Abstract
The rising prevalence of Alzheimer’s disease (AD) is rapidly becoming one of the largest health and economic challenges in the world. There is a growing need for the development and implementation of reliable biomarkers for AD that can be used to assist in diagnosis, inform disease progression, and monitor therapeutic efficacy. Preclinical models permit the evaluation of candidate biomarkers and assessment of pipeline agents before clinical trials are initiated and provide a translational opportunity to advance biomarker discovery. Fast and inexpensive data can be obtained from examination of peripheral markers, though they currently lack the sensitivity and consistency of imaging techniques such as MRI or PET. Plasma and cerebrospinal fluid (CSF) biomarkers in animal models can assist in development and implementation of similar approaches in clinical populations. These biomarkers may also be invaluable in decisions to advance a treatment to human testing. Longitudinal studies in AD models can determine initial presentation and progression of biomarkers that may also be used to evaluate disease-modifying efficacy of drugs. The refinement of biomarker approaches in preclinical systems will not only aid in drug development, but may facilitate diagnosis and disease monitoring in AD patients.
Keywords: Alzheimer’s disease, biomarkers, animal models, drug development
Introduction
Alzheimer’s disease (AD) represents a looming crisis as the number of victims continues to increase and limited treatments are available. There is a critical need for advances in the treatment of AD and in the tools used to detect cases early and monitor progression. Studies of AD populations show that biomarkers, including imaging approaches [1-3] and evaluation of cerebrospinal fluid (CSF) and blood [4-6], have value in diagnosis and tracking disease progression. Numerous candidate markers have been identified such as CSF or plasma levels of Beta-amyloid protein (Aβ) and phosphorylated tau (ptau); however, many lack the specificity necessary to be used diagnostically or do not correlate with clinical progression. An opportunity exists with the animal model systems that are employed in the study of AD mechanisms to examine potential biomarkers. Many of the investigations of mechanisms in AD also provide suitable targets for evaluation of novel treatment efficacy in both animal models and clinical populations. Investigation of potential biomarkers in animal model systems lends itself to examination of mechanisms underlying correspondence between central pathology and peripheral markers. Moreover, relationships in animal models can be explored between peripheral biomarkers and readily available neuropathology; these can be translated into human studies where biomarkers are accessible but neuropathology is often not.
Biomarkers can assist in monitoring the progression of AD and inform treatment approaches as the disease advances. Aβ targets, for example, may be more appropriate very early in the disease course and tau targets may be more relevant later in the disease. The complex evaluation and staging of treatment of AD requires assessment in animal models exhibiting core pathological features.
In this review we highlight several candidate biomarkers in AD that have been evaluated in preclinical animal models. Several potential biomarkers have been suggested related to core disease pathologies (Aβ and tau), as well as those associated with AD risk factors (such as microglial and immune activation). We also discuss the need for greater evaluation of candidate biomarkers in preclinical systems in parallel with the investigations of brain pathology that is the central approach to animal models of AD.
Peripheral markers
The amyloid cascade hypothesis posits that early accumulation of Aβ leads to synaptic dysfunction, neurodegeneration, and cognitive deficits [7,8]. Although substantial evidence exists to support this hypothesis, the mechanisms underlying the pathogenic contributions of Aβ remain elusive. Whether the deposition of Aβ into insoluble plaques or smaller, soluble species of Aβ drive neurodegeneration is unclear; however, recent evidence suggests Aβ oligomers (oAβ) may be more neurotoxic [9-11]. Early work with Tg2576 mice demonstrated a reduction in CSF and plasma levels of Aβ42 [12], findings which mirror AD patient data. In PDAPP mice, Aβ42 levels positively correlated with the abundance of plaques [13], contrary to what has been found in AD [14]. Canines represent a strong model for investigating disease-state mechanisms as they develop age-related Aβ deposition and cognitive deficits, which show similarities to human AD [15]. CSF Aβ42 and oAβ levels decreased with age and inversely correlated with plaque load, indicating these markers may reflect brain amyloidosis [16]. These observations mirror those in humans. In aged canines with mild cognitive impairment (MCI), plasma Aβ42 levels were increased compared to unimpaired or severely impaired dogs [17].
The detection and quantification of Aβ species in vivo has encountered difficulties, and questions have been raised about the feasibility of reliably measuring oligomers in CSF [18]. Specific ELISAs have been developed for the quantification of Aβ levels, which have been demonstrated to increase with age in the brains of transgenic (Tg) AD mice [18,19]. However, depending on the specific antibodies utilized, oAβ has not been consistently detected in human CSF [18,20]. Using flow cytometry, the increased presence of Aβ has been reported in the CSF of AD patients compared to controls [21]. The reliable quantification of Aβ species from biological fluids could serve as an important measure of therapeutic efficacy in preclinical models, underscoring the import of the development of a precise detection technique.
One of the principal enzymes responsible for amyloidogenesis, β-site amyloid precursor protein cleaving enzyme 1 (BACE1), has promise as a therapeutic target. Novel techniques for quantifying BACE1 levels from the CSF and plasma are emerging, permitting a primary measure of target engagement [22,23]. Inhibitors of BACE1 decreased CSF and plasma levels of Aβ40 and Aβ42 in mice, guinea pigs, and non-human primates [23-26]. Levels of soluble amyloid precursor protein β (sAPPβ) are also decreased in plasma following BACE inhibition in rhesus monkeys [23], while mixed results have been observed from human AD plasma [23,27]. An inhibitor of cathepsin B, one of the enzymes in the β-secretase complex along with BACE1 [28], reduced Aβ40 and Aβ42 in the CSF and plasma of guinea pigs [29]. Extending these findings and approaches to Tg models of AD could offer additional insight into therapeutic candidate efficacy and mechanisms of disease. BACE1 activity can be measured in human CSF permitting it to be used as a measure of target engagement by inhibitors of this enzyme.
Inhibition of gamma-secretase has been a target of therapeutic development despite initial high-profile failures [30]; more recent drugs have attempted to avoid Notch-related toxicity and side effects [31,32]. Peripheral measurement of Aβ following administration of gamma-secretase modulators could provide a fast and reliable readout in addition to behavioral outcomes. Gamma-secretase modulators and inhibitors have been shown to reduce CSF and plasma Aβ levels in non-Tg rats and guinea pigs [33-35], reinforcing the utility of investigating peripheral biomarkers in animals and clinical trials. Using stable-isotope-labeling kinetics (SILK) methods, a gamma-secretase inhibitor was shown to reduce CSF Aβ production in rhesus monkeys without a subsequent rise in Aβ production [36]. Because gamma-secretase inhibitors lead to altered proteolysis of APP and the generation of shorter Aβ fragments (i.e. Aβ (1-15)), assays that target these isoforms may provide a reliable measure of drug activity, as has been demonstrated in canine CSF [37].
A number of immunotherapies have been developed and tested in AD models, most of which target amyloid pathology. Central immunotherapy may produce secondary, undesirable immune responses, and it is important to evaluate markers of inflammation as possible indicators of an adverse response. Plasma levels of interleukin (IL)-10, an anti-inflammatory cytokine, have been found to be increased following Aβ immunotherapy in Tg2576 mice [38,39]. In contrast, T cell infusions specific for Aβ or administration of granulocyte colony stimulating factor (GM-CSF) reduced plasma levels of IL-4, TNF-α, and other cytokines [40,41]. Inflammatory mechanisms and their peripheral markers can be further explored with parallel observations in humans and animals.
Isoprostanes, which reflect lipid peroxidation and oxidative stress [42], are elevated in plasma from Tg2576 mice in advance of plaque formation [43], suggesting isoprostane levels may have utility as a predictive biomarker.
Microgliosis is an important indicator of drug activity and a common pathological finding in AD [44]. Although microglial activity may be assessed with neuroimaging techniques, important information can be quickly and inexpensively obtained through peripheral measures. In rhesus monkey plasma, a non-viral Aβ vaccine did not alter chemokine (C-C motif) ligand 2 (CCL2) expression [45]. More studies may benefit from investigation of microglial markers in the future. However, care must be taken in the interpretation of immune responses from mice as recent evidence suggests they may not translate well to human inflammatory diseases [46].
Currently the measurement of tau and phosphorylated tau (p-tau) from human or animal fluids is restricted to the CSF. Studies with Tg mice or rats have demonstrated elevated CSF p-tau and tau levels with age [47,48]. Recent work has indicated promising data in the development of assays to reliably quantify ptau and tau from plasma and serum that would significantly advance the utility of blood-based markers [49].
Imaging
Imaging approaches in AD models have demonstrated strong correspondence with findings from AD populations, suggesting these techniques may have considerable translational utility. Imaging studies may complement behavioral and postmortem readouts in animals, especially in the evaluation of treatments designed to target different stages of pathological severity. Several imaging approaches have been employed in animal models of AD, including positron emission tomography (PET), magnetic resonance imaging (MRI), and magnetic resonance spectroscopy (MRS).
Fluorodeoxyglucose (FDG)-PET imaging shows consistent patterns in AD making it a suitable measure of drug efficacy in patients and animal models. Similar to AD, reduced FDG uptake has been observed in PDAPP mice [50-52], PSAPP mice [53], 3xTg-AD mice [54], and PLB1 mice [55]. However, null effects in other studies suggest the small size of mouse brains may limit the utility of FDG-microPET in mice [56,57].
Radiolabeled amyloid imaging agents provide a measure of plaque load and may serve to longitudinally track changes in amyloidosis during clinical trials. Despite initial difficulty with uptake and retention of Pittsburgh compound B (PIB) in Tg AD mice [58,59], recent studies indicate PSAPP mice display strong, age-related amyloid loads with PIB [60], while others have demonstrated that a high specific radioactivity facilitates PIB-microPET imaging in APP23 mice [61,62]. Additional investigations have demonstrated utility in voxel-based analyses of plaque load using PIB in APP/PS1 double transgenic mice [63]. Work with monkeys shows that amyloid burden can be observed with PIB in multiple species [64]. An alternative radiotracer, 2-(1-{6-[(2-18F-fluoroethyl)(methyl) amino]-2-naphthyl}ethylidene)malononitrile (FDDNP), binds to plaques and tangles, displaying increased retention in aged macaques [64] and Tg AD rats [65]. FDDNP binding to tangles has not been demonstrated in animal models; another ligand --- 18F-THK523 --- binds selectively to tangles in Tg mice expressing mutant tau [66].
A novel amyloid imaging probe recently approved by the US Food and Drug Administration (FDA) for use in PET imaging, florbetapir (AmyvidTM), shows robust labeling in monkeys and PSAPP mice [67]. Another study with florbetapir revealed strong retention in the cortex, hippocampus, and striatum of PSAPP mice [68], areas rich in plaques. Each of these amyloid imaging agents could act as signpost markers of disease progression or determine Aβ immunotherapy efficacy. There is a consistent inverse correlation between CSF Aβ levels and brain amyloid load as demonstrated with amyloid imaging, indicating that the fluid marker is a good guide to this central aspect of AD.
The microglial probe 11C-(R)-PK11195 binds to the 18 kDa translocator protein (TSPO), also known as the peripheral benzodiazepine receptor, which reflects neuroinflammation [69]. PSAPP mice display progressively increased retention of this ligand, which mirrored the postmortem abundance of microglia [70]. Work with other TSPO probes suggests microgliosis may be more closely associated with tau pathology than amyloid pathology in mutant tau or APP mouse models, respectively [71].
MRI provides superior spatial resolution compared to PET, indicating it may be better suited to microimaging in Tg mice. Similar to AD populations, several Tg AD mouse lines have shown reduced volume of several brain structures including the hippocampus and cortex [72-77]. Other approaches to model AD also revealed brain atrophy with MRI, including aged rabbits chronically administered Aβ42 [78] and monkeys administered streptozotocin, a drug that disrupts insulin signaling and induces a diabetes phenotype and AD pathologies [79]. Because MRI-detected atrophy is progressive, a staging could be established which correlates with tau pathology or other markers of neurodegeneration and permits the evaluation of various stage-dependent therapeutics (i.e. MCI vs. advanced AD).
A variation on MRI, arterial spin labeling (ASL), is able to quantify differences in regional cerebral blood flow, which is typically reduced in AD [80-82]. Similarly, Tg mouse models of AD display cerebral hypoperfusion compared to wild-type (WT) mice during ASL-MRI [83,84]. Combined with other imaging techniques or fluid biomarkers, ASL-MRI may assist in diagnosis and evaluation of clinical trial outcomes.
Using MRI, amyloid-specific contrast agents can visualize in vivo plaque load. A variety of approaches have been used to image plaques in Tg AD mice by coupling amyloid compounds to specific probes, including gadolinium to Aβ [85-89], 19F or 1H amyloidophilic agents [90,91], and nanoparticle-based probes [92-94]. Although these compounds have not yet been tested in AD patients, their utility for measuring amyloid burden during testing of pipeline agents should be investigated. The enhanced spatial frequency associated with MRI makes it preferable to PET-based probes, especially for detecting subtle, region-specific differences. Plaques have also been visualized in AD mouse models without the help of a contrast agent because of their high iron content using high field intensity MRI [95-99]. Concerns about the consistency of interplaque metal content and non-specific arterial binding limit the application of this approach [99,100].
Deficits in axonal transport have been discovered in Tg2576 and 3xTg-AD mice before Aβ and tau pathology with manganese-enhanced MRI (MEMRI) [101,102]. Although it has not been demonstrated that these changes can be visualized in AD, this technique may prove useful for detecting early neuronal impairments in at-risk, aged individuals. Using ultra-high field diffusion tensor imaging (DTI), gray and white matter degeneration were observed in PSAPP mice [103], similar to what has been found in AD brains.
Proton magnetic resonance spectroscopy (1H-MRS) quantifies neurochemical biomarkers that are different in AD compared to age-matched controls [104,105]. AD mouse models show a similar pattern of metabolite changes [106], with decreased levels of N-acetyl aspartate (NAA) and increased levels of taurine [107]. Other studies show increased ratios of myoinositol and decreased ratios of NAA levels compared to total creatine [108-112]. Because NAA levels likely reflect neuronal viability [113], 1H-MRS data may serve as an early marker for neurodegeneration or conversely, as a measure of the neuroprotective potential of therapeutic agents. Accordingly, chronic administration of non-steroidal anti-inflammatory drugs (NSAIDs) to aged PSAPP mice mitigated the decrease in NAA and glutamate levels, while reducing plaque burden [114].
Atrophy as measured by MRI and reduced metabolism as assessed by FDG-PET progress in the course of AD and correlate with cognitive decline. This contrasts with amyloid PET where the burden of pathology is relatively stable throughout the MCI and dementia phases of AD. This may make MRI and FDG-PET more relevant as outcome measures for clinical trials, especially for agents not targeting amyloid-related processes.
Future directions
Proteomic approaches allow the unbiased investigation of a multitude of fluid biomarkers simultaneously in order to increase diagnostic and prognostic accuracy, as well as identify novel targets. With advances in mass spectrometry (MS) and microarray techniques, many groups have recently undertaken proteomic examinations in the CSF and serum/plasma of AD and MCI patients. Although some results have been inconsistent, levels of several proteins have been found to differentiate AD compared to controls, including various apolipoproteins, α1-antitrypsin, and β2-microglobulin [115-121]. Proteomic analyses have been used to differentiate AD from other dementias [122] and predict progression of MCI to AD [123]. Although proteome-based studies of plasma and serum suggest AD may be sensitively characterized with this approach [124-128], few of these reports have been replicated or confirmed.
Proteomic analyses in animal systems are preliminary. A study in tau Tg mice investigated the proteome in blood plasma at a presymptomatic and symptomatic age. A few proteins were identified as potential biomarker candidates such as adenosine triphosphate (ATP) synthase and adenosine kinase [129]. Examining the proteome both in multiple preclinical models and AD could lead to the characterization of a biomarker panel specific to the disorder.
MicroRNAs (miRNAs) have also been implicated in AD pathogenesis and suggested as a putative biomarker [130,131]. In non-Tg mice fed a high-fat diet, decreased expression of multiple miRNAs was observed in the serum [132]. Substantial translational work is required before miRNAs can be used in the clinic; however, the approach is advancing rapidly.
Alternative approaches have emerged recently that have considerable promise in investigations of cellular variations in core pathological markers. For example, attention is being directed towards the measurement and understanding of post-translational modifications of Aβ. Tg mice carrying the Arctic APP mutation provide an opportunity to study protofibrillar Aβ42; the development of protofibril-specific assays has permitted the quantification of CSF protofibrillar Aβ in these mice [133,134].
Conclusions
The use of several animal model systems has provided invaluable data regarding AD pathologies and mechanisms involved in neurodegeneration. In this research, the primary focus has been on the examination of central tissues, and to a lesser extent on peripheral markers. For the investigations that have included peripheral measures, considerable progress has been made in the identification of needed biomarkers in AD. The literature documents the extensive effort that is underway in animal model systems to associate core pathological markers with less invasive and peripheral markers that may translate to the clinic readily (see Table 1). Given the progress that has been made, a greater emphasis in probing both central and peripheral tissues in multiple animal model systems would be advantageous. This approach may also greatly accelerate translational efforts to impact clinical research. Equally pressing is the need for more data from animal systems investigating biomarkers that can be directly translated to human biomarkers. Continued progress is needed in investigations of the same and over lapping markers in animal models and clinical populations to serve as a translational bridge between animal models systems and clinical populations [135]. Given the emphasis on multiple brain regions in histological analyses of AD animal models and the speed at which studies can be carried out, a directed effort of evaluating candidate biomarkers in parallel with central markers has tremendous potential to enlighten clinical AD biomarker approaches.
Table 1.
Biomarker | Reference(s) |
---|---|
Aβ42 | [12,13,16,17,23-26,29,33-35] |
Aβ oligomers | [16] |
Protofibrillar Aβ42 | [133,134] |
Soluble APPβ | [23] |
Fragmented Aβ | [37] |
Cytokines | [38-41] |
Isoprostanes | [43] |
Tau | [47] |
Ptau | [48] |
FDG-PET | [50-57] |
Pittsburgh Compound B | [58-64] |
FDDNP | [64,65] |
18F-THK523 | [66] |
Florbetapir | [67,68] |
Microglial probes | [70,71] |
MRI-based atrophy | [72-79] |
ASL-MRI | [83,84] |
MRI amyloid contrast agents | [85-94] |
MRI amyloid without contrast | [95-99] |
Axonal transport via MEMRI | [101,102] |
DTI | [103] |
1H-MRS metabolites | [106-112,114] |
Proteome | [129] |
MicroRNAs | [132] |
Disclosure of conflict of interest
Dr. Cummings has provided consultation to Abbott, Acadia, ADAMAS, Anavex, Avanir, Baxter, Bristol-Myers Squibb, Eisai, EnVivo, Forest, Genentech, GlaxoSmithKline, Grifols, Janssen, Lilly, Medtronics, Merck, Novartis, Pain Therapeutics, Pfizer, Prana, QR, Sanofi, Sonexa, Takeda, and Toyama pharmaceutical companies.
References
- 1.Chao LL, Buckley ST, Kornak J, Schuff N, Madison C, Yaffe K, Miller BL, Kramer JH, Weiner MW. ASL perfusion MRI predicts cognitive decline and conversion from MCI to dementia. Alzheimer Dis Assoc Disord. 2010;24:19–27. doi: 10.1097/WAD.0b013e3181b4f736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Jack CR Jr, Lowe VJ, Weigand SD, Wiste HJ, Senjem ML, Knopman DS, Shiung MM, Gunter JL, Boeve BF, Kemp BJ, Weiner M, Petersen RC. Alzheimer’s Disease Neuroimaging Initiative. Serial PIB and MRI in normal, mild cognitive impairment and alzheimer’s disease: Implications for sequence of pathological events in alzheimer’s disease. Brain. 2009;132:1355–65. doi: 10.1093/brain/awp062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Liu E, Morris JC, Petersen RC, Saykin AJ, Schmidt ME, Shaw L, Siuciak JA, Soares H, Toga AW, Trojanowski JQ. Alzheimer’s Disease Neuroimaging Initiative. The alzheimer’s disease neuroimaging initiative: A review of papers published since its inception. Alzheimers Dement. 2012;8(Suppl 1):S1–68. doi: 10.1016/j.jalz.2011.09.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Blennow K. Cerebrospinal fluid protein biomarkers for alzheimer’s disease. NeuroRx. 2004;1:213–25. doi: 10.1602/neurorx.1.2.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cummings JL. Biomarkers in alzheimer’s disease drug development. Alzheimers Dement. 2011;7:e13–44. doi: 10.1016/j.jalz.2010.06.004. [DOI] [PubMed] [Google Scholar]
- 6.Hansson O, Zetterberg H, Vanmechelen E, Vanderstichele H, Andreasson U, Londos E, Wallin A, Minthon L, Blennow K. Evaluation of plasma abeta(40) and abeta(42) as predictors of conversion to alzheimer’s disease in patients with mild cognitive impairment. Neurobiol Aging. 2010;31:357–67. doi: 10.1016/j.neurobiolaging.2008.03.027. [DOI] [PubMed] [Google Scholar]
- 7.Hardy JA, Higgins GA. Alzheimer’s disease: The amyloid cascade hypothesis. Science. 1992;256:184–5. doi: 10.1126/science.1566067. [DOI] [PubMed] [Google Scholar]
- 8.Hardy J. Alzheimer’s disease: The amyloid cascade hypothesis: An update and reappraisal. J Alzheimers Dis. 2006;9(Suppl 3):151–3. doi: 10.3233/jad-2006-9s317. [DOI] [PubMed] [Google Scholar]
- 9.Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C, Finch CE, Krafft GA, Klein WL. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448–53. doi: 10.1073/pnas.95.11.6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Selkoe DJ. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav Brain Res. 2008;192:106–13. doi: 10.1016/j.bbr.2008.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–9. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
- 12.Kawarabayashi T, Younkin LH, Saido TC, Shoji M, Ashe KH, Younkin SG. Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of alzheimer’s disease. J Neurosci. 2001;21:372–81. doi: 10.1523/JNEUROSCI.21-02-00372.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.DeMattos RB, Bales KR, Parsadanian M, O’Dell MA, Foss EM, Paul SM, Holtzman DM. Plaque-associated disruption of CSF and plasma amyloid-beta (abeta) equilibrium in a mouse model of alzheimer’s disease. J Neurochem. 2002;81:229–36. doi: 10.1046/j.1471-4159.2002.00889.x. [DOI] [PubMed] [Google Scholar]
- 14.Fagan AM, Mintun MA, Mach RH, Lee SY, Dence CS, Shah AR, LaRossa GN, Spinner ML, Klunk WE, Mathis CA, DeKosky ST, Morris JC, Holtzman DM. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta42 in humans. Ann Neurol. 2006;59:512–9. doi: 10.1002/ana.20730. [DOI] [PubMed] [Google Scholar]
- 15.Cummings BJ, Head E, Afagh AJ, Milgram NW, Cotman CW. Beta-amyloid accumulation correlates with cognitive dysfunction in the aged canine. Neurobiol Learn Mem. 1996;66:11–23. doi: 10.1006/nlme.1996.0039. [DOI] [PubMed] [Google Scholar]
- 16.Head E, Pop V, Sarsoza F, Kayed R, Beckett TL, Studzinski CM, Tomic JL, Glabe CG, Murphy MP. Amyloid-beta peptide and oligomers in the brain and cerebrospinal fluid of aged canines. J Alzheimers Dis. 2010;20:637–46. doi: 10.3233/JAD-2010-1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gonzalez-Martinez A, Rosado B, Pesini P, Suarez ML, Santamarina G, Garcia-Belenguer S, Villegas A, Monleon I, Sarasa M. Plasma beta-amyloid peptides in canine aging and cognitive dysfunction as a model of alzheimer’s disease. Exp Gerontol. 2011;46:590–6. doi: 10.1016/j.exger.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 18.Yang T, Hong S, O’Malley T, Sperling RA, Walsh DM, Selkoe DJ. New ELISAs with high specificity for soluble oligomers of amyloid beta-protein detect natural abeta oligomers in human brain but not CSF. Alzheimers Dement. 2013;9:99–112. doi: 10.1016/j.jalz.2012.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bruggink KA, Jongbloed W, Biemans EA, Veerhuis R, Claassen JA, Kuiperij HB, Verbeek MM. Amyloid-beta oligomer detection by ELISA in cerebrospinal fluid and brain tissue. Anal Biochem. 2013;433:112–20. doi: 10.1016/j.ab.2012.09.014. [DOI] [PubMed] [Google Scholar]
- 20.Tai LM, Bilousova T, Jungbauer L, Roeske SK, Youmans KL, Yu C, Poon WW, Cornwell LB, Miller CA, Vinters HV, Van Eldik LJ, Fardo DW, Estus S, Bu G, Gylys KH, Ladu MJ. Levels of soluble apolipoprotein E/amyloid-beta (abeta) complex are reduced and oligomeric abeta increased with APOE4 and alzheimer disease in a transgenic mouse model and human samples. J Biol Chem. 2013;288:5914–26. doi: 10.1074/jbc.M112.442103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Santos AN, Ewers M, Minthon L, Simm A, Silber RE, Blennow K, Prvulovic D, Hansson O, Hampel H. Amyloid-beta oligomers in cerebrospinal fluid are associated with cognitive decline in patients with alzheimer’s disease. J Alzheimers Dis. 2012;29:171–6. doi: 10.3233/JAD-2012-111361. [DOI] [PubMed] [Google Scholar]
- 22.Gonzales A, Decourt B, Walker A, Condjella R, Nural H, Sabbagh MN. Development of a specific ELISA to measure BACE1 levels in human tissues. J Neurosci Methods. 2011;202:70–6. doi: 10.1016/j.jneumeth.2011.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wu G, Sankaranarayanan S, Wong J, Tugusheva K, Michener MS, Shi X, Cook JJ, Simon AJ, Savage MJ. Characterization of plasma beta-secretase (BACE1) activity and soluble amyloid precursor proteins as potential biomarkers for alzheimer’s disease. J Neurosci Res. 2012;90:2247–58. doi: 10.1002/jnr.23122. [DOI] [PubMed] [Google Scholar]
- 24.Gravenfors Y, Viklund J, Blid J, Ginman T, Karlstrom S, Kihlstrom J, Kolmodin K, Lindstrom J, von Berg S, von Kieseritzky F, Bogar K, Slivo C, Swahn BM, Olsson LL, Johansson P, Eketjall S, Falting J, Jeppsson F, Stromberg K, Janson J, Rahm F. New aminoimidazoles as beta-secretase (BACE-1) inhibitors showing amyloid-beta (abeta) lowering in brain. J Med Chem. 2012;55:9297–311. doi: 10.1021/jm300991n. [DOI] [PubMed] [Google Scholar]
- 25.Jeppsson F, Eketjall S, Janson J, Karlstrom S, Gustavsson S, Olsson LL, Radesater AC, Ploeger B, Cebers G, Kolmodin K, Swahn BM, von Berg S, Bueters T, Falting J. Discovery of AZD3839, a potent and selective BACE1 inhibitor clinical candidate for the treatment of alzheimer disease. J Biol Chem. 2012;287:41245–57. doi: 10.1074/jbc.M112.409110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sankaranarayanan S, Holahan MA, Colussi D, Crouthamel MC, Devanarayan V, Ellis J, Espeseth A, Gates AT, Graham SL, Gregro AR, Hazuda D, Hochman JH, Holloway K, Jin L, Kahana J, Lai MT, Lineberger J, McGaughey G, Moore KP, Nantermet P, Pietrak B, Price EA, Rajapakse H, Stauffer S, Steinbeiser MA, Seabrook G, Selnick HG, Shi XP, Stanton MG, Swestock J, Tugusheva K, Tyler KX, Vacca JP, Wong J, Wu G, Xu M, Cook JJ, Simon AJ. First demonstration of cerebrospinal fluid and plasma A beta lowering with oral administration of a beta-site amyloid precursor protein-cleaving enzyme 1 inhibitor in nonhuman primates. J Pharmacol Exp Ther. 2009;328:131–40. doi: 10.1124/jpet.108.143628. [DOI] [PubMed] [Google Scholar]
- 27.Perneczky R, Guo LH, Kagerbauer SM, Werle L, Kurz A, Martin J, Alexopoulos P. Soluble amyloid precursor protein beta as blood-based biomarker of alzheimer’s disease. Transl Psychiatry. 2013;3:e227. doi: 10.1038/tp.2013.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tagawa K, Kunishita T, Maruyama K, Yoshikawa K, Kominami E, Tsuchiya T, Suzuki K, Tabira T, Sugita H, Ishiura S. Alzheimer’s disease amyloid beta-clipping enzyme (APP secretase): Identification, purification, and characterization of the enzyme. Biochem Biophys Res Commun. 1991;177:377–87. doi: 10.1016/0006-291x(91)91994-n. [DOI] [PubMed] [Google Scholar]
- 29.Hook G, Hook V, Kindy M. The cysteine protease inhibitor, E64d, reduces brain amyloid-beta and improves memory deficits in alzheimer’s disease animal models by inhibiting cathepsin B, but not BACE1, beta-secretase activity. J Alzheimers Dis. 2011;26:387–408. doi: 10.3233/JAD-2011-110101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cummings J. What can be inferred from the interruption of the semagacestat trial for treatment of alzheimer’s disease? Biol Psychiatry. 2010;68:876–8. doi: 10.1016/j.biopsych.2010.09.020. [DOI] [PubMed] [Google Scholar]
- 31.Basi GS, Hemphill S, Brigham EF, Liao A, Aubele DL, Baker J, Barbour R, Bova M, Chen XH, Dappen MS, Eichenbaum T, Goldbach E, Hawkinson J, Lawler-Herbold R, Hu K, Hui T, Jagodzinski JJ, Keim PS, Kholodenko D, Latimer LH, Lee M, Marugg J, Mattson MN, McCauley S, Miller JL, Motter R, Mutter L, Neitzel ML, Ni H, Nguyen L, Quinn K, Ruslim L, Semko CM, Shapiro P, Smith J, Soriano F, Szoke B, Tanaka K, Tang P, Tucker JA, Ye XM, Yu M, Wu J, Xu YZ, Garofalo AW, Sauer JM, Konradi AW, Ness D, Shopp G, Pleiss MA, Freedman SB, Schenk D. Amyloid precursor protein selective gamma-secretase inhibitors for treatment of alzheimer’s disease. Alzheimers Res Ther. 2010;2:36. doi: 10.1186/alzrt60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Imbimbo BP, Giardino L, Sivilia S, Giuliani A, Gusciglio M, Pietrini V, Del Giudice E, D’Arrigo A, Leon A, Villetti G, Calza L. CHF5074, a novel gamma-secretase modulator, restores hippocampal neurogenesis potential and reverses contextual memory deficit in a transgenic mouse model of alzheimer’s disease. J Alzheimers Dis. 2010;20:159–73. doi: 10.3233/JAD-2010-1366. [DOI] [PubMed] [Google Scholar]
- 33.Anderson JJ, Holtz G, Baskin PP, Turner M, Rowe B, Wang B, Kounnas MZ, Lamb BT, Barten D, Felsenstein K, McDonald I, Srinivasan K, Munoz B, Wagner SL. Reductions in beta-amyloid concentrations in vivo by the gamma-secretase inhibitors BMS-289948 and BMS-299897. Biochem Pharmacol. 2005;69:689–98. doi: 10.1016/j.bcp.2004.11.015. [DOI] [PubMed] [Google Scholar]
- 34.El Mouedden M, Vandermeeren M, Meert T, Mercken M. Reduction of abeta levels in the sprague dawley rat after oral administration of the functional gamma-secretase inhibitor, DAPT: A novel non-transgenic model for abeta production inhibitors. Curr Pharm Des. 2006;12:671–6. doi: 10.2174/138161206775474233. [DOI] [PubMed] [Google Scholar]
- 35.Hawkins J, Harrison DC, Ahmed S, Davis RP, Chapman T, Marshall I, Smith B, Mead TL, Medhurst A, Giblin GM, Hall A, Gonzalez MI, Richardson J, Hussain I. Dynamics of Abeta42 reduction in plasma, CSF and brain of rats treated with the gamma-secretase modulator, GSM-10h. Neurodegener Dis. 2011;8:455–64. doi: 10.1159/000324511. [DOI] [PubMed] [Google Scholar]
- 36.Cook JJ, Wildsmith KR, Gilberto DB, Holahan MA, Kinney GG, Mathers PD, Michener MS, Price EA, Shearman MS, Simon AJ, Wang JX, Wu G, Yarasheski KE, Bateman RJ. Acute gamma-secretase inhibition of nonhuman primate CNS shifts amyloid precursor protein (APP) metabolism from amyloid-beta production to alternative APP fragments without amyloid-beta rebound. J Neurosci. 2010;30:6743–50. doi: 10.1523/JNEUROSCI.1381-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Portelius E, Van Broeck B, Andreasson U, Gustavsson MK, Mercken M, Zetterberg H, Borghys H, Blennow K. Acute effect on the abeta isoform pattern in CSF in response to gamma-secretase modulator and inhibitor treatment in dogs. J Alzheimers Dis. 2010;21:1005–12. doi: 10.3233/JAD-2010-100573. [DOI] [PubMed] [Google Scholar]
- 38.Kim HD, Tahara K, Maxwell JA, Lalonde R, Fukuiwa T, Fujihashi K, Van Kampen KR, Kong FK, Tang DC, Fukuchi K. Nasal inoculation of an adenovirus vector encoding 11 tandem repeats of Abeta1-6 upregulates IL-10 expression and reduces amyloid load in a mo/hu APPswe PS1dE9 mouse model of alzheimer’s disease. J Gene Med. 2007;9:88–98. doi: 10.1002/jgm.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Town T, Vendrame M, Patel A, Poetter D, DelleDonne A, Mori T, Smeed R, Crawford F, Klein T, Tan J, Mullan M. Reduced Th1 and enhanced Th2 immunity after immunization with alzheimer’s beta-amyloid(1-42) J Neuroimmunol. 2002;132:49–59. doi: 10.1016/s0165-5728(02)00307-7. [DOI] [PubMed] [Google Scholar]
- 40.Cao C, Arendash GW, Dickson A, Mamcarz MB, Lin X, Ethell DW. Abeta-specific Th2 cells provide cognitive and pathological benefits to alzheimer’s mice without infiltrating the CNS. Neurobiol Dis. 2009;34:63–70. doi: 10.1016/j.nbd.2008.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sanchez-Ramos J, Song S, Sava V, Catlow B, Lin X, Mori T, Cao C, Arendash GW. Granulocyte colony stimulating factor decreases brain amyloid burden and reverses cognitive impairment in alzheimer’s mice. Neuroscience. 2009;163:55–72. doi: 10.1016/j.neuroscience.2009.05.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pratico D. The neurobiology of isoprostanes and alzheimer’s disease. Biochim Biophys Acta. 2010;1801:930–3. doi: 10.1016/j.bbalip.2010.01.009. [DOI] [PubMed] [Google Scholar]
- 43.Pratico D, Uryu K, Leight S, Trojanoswki JQ, Lee VM. Increased lipid peroxidation precedes amyloid plaque formation in an animal model of alzheimer amyloidosis. J Neurosci. 2001;21:4183–7. doi: 10.1523/JNEUROSCI.21-12-04183.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, Culliford D, Perry VH. Systemic inflammation and disease progression in alzheimer disease. Neurology. 2009;73:768–74. doi: 10.1212/WNL.0b013e3181b6bb95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tokita Y, Kaji K, Lu J, Okura Y, Kohyama K, Matsumoto Y. Assessment of non-viral amyloid-beta DNA vaccines on amyloid-beta reduction and safety in rhesus monkeys. J Alzheimers Dis. 2010;22:1351–61. doi: 10.3233/JAD-2010-100978. [DOI] [PubMed] [Google Scholar]
- 46.Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, Finnerty CC, Lopez CM, Honari S, Moore EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein BH, Miller-Graziano C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF, Maier RV, Moldawer LL, Herndon DN, Davis RW, Xiao W, Tompkins RG. Inflammation and Host Response to Injury, Large Scale Collaborative Research Program. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2013;110:3507–12. doi: 10.1073/pnas.1222878110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Barten DM, Cadelina GW, Hoque N, DeCarr LB, Guss VL, Yang L, Sankaranarayanan S, Wes PD, Flynn ME, Meredith JE, Ahlijanian MK, Albright CF. Tau transgenic mice as models for cerebrospinal fluid tau biomarkers. J Alzheimers Dis. 2011;24(Suppl 2):127–41. doi: 10.3233/JAD-2011-110161. [DOI] [PubMed] [Google Scholar]
- 48.Zilka N, Korenova M, Kovacech B, Iqbal K, Novak M. CSF phospho-tau correlates with behavioural decline and brain insoluble phospho-tau levels in a rat model of tauopathy. Acta Neuropathol. 2010;119:679–87. doi: 10.1007/s00401-010-0680-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J, Hansson O. Plasma tau levels in alzheimer’s disease. Alzheimers Res Ther. 2013;5:9. doi: 10.1186/alzrt163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Reiman EM, Uecker A, Gonzalez-Lima F, Minear D, Chen K, Callaway NL, Berndt JD, Games D. Tracking alzheimer’s disease in transgenic mice using fluorodeoxyglucose autoradiography. Neuroreport. 2000;11:987–91. doi: 10.1097/00001756-200004070-00018. [DOI] [PubMed] [Google Scholar]
- 51.Valla J, Chen K, Berndt JD, Gonzalez-Lima F, Cherry SR, Games D, Reiman EM. Effects of image resolution on autoradiographic measurements of posterior cingulate activity in PDAPP mice: Implications for functional brain imaging studies of transgenic mouse models of alzheimer’s disease. Neuroimage. 2002;16:1–6. doi: 10.1006/nimg.2002.1080. [DOI] [PubMed] [Google Scholar]
- 52.Valla J, Schneider LE, Gonzalez-Lima F, Reiman EM. Nonprogressive transgene-related callosal and hippocampal changes in PDAPP mice. Neuroreport. 2006;17:829–32. doi: 10.1097/01.wnr.0000220140.91294.15. [DOI] [PubMed] [Google Scholar]
- 53.Dubois A, Herard AS, Delatour B, Hantraye P, Bonvento G, Dhenain M, Delzescaux T. Detection by voxel-wise statistical analysis of significant changes in regional cerebral glucose uptake in an APP/PS1 transgenic mouse model of alzheimer’s disease. Neuroimage. 2010;51:586–98. doi: 10.1016/j.neuroimage.2010.02.074. [DOI] [PubMed] [Google Scholar]
- 54.Nicholson RM, Kusne Y, Nowak LA, LaFerla FM, Reiman EM, Valla J. Regional cerebral glucose uptake in the 3xTG model of alzheimer’s disease highlights common regional vulnerability across AD mouse models. Brain Res. 2010;1347:179–85. doi: 10.1016/j.brainres.2010.05.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Platt B, Drever B, Koss D, Stoppelkamp S, Jyoti A, Plano A, Utan A, Merrick G, Ryan D, Melis V, Wan H, Mingarelli M, Porcu E, Scrocchi L, Welch A, Riedel G. Abnormal cognition, sleep, EEG and brain metabolism in a novel knock-in alzheimer mouse, PLB1. PLoS One. 2011;6:e27068. doi: 10.1371/journal.pone.0027068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kuntner C, Kesner AL, Bauer M, Kremslehner R, Wanek T, Mandler M, Karch R, Stanek J, Wolf T, Muller M, Langer O. Limitations of small animal PET imaging with [18F] FDDNP and FDG for quantitative studies in a transgenic mouse model of alzheimer’s disease. Mol Imaging Biol. 2009;11:236–40. doi: 10.1007/s11307-009-0198-z. [DOI] [PubMed] [Google Scholar]
- 57.Luo F, Rustay NR, Ebert U, Hradil VP, Cole TB, Llano DA, Mudd SR, Zhang Y, Fox GB, Day M. Characterization of 7- and 19-month-old Tg2576 mice using multimodal in vivo imaging: Limitations as a translatable model of alzheimer’s disease. Neurobiol Aging. 2012;33:933–44. doi: 10.1016/j.neurobiolaging.2010.08.005. [DOI] [PubMed] [Google Scholar]
- 58.Klunk WE, Lopresti BJ, Ikonomovic MD, Lefterov IM, Koldamova RP, Abrahamson EE, Debnath ML, Holt DP, Huang GF, Shao L, DeKosky ST, Price JC, Mathis CA. Binding of the positron emission tomography tracer pittsburgh compound-B reflects the amount of amyloid-beta in alzheimer’s disease brain but not in transgenic mouse brain. J Neurosci. 2005;25:10598–606. doi: 10.1523/JNEUROSCI.2990-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Toyama H, Ye D, Ichise M, Liow JS, Cai L, Jacobowitz D, Musachio JL, Hong J, Crescenzo M, Tipre D, Lu JQ, Zoghbi S, Vines DC, Seidel J, Katada K, Green MV, Pike VW, Cohen RM, Innis RB. PET imaging of brain with the beta-amyloid probe, [11C] 6-OH-BTA-1, in a transgenic mouse model of alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2005;32:593–600. doi: 10.1007/s00259-005-1780-5. [DOI] [PubMed] [Google Scholar]
- 60.Manook A, Yousefi BH, Willuweit A, Platzer S, Reder S, Voss A, Huisman M, Settles M, Neff F, Velden J, Schoor M, von der Kammer H, Wester HJ, Schwaiger M, Henriksen G, Drzezga A. Small-animal PET imaging of amyloid-beta plaques with [11C] PiB and its multi-modal validation in an APP/PS1 mouse model of alzheimer’s disease. PLoS One. 2012;7:e31310. doi: 10.1371/journal.pone.0031310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Higuchi M. Visualization of brain amyloid and microglial activation in mouse models of alzheimer’s disease. Curr Alzheimer Res. 2009;6:137–43. doi: 10.2174/156720509787602906. [DOI] [PubMed] [Google Scholar]
- 62.Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T, Staufenbiel M, Iwata N, Ono M, Saido TC, Suzuki K, Mori H, Higuchi M, Suhara T. Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of alzheimer’s disease enabled by positron emission tomography. J Neurosci. 2007;27:10957–68. doi: 10.1523/JNEUROSCI.0673-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.von Reutern B, Grunecker B, Yousefi BH, Henriksen G, Czisch M, Drzezga A. Voxel-based analysis of amyloid-burden measured with [11C] PiB PET in a double transgenic mouse model of alzheimer’s disease. Mol Imaging Biol. 2013 Apr 10; doi: 10.1007/s11307-013-0625-z. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 64.Noda A, Murakami Y, Nishiyama S, Fukumoto D, Miyoshi S, Tsukada H, Nishimura S. Amyloid imaging in aged and young macaques with [11C] PIB and [18F] FDDNP. Synapse. 2008;62:472–5. doi: 10.1002/syn.20508. [DOI] [PubMed] [Google Scholar]
- 65.Teng E, Kepe V, Frautschy SA, Liu J, Satyamurthy N, Yang F, Chen PP, Cole GB, Jones MR, Huang SC, Flood DG, Trusko SP, Small GW, Cole GM, Barrio JR. F-18] FDDNP microPET imaging correlates with brain abeta burden in a transgenic rat model of alzheimer disease: Effects of aging, in vivo blockade, and anti-abeta antibody treatment. Neurobiol Dis. 2011;43:565–75. doi: 10.1016/j.nbd.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fodero-Tavoletti MT, Okamura N, Furumoto S, Mulligan RS, Connor AR, McLean CA, Cao D, Rigopoulos A, Cartwright GA, O’Keefe G, Gong S, Adlard PA, Barnham KJ, Rowe CC, Masters CL, Kudo Y, Cappai R, Yanai K, Villemagne VL. 18F-THK523: A novel in vivo tau imaging ligand for alzheimer’s disease. Brain. 2011;134:1089–100. doi: 10.1093/brain/awr038. [DOI] [PubMed] [Google Scholar]
- 67.Choi SR, Golding G, Zhuang Z, Zhang W, Lim N, Hefti F, Benedum TE, Kilbourn MR, Skovronsky D, Kung HF. Preclinical properties of 18F-AV-45: A PET agent for abeta plaques in the brain. J Nucl Med. 2009;50:1887–94. doi: 10.2967/jnumed.109.065284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Poisnel G, Dhilly M, Moustie O, Delamare J, Abbas A, Guilloteau D, Barre L. PET imaging with [18F] AV-45 in an APP/PS1-21 murine model of amyloid plaque deposition. Neurobiol Aging. 2012 Nov;33:2561–71. doi: 10.1016/j.neurobiolaging.2011.12.024. [DOI] [PubMed] [Google Scholar]
- 69.Lang S. The role of peripheral benzodiazepine receptors (PBRs) in CNS pathophysiology. Curr Med Chem. 2002;9:1411–5. doi: 10.2174/0929867023369745. [DOI] [PubMed] [Google Scholar]
- 70.Venneti S, Lopresti BJ, Wang G, Hamilton RL, Mathis CA, Klunk WE, Apte UM, Wiley CA. PK11195 labels activated microglia in alzheimer’s disease and in vivo in a mouse model using PET. Neurobiol Aging. 2009;30:1217–26. doi: 10.1016/j.neurobiolaging.2007.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Maeda J, Zhang MR, Okauchi T, Ji B, Ono M, Hattori S, Kumata K, Iwata N, Saido TC, Trojanowski JQ, Lee VM, Staufenbiel M, Tomiyama T, Mori H, Fukumura T, Suhara T, Higuchi M. In vivo positron emission tomographic imaging of glial responses to amyloid-beta and tau pathologies in mouse models of alzheimer’s disease and related disorders. J Neurosci. 2011;31:4720–30. doi: 10.1523/JNEUROSCI.3076-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lau JC, Lerch JP, Sled JG, Henkelman RM, Evans AC, Bedell BJ. Longitudinal neuroanatomical changes determined by deformation-based morphometry in a mouse model of alzheimer’s disease. Neuroimage. 2008;42:19–27. doi: 10.1016/j.neuroimage.2008.04.252. [DOI] [PubMed] [Google Scholar]
- 73.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;100:1381–6. doi: 10.1073/pnas.242746599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sykova E, Vorisek I, Antonova T, Mazel T, Meyer-Luehmann M, Jucker M, Hajek M, Ort 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;102:479–84. doi: 10.1073/pnas.0408235102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Van Broeck B, Vanhoutte G, Pirici D, Van Dam D, Wils H, Cuijt I, Vennekens K, Zabielski M, Michalik A, Theuns J, De Deyn PP, Van der Linden A, Van Broeckhoven C, Kumar-Singh S. Intraneuronal amyloid beta and reduced brain volume in a novel APP T714I mouse model for alzheimer’s disease. Neurobiol Aging. 2008;29:241–52. doi: 10.1016/j.neurobiolaging.2006.10.016. [DOI] [PubMed] [Google Scholar]
- 76.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;11:425–33. doi: 10.1006/nbdi.2002.0555. [DOI] [PubMed] [Google Scholar]
- 77.Yang D, Xie Z, Stephenson D, Morton D, Hicks CD, Brown TM, Sriram R, O’Neill S, Raunig D, Bocan T. Volumetric MRI and MRS provide sensitive measures of alzheimer’s disease neuropathology in inducible tau transgenic mice (rTg4510) Neuroimage. 2011;54:2652–8. doi: 10.1016/j.neuroimage.2010.10.067. [DOI] [PubMed] [Google Scholar]
- 78.Ramesh BN, Raichurkar KP, Shamasundar NM, Rao TS, Rao KS. Abeta(42) induced MRI changes in aged rabbit brain resembles AD brain. Neurochem Int. 2011;59:637–42. doi: 10.1016/j.neuint.2011.06.003. [DOI] [PubMed] [Google Scholar]
- 79.Heo JH, Lee SR, Lee ST, Lee KM, Oh JH, Jang DP, Chang KT, Cho ZH. Spatial distribution of glucose hypometabolism induced by intracerebroventricular streptozotocin in monkeys. J Alzheimers Dis. 2011;25:517–23. doi: 10.3233/JAD-2011-102079. [DOI] [PubMed] [Google Scholar]
- 80.Alsop DC, Detre JA, Grossman M. Assessment of cerebral blood flow in alzheimer’s disease by spin-labeled magnetic resonance imaging. Ann Neurol. 2000;47:93–100. [PubMed] [Google Scholar]
- 81.Chen Y, Wolk DA, Reddin JS, Korczykowski M, Martinez PM, Musiek ES, Newberg AB, Julin P, Arnold SE, Greenberg JH, Detre JA. Voxel-level comparison of arterial spin-labeled perfusion MRI and FDG-PET in alzheimer disease. Neurology. 2011;77:1977–85. doi: 10.1212/WNL.0b013e31823a0ef7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Johnson NA, Jahng GH, Weiner MW, Miller BL, Chui HC, Jagust WJ, Gorno-Tempini ML, Schuff N. Pattern of cerebral hypoperfusion in alzheimer disease and mild cognitive impairment measured with arterial spin-labeling MR imaging: Initial experience. Radiology. 2005;234:851–9. doi: 10.1148/radiol.2343040197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.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;32:407–18. doi: 10.1016/j.neurobiolaging.2009.03.009. [DOI] [PubMed] [Google Scholar]
- 84.Weidensteiner C, Metzger F, Bruns A, Bohrmann B, Kuennecke B, von Kienlin M. Cortical hypoperfusion in the B6. PS2APP mouse model for alzheimer’s disease: Comprehensive phenotyping of vascular and tissular parameters by MRI. Magn Reson Med. 2009;62:35–45. doi: 10.1002/mrm.21985. [DOI] [PubMed] [Google Scholar]
- 85.Kandimalla KK, Wengenack TM, Curran GL, Gilles EJ, Poduslo JF. Pharmacokinetics and amyloid plaque targeting ability of a novel peptide-based magnetic resonance contrast agent in wild-type and alzheimer’s disease transgenic mice. J Pharmacol Exp Ther. 2007;322:541–9. doi: 10.1124/jpet.107.119883. [DOI] [PubMed] [Google Scholar]
- 86.Sigurdsson EM, Wadghiri YZ, Mosconi L, Blind JA, Knudsen E, Asuni A, Scholtzova H, Tsui WH, Li Y, Sadowski M, Turnbull DH, de Leon MJ, Wisniewski T. A non-toxic ligand for voxel-based MRI analysis of plaques in AD transgenic mice. Neurobiol Aging. 2008;29:836–47. doi: 10.1016/j.neurobiolaging.2006.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wengenack TM, Jack CR Jr, Garwood M, Poduslo JF. MR microimaging of amyloid plaques in alzheimer’s disease transgenic mice. Eur J Nucl Med Mol Imaging. 2008;35(Suppl 1):S82–8. doi: 10.1007/s00259-007-0706-9. [DOI] [PubMed] [Google Scholar]
- 88.Poduslo JF, Wengenack TM, Curran GL, Wisniewski T, Sigurdsson EM, Macura SI, Borowski BJ, Jack CR Jr. Molecular targeting of alzheimer’s amyloid plaques for contrast-enhanced magnetic resonance imaging. Neurobiol Dis. 2002;11:315–29. doi: 10.1006/nbdi.2002.0550. [DOI] [PubMed] [Google Scholar]
- 89.Wadghiri YZ, Sigurdsson EM, Sadowski M, Elliott JI, Li Y, Scholtzova H, Tang CY, Aguinaldo G, Pappolla M, Duff K, Wisniewski T, Turnbull DH. Detection of alzheimer’s amyloid in transgenic mice using magnetic resonance microimaging. Magn Reson Med. 2003;50:293–302. doi: 10.1002/mrm.10529. [DOI] [PubMed] [Google Scholar]
- 90.Higuchi M, Iwata N, Matsuba Y, Sato K, Sasamoto K, Saido TC. 19F and 1H MRI detection of amyloid beta plaques in vivo. Nat Neurosci. 2005;8:527–33. doi: 10.1038/nn1422. [DOI] [PubMed] [Google Scholar]
- 91.Yanagisawa D, Amatsubo T, Morikawa S, Taguchi H, Urushitani M, Shirai N, Hirao K, Shiino A, Inubushi T, Tooyama I. In vivo detection of amyloid beta deposition using (1)(9)F magnetic resonance imaging with a (1)(9)F-containing curcumin derivative in a mouse model of alzheimer’s disease. Neuroscience. 2011;184:120–7. doi: 10.1016/j.neuroscience.2011.03.071. [DOI] [PubMed] [Google Scholar]
- 92.Koffie RM, Farrar CT, Saidi LJ, William CM, Hyman BT, Spires-Jones TL. Nanoparticles enhance brain delivery of blood-brain barrier-impermeable probes for in vivo optical and magnetic resonance imaging. Proc Natl Acad Sci U S A. 2011;108:18837–42. doi: 10.1073/pnas.1111405108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Poduslo JF, Hultman KL, Curran GL, Preboske GM, Chamberlain R, Marjanska M, Garwood M, Jack CR Jr, Wengenack TM. Targeting vascular amyloid in arterioles of alzheimer disease transgenic mice with amyloid beta protein antibody-coated nanoparticles. J Neuropathol Exp Neurol. 2011;70:653–61. doi: 10.1097/NEN.0b013e318225038c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yang J, Wadghiri YZ, Hoang DM, Tsui W, Sun Y, Chung E, Li Y, Wang A, de Leon M, Wisniewski T. Detection of amyloid plaques targeted by USPIO-Abeta1-42 in alzheimer’s disease transgenic mice using magnetic resonance microimaging. Neuroimage. 2011;55:1600–9. doi: 10.1016/j.neuroimage.2011.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Borthakur A, Gur T, Wheaton AJ, Corbo M, Trojanowski JQ, Lee VM, Reddy R. In vivo measurement of plaque burden in a mouse model of alzheimer’s disease. J Magn Reson Imaging. 2006;24:1011–7. doi: 10.1002/jmri.20751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jack CR Jr, Garwood M, Wengenack TM, Borowski B, Curran GL, Lin J, Adriany G, Grohn OH, Grimm R, Poduslo JF. In vivo visualization of alzheimer’s amyloid plaques by magnetic resonance imaging in transgenic mice without a contrast agent. Magn Reson Med. 2004;52:1263–71. doi: 10.1002/mrm.20266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jack CR Jr, Wengenack TM, Reyes DA, Garwood M, Curran GL, Borowski BJ, Lin J, Preboske GM, Holasek SS, Adriany G, Poduslo JF. In vivo magnetic resonance microimaging of individual amyloid plaques in alzheimer’s transgenic mice. J Neurosci. 2005;25:10041–8. doi: 10.1523/JNEUROSCI.2588-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lee SP, Falangola MF, Nixon RA, Duff K, Helpern JA. Visualization of beta-amyloid plaques in a transgenic mouse model of alzheimer’s disease using MR microscopy without contrast reagents. Magn Reson Med. 2004;52:538–44. doi: 10.1002/mrm.20196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Vanhoutte G, Dewachter I, Borghgraef P, Van Leuven F, Van der Linden A. Noninvasive in vivo MRI detection of neuritic plaques associated with iron in APP[V717I] transgenic mice, a model for alzheimer’s disease. Magn Reson Med. 2005;53:607–13. doi: 10.1002/mrm.20385. [DOI] [PubMed] [Google Scholar]
- 100.Dhenain M, Privat N, Duyckaerts C, Jacobs RE. Senile plaques do not induce susceptibility effects in T2*-weighted MR microscopic images. NMR Biomed. 2002;15:197–203. doi: 10.1002/nbm.760. [DOI] [PubMed] [Google Scholar]
- 101.Kim J, Choi IY, Michaelis ML, Lee P. Quantitative in vivo measurement of early axonal transport deficits in a triple transgenic mouse model of alzheimer’s disease using manganese-enhanced MRI. Neuroimage. 2011;56:1286–92. doi: 10.1016/j.neuroimage.2011.02.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Smith KD, Kallhoff V, Zheng H, Pautler RG. In vivo axonal transport rates decrease in a mouse model of alzheimer’s disease. Neuroimage. 2007;35:1401–8. doi: 10.1016/j.neuroimage.2007.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zerbi V, Kleinnijenhuis M, Fang X, Jansen D, Veltien A, Van Asten J, Timmer N, Dederen PJ, Kiliaan AJ, Heerschap A. Gray and white matter degeneration revealed by diffusion in an alzheimer mouse model. Neurobiol Aging. 2013;34:1440–50. doi: 10.1016/j.neurobiolaging.2012.11.017. [DOI] [PubMed] [Google Scholar]
- 104.Jessen F, Gur O, Block W, Ende G, Frolich L, Hammen T, Wiltfang J, Kucinski T, Jahn H, Heun R, Maier W, Kolsch H, Kornhuber J, Traber F. A multicenter (1)H-MRS study of the medial temporal lobe in AD and MCI. Neurology. 2009;72:1735–40. doi: 10.1212/WNL.0b013e3181a60a20. [DOI] [PubMed] [Google Scholar]
- 105.Frederick BD, Lyoo IK, Satlin A, Ahn KH, Kim MJ, Yurgelun-Todd DA, Cohen BM, Renshaw PF. In vivo proton magnetic resonance spectroscopy of the temporal lobe in alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:1313–22. doi: 10.1016/j.pnpbp.2004.08.013. [DOI] [PubMed] [Google Scholar]
- 106.Lee MR, Denic A, Hinton DJ, Mishra PK, Choi DS, Pirko I, Rodriguez M, Macura SI. Preclinical (1)H-MRS neurochemical profiling in neurological and psychiatric disorders. Bioanalysis. 2012;4:1787–804. doi: 10.4155/bio.12.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Dedeoglu A, Choi JK, Cormier K, Kowall NW, Jenkins BG. Magnetic resonance spectroscopic analysis of alzheimer’s disease mouse brain that express mutant human APP shows altered neurochemical profile. Brain Res. 2004;1012:60–5. doi: 10.1016/j.brainres.2004.02.079. [DOI] [PubMed] [Google Scholar]
- 108.Chen SQ, Wang PJ, Ten GJ, Zhan W, Li MH, Zang FC. Role of myo-inositol by magnetic resonance spectroscopy in early diagnosis of alzheimer’s disease in APP/PS1 transgenic mice. Dement Geriatr Cogn Disord. 2009;28:558–66. doi: 10.1159/000261646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Marjanska M, Curran GL, Wengenack TM, Henry PG, Bliss RL, Poduslo JF, Jack CR Jr, Ugurbil K, Garwood M. Monitoring disease progression in transgenic mouse models of alzheimer’s disease with proton magnetic resonance spectroscopy. Proc Natl Acad Sci U S A. 2005;102:11906–10. doi: 10.1073/pnas.0505513102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Oberg J, Spenger C, Wang FH, Andersson A, Westman E, Skoglund P, Sunnemark D, Norinder U, Klason T, Wahlund LO, Lindberg M. Age related changes in brain metabolites observed by 1H MRS in APP/PS1 mice. Neurobiol Aging. 2008;29:1423–33. doi: 10.1016/j.neurobiolaging.2007.03.002. [DOI] [PubMed] [Google Scholar]
- 111.Woo DC, Lee SH, Lee DW, Kim SY, Kim GY, Rhim HS, Choi CB, Kim HY, Lee CU, Choe BY. Regional metabolic alteration of alzheimer’s disease in mouse brain expressing mutant human APP-PS1 by 1H HR-MAS. Behav Brain Res. 2010;211:125–31. doi: 10.1016/j.bbr.2010.03.026. [DOI] [PubMed] [Google Scholar]
- 112.Xu W, Zhan Y, Huang W, Wang X, Zhang S, Lei H. Reduction of hippocampal N-acetyl aspartate level in aged APP(swe)/PS1(dE9) transgenic mice is associated with degeneration of CA3 pyramidal neurons. J Neurosci Res. 2010;88:3155–60. doi: 10.1002/jnr.22479. [DOI] [PubMed] [Google Scholar]
- 113.Moffett JR, Ross B, Arun P, Madhavarao CN, Namboodiri AM. N-acetylaspartate in the CNS: From neurodiagnostics to neurobiology. Prog Neurobiol. 2007;81:89–131. doi: 10.1016/j.pneurobio.2006.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Choi JK, Jenkins BG, Carreras I, Kaymakcalan S, Cormier K, Kowall NW, Dedeoglu A. Anti-inflammatory treatment in AD mice protects against neuronal pathology. Exp Neurol. 2010;223:377–84. doi: 10.1016/j.expneurol.2009.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Carrette O, Demalte I, Scherl A, Yalkinoglu O, Corthals G, Burkhard P, Hochstrasser DF, Sanchez JC. A panel of cerebrospinal fluid potential biomarkers for the diagnosis of alzheimer’s disease. Proteomics. 2003;3:1486–94. doi: 10.1002/pmic.200300470. [DOI] [PubMed] [Google Scholar]
- 116.Craig-Schapiro R, Kuhn M, Xiong C, Pickering EH, Liu J, Misko TP, Perrin RJ, Bales KR, Soares H, Fagan AM, Holtzman DM. Multiplexed immunoassay panel identifies novel CSF biomarkers for alzheimer’s disease diagnosis and prognosis. PLoS One. 2011;6:e18850. doi: 10.1371/journal.pone.0018850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Davidsson P, Westman-Brinkmalm A, Nilsson CL, Lindbjer M, Paulson L, Andreasen N, Sjogren M, Blennow K. Proteome analysis of cerebrospinal fluid proteins in alzheimer patients. Neuroreport. 2002;13:611–5. doi: 10.1097/00001756-200204160-00015. [DOI] [PubMed] [Google Scholar]
- 118.Finehout EJ, Franck Z, Choe LH, Relkin N, Lee KH. Cerebrospinal fluid proteomic biomarkers for alzheimer’s disease. Ann Neurol. 2007;61:120–9. doi: 10.1002/ana.21038. [DOI] [PubMed] [Google Scholar]
- 119.Puchades M, Hansson SF, Nilsson CL, Andreasen N, Blennow K, Davidsson P. Proteomic studies of potential cerebrospinal fluid protein markers for alzheimer’s disease. Brain Res Mol Brain Res. 2003;118:140–6. doi: 10.1016/j.molbrainres.2003.08.005. [DOI] [PubMed] [Google Scholar]
- 120.Simonsen AH, McGuire J, Podust VN, Davies H, Minthon L, Skoog I, Andreasen N, Wallin A, Waldemar G, Blennow K. Identification of a novel panel of cerebrospinal fluid biomarkers for alzheimer’s disease. Neurobiol Aging. 2008;29:961–8. doi: 10.1016/j.neurobiolaging.2007.01.011. [DOI] [PubMed] [Google Scholar]
- 121.Zhang J, Goodlett DR, Quinn JF, Peskind E, Kaye JA, Zhou Y, Pan C, Yi E, Eng J, Wang Q, Aebersold RH, Montine TJ. Quantitative proteomics of cerebrospinal fluid from patients with alzheimer disease. J Alzheimers Dis. 2005;7:125–33. doi: 10.3233/jad-2005-7205. discussion 173-80. [DOI] [PubMed] [Google Scholar]
- 122.Abdi F, Quinn JF, Jankovic J, McIntosh M, Leverenz JB, Peskind E, Nixon R, Nutt J, Chung K, Zabetian C, Samii A, Lin M, Hattan S, Pan C, Wang Y, Jin J, Zhu D, Li GJ, Liu Y, Waichunas D, Montine TJ, Zhang J. Detection of biomarkers with a multiplex quantitative proteomic platform in cerebrospinal fluid of patients with neurodegenerative disorders. J Alzheimers Dis. 2006;9:293–348. doi: 10.3233/jad-2006-9309. [DOI] [PubMed] [Google Scholar]
- 123.Simonsen AH, McGuire J, Podust VN, Hagnelius NO, Nilsson TK, Kapaki E, Vassilopoulos D, Waldemar G. A novel panel of cerebrospinal fluid biomarkers for the differential diagnosis of alzheimer’s disease versus normal aging and frontotemporal dementia. Dement Geriatr Cogn Disord. 2007;24:434–40. doi: 10.1159/000110576. [DOI] [PubMed] [Google Scholar]
- 124.German DC, Gurnani P, Nandi A, Garner HR, Fisher W, Diaz-Arrastia R, O’Suilleabhain P, Rosenblatt KP. Serum biomarkers for alzheimer’s disease: Proteomic discovery. Biomed Pharmacother. 2007;61:383–9. doi: 10.1016/j.biopha.2007.05.009. [DOI] [PubMed] [Google Scholar]
- 125.Hye A, Lynham S, Thambisetty M, Causevic M, Campbell J, Byers HL, Hooper C, Rijsdijk F, Tabrizi SJ, Banner S, Shaw CE, Foy C, Poppe M, Archer N, Hamilton G, Powell J, Brown RG, Sham P, Ward M, Lovestone S. Proteome-based plasma biomarkers for alzheimer’s disease. Brain. 2006;129:3042–50. doi: 10.1093/brain/awl279. [DOI] [PubMed] [Google Scholar]
- 126.Lopez MF, Mikulskis A, Kuzdzal S, Bennett DA, Kelly J, Golenko E, DiCesare J, Denoyer E, Patton WF, Ediger R, Sapp L, Ziegert T, Lynch C, Kramer S, Whiteley GR, Wall MR, Mannion DP, Della Cioppa G, Rakitan JS, Wolfe GM. High-resolution serum proteomic profiling of alzheimer disease samples reveals disease-specific, carrier-protein-bound mass signatures. Clin Chem. 2005;51:1946–54. doi: 10.1373/clinchem.2005.053090. [DOI] [PubMed] [Google Scholar]
- 127.Ray S, Britschgi M, Herbert C, Takeda-Uchimura Y, Boxer A, Blennow K, Friedman LF, Galasko DR, Jutel M, Karydas A, Kaye JA, Leszek J, Miller BL, Minthon L, Quinn JF, Rabinovici GD, Robinson WH, Sabbagh MN, So YT, Sparks DL, Tabaton M, Tinklenberg J, Yesavage JA, Tibshirani R, Wyss-Coray T. Classification and prediction of clinical alzheimer’s diagnosis based on plasma signaling proteins. Nat Med. 2007;13:1359–62. doi: 10.1038/nm1653. [DOI] [PubMed] [Google Scholar]
- 128.Thambisetty M, Hye A, Foy C, Daly E, Glover A, Cooper A, Simmons A, Murphy D, Lovestone S. Proteome-based identification of plasma proteins associated with hippocampal metabolism in early alzheimer’s disease. J Neurol. 2008;255:1712–20. doi: 10.1007/s00415-008-0006-8. [DOI] [PubMed] [Google Scholar]
- 129.Kim YH, Lee EK, Park SA, Kim NH, Kim CW. Proteomic analysis of plasma from a tau transgenic mouse. Int J Dev Neurosci. 2012;30:277–83. doi: 10.1016/j.ijdevneu.2012.01.011. [DOI] [PubMed] [Google Scholar]
- 130.Wang LL, Huang Y, Wang G, Chen SD. The potential role of microRNA-146 in alzheimer’s disease: Biomarker or therapeutic target? Med Hypotheses. 2012;78:398–401. doi: 10.1016/j.mehy.2011.11.019. [DOI] [PubMed] [Google Scholar]
- 131.Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang G, Huang Q, Rigoutsos I, Nelson PT. The expression of microRNA miR-107 decreases early in alzheimer’s disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28:1213–23. doi: 10.1523/JNEUROSCI.5065-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Geekiyanage H, Jicha GA, Nelson PT, Chan C. Blood serum miRNA: Non-invasive biomarkers for alzheimer’s disease. Exp Neurol. 2012;235:491–6. doi: 10.1016/j.expneurol.2011.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Englund H, Sehlin D, Johansson AS, Nilsson LN, Gellerfors P, Paulie S, Lannfelt L, Pettersson FE. Sensitive ELISA detection of amyloid-beta protofibrils in biological samples. J Neurochem. 2007;103:334–45. doi: 10.1111/j.1471-4159.2007.04759.x. [DOI] [PubMed] [Google Scholar]
- 134.Lord A, Englund H, Soderberg L, Tucker S, Clausen F, Hillered L, Gordon M, Morgan D, Lannfelt L, Pettersson FE, Nilsson LN. Amyloid-beta 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]
- 135.Sabbagh JJ, Kinney JW, Cummings JL. Alzheimer’s disease biomarkers: Correspondence between human studies and animal models. Neurobiol Dis. 2013 Aug;56:116–30. doi: 10.1016/j.nbd.2013.04.010. [DOI] [PubMed] [Google Scholar]