011.03.049. [DOI] [PubMed] [Google Scholar]
  316. Walsh RN, Cummins RA. The open-field test: a critical review. Psychol. Bull. 1976;83:482–504. [PubMed] [Google Scholar]
  317. Wang J, Tanila H, Puoliväli J, Kadish I, van Groen T. Gender differences in the amount and deposition of amyloidβ in APPswe and PS1 double transgenic mice. Neurobiol. Dis. 2003;14:318–327. doi: 10.1016/j.nbd.2003.08.009. [DOI] [PubMed] [Google Scholar]
  318. Wegiel J, Imaki H, Wang K-C, Wegiel T, Wronska A, Osuchowski M, Rubenstein R. Origin and turnover of microglial cells in fibrillar plaques of APPsw transgenic mice. Acta Neuropathol. 2003;105:392–402. doi: 10.1007/s00401-002-0660-3. [DOI] [PubMed] [Google Scholar]
  319. Wegiel J, Wang K-C, Imaki H, Rubenstein R, Wronska A, Osuchowski M, Lipinski WJ, Walker LC, LeVine H. The role of microglial cells and astrocytes in fibrillar plaque evolution in transgene APPSW mice. Neurobiol. Aging. 2001;22:49–61. doi: 10.1016/s0197-4580(00)00181-0. [DOI] [PubMed] [Google Scholar]
  320. Weintraub S, Mesulam MM. Four neuropsychological profiles in dementia. In: Spinnler H, Boller F, editors. Handbook of Neuropsychology. vol. 8. Amsterdam: Elsevier; 1993. pp. 253–282. [Google Scholar]
  321. Weiss C, Venkatasubramanian PN, Aguado AS, Power JM, Tom BC, Li L, Chen KS, Disterhoft JF, Wyrwicz AM. Impaired eyeblink conditioning and decreased hippocampal volume in PDAPP V717F mice. Neurobiol. Dis. 2002;11:425–433. doi: 10.1006/nbdi.2002.0555. [DOI] [PubMed] [Google Scholar]
  322. Westerman MA, Cooper-Blacketer D, Mariash A, Kotilinek L, Kawarabayashi T, Younkin LH, Carlson GA, Younkin SG, Ashe KH. The relationship between Aβ and memory in the Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 2002;22:1858–1867. doi: 10.1523/JNEUROSCI.22-05-01858.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Whitford GM. Alzheimer's disease and serotonin: a review. Neuropsychobiology. 1986;15:133–142. doi: 10.1159/000118256. [DOI] [PubMed] [Google Scholar]
  324. Winkler DT, Bondolfi L, Herzig MC, Jann L, Calhoun ME, Wiederhold K-H, Tolnay M, Staufenbiel M, Jucker M. Spontaneous hemorrhagic stroke in a mouse model of cerebral amyloid angiopathy. J. Neurosci. 2001;21:1619–1627. doi: 10.1523/JNEUROSCI.21-05-01619.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Wirths O, Bayer TA. Neuron loss in transgenic mouse models of Alzheimer's disease. Int. J. Alzheimers Dis. 2010 doi: 10.4061/2010/723782. pii: 723782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Wirths O, Breyhan H, Schäfer S, Roth C, Bayer TA. Deficits in working memory and motor performance in the APP/PS1ki mouse model for Alzheimer's disease. Neurobiol. Aging. 2008;29:891–901. doi: 10.1016/j.neurobiolaging.2006.12.004. [DOI] [PubMed] [Google Scholar]
  327. Woodruff-Pak DS, Papka M. Alzheimer's disease and eyeblink conditioning: 750 ms trace vs. 400 ms delay paradigm. Neurobiol. Aging. 1996;17:397–404. doi: 10.1016/0197-4580(96)00022-x. [DOI] [PubMed] [Google Scholar]
  328. Woodruff-Pak DS, Romano S, Papka M. Training to criterion in eyeblink classical conditioning in Alzheimer's disease, Down's syndrome with Alzheimer's disease, and healthy elderly. Behav. Neurosci. 1996;110:22–29. doi: 10.1037//0735-7044.110.1.22. [DOI] [PubMed] [Google Scholar]
  329. Wu C-C, Chawla F, Games D, Rydel RE, Freedman S, Schenk D, Young WG, Morrison JH, Bloom FE. Selective vulnerability of dentate granule cells prior to amyloid deposition in PDAPP mice: digital morphometric analyses. Proc. Natl. Acad. Sci. U.S.A. 2004;101:7141–7146. doi: 10.1073/pnas.0402147101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Yamaguchi F, Richards S-J, Beyruther K, Salbaum M, Carlson GA, Dunnett SB. Transgenic mice for the amyloid precursor protein 695 isoform have impaired spatial memory. NeuroReport. 1991;2:781–784. doi: 10.1097/00001756-199112000-00013. [DOI] [PubMed] [Google Scholar]

RESOURCES