Elsevier

Progress in Neurobiology

Volume 95, Issue 4, December 2011, Pages 547-556
Progress in Neurobiology

Development of Alzheimer-disease neuroimaging-biomarkers using mouse models with amyloid-precursor protein-transgene expression

https://doi.org/10.1016/j.pneurobio.2011.05.004Get rights and content

Abstract

There are important recent developments in Alzheimer's disease (AD) translational research, especially with respect to the imaging of amyloid pathology in vivo using MRI and PET technologies. Here we exploit the most widely used transgenic mouse models of amyloid pathology in order to relate the imaging findings to our knowledge about the histopathological phenotype of these models. The development of new diagnostic criteria of AD necessitates the use of biological markers to diagnose AD even in the absence of overt dementia or early symptomatic mild cognitive impairment. The validity of the diagnosis will depend on the availability of an in vivo marker to reflect underlying neurobiological changes of AD. Transgenic models with essential features of AD pathology and mechanisms provide a test setting for the development and evaluation of new biological imaging markers.

Among the best established imaging markers of amyloid pathology in transgenic animals are high-field MRI of brain atrophy, proton spectroscopy of neurochemical changes, high-field MRI of amyloid plaque load, and in vivo plaque imaging using radio-labelled ligands with PET. We discuss the implications of the findings as well as the methodological limitations and the specific requirements of these technologies. We furthermore outline future directions of transgene-imaging research. Transgene imaging is an emerging area of translational research that implies strong multi- and interdisciplinary collaborations. It will become ever more valuable with the introduction of new diagnostic standards and novel treatment approaches which will require valid and reliable biological markers to improve the diagnosis and early treatment of AD patients.

Highlights

► Ultrahighfield MRI of transgenic models allows detection of amyloid pathology in vivo. ► Neurochemical profiles of amyloid pathology show discrepancies between human disease and animal models. ► PET and SPECT with radiolabelled compounds are the most promising translational markers of amyloid pathology in animal and human studies. ► Imaging features of transgenic models of amyloid pathology reveal the underlying substrate of signal changes in imaging studies in humans.

Introduction

Imaging-derived biomarkers are currently being implemented as a defining criterion for the diagnosis of AD according to currently discussed revised research criteria (Dubois et al., 2007, Dubois et al., 2009). The revision of the NINCDS-ADRDA criteria of AD (McKhann et al., 1984) suggests several new diagnostic entities (http://www.alz.org/research/diagnostic_criteria): the presence of positive biomarkers including imaging and CSF markers together with clinically probable AD defines the entity of “highly probable AD”; together with positive biomarkers the clinical syndrome of amnestic mild cognitive impairment (MCI) becomes “MCI of the Alzheimer type” or even “prodromal AD”. Moreover, a new entity of “preclinical AD” will solely be defined by CSF and imaging biomarkers in the absence of any cognitive decline. These new entities are intended to enable the early diagnosis in clinical diagnostic studies and the enrichment and stratification of samples for testing the effects of potential disease modification in clinical trials of AD (Hampel et al., 2010, Hampel et al., 2011).

With regard to current discussions about diagnostic criteria, the question remains, what the evidence might be that imaging markers of atrophy or neurochemical and functional abnormalities represent essential features of (preclinical) AD pathology? The validity of biomarkers regarding their supposed neurobiological substrate is crucial for the evaluation of disease-modifying treatment strategies. Several lines of research support the use of imaging biomarkers for the early diagnosis or the detection of intervention effects, including the clinical validity of a marker to predict a clinically relevant endpoint, such as cognitive decline (Teipel et al., 2008), the pathological validity of an atrophy marker to represent regional neuronal density (Bobinski et al., 2000) or the neurochemical validity of a marker to specifically bind to Aβ-containing amyloid plaques in the brain (Lockhart et al., 2007). One important line of evidence that is gaining increasing relevance for the validation of in vivo markers of AD are studies in transgenic animal models with amyloid pathology or other AD-related pathological features. Studies in transgenic animals serve a double function: Firstly, they provide independent evidence for potential pathological processes underlying specific imaging changes. Secondly, they represent a testing field for the development of new imaging technologies and sequences. Below we will give an overview of widely used transgenic models of amyloid pathology and discuss the findings on changes of cerebral anatomy, neurochemistry and neuronal function using in vivo imaging techniques in these models.

Section snippets

Transgene models of cerebral β-amyloidosis

The nosological models of AD keep shifting as a consequence of the lack of more precise knowledge about its etiology. This makes it difficult to design animal models precisely according to the human disease context and always requires an adaptation of mouse models to the changing concepts of human AD. Mutations in the amyloid precursor protein (APP) or one of the presenilins are sufficient to cause the complete AD phenotype of plaques, tangles, neuronal loss, and clinically progressive

In vivo imaging of transgenic models of AD

In vivo imaging techniques in transgenic models focus on (i) structural changes, (ii) biochemical changes, and (iii) in vivo amyloid-plaque load, employing technologies of high-field MRI, MR spectroscopy and PET. A summary of the methodological aspects is given in Table 2, while details are discussed in the following section.

Summary

A number of transgenic models replicate essential features of AD histopathology and molecular mechanisms. However, there is no single model that comprehensively resembles human AD pathophysiology. Most likely, there will never be a transgenic model which completely and fully replicates the converging mechanistic and pathological spectrum of sporadic AD, because the chronic decade-long time frame and progression of disease development as well as the complexity of the underlying neuronal

Acknowledgements

Part of this work was supported by grants of from the Interdisciplinary Faculty, Department “Aging Science and Humanities”, University of Rostock to S.J.T. and J.P., the Alzheimer Forschung Initiative e.V. and the Exzellenz Initiative Mecklenburg-Vorpommern to J.P., and of the Hirnliga e.V. (Nürmbrecht, Germany) to S.J.T. The work was further supported by the Science Foundation Ireland (SFI) investigator program award 08/IN.1/B1846 to H.H. Partly supported by the “Landesoffensive zur

References (104)

  • M.P. Kung et al.

    IMPY: an improved thioflavin-T derivative for in vivo labeling of beta-amyloid plaques

    Brain Res.

    (2002)
  • J.C. Lau et al.

    Longitudinal neuroanatomical changes determined by deformation-based morphometry in a mouse model of Alzheimer's disease

    Neuroimage

    (2008)
  • Y. Ma et al.

    A three-dimensional digital atlas database of the adult C57BL/6J mouse brain by magnetic resonance microscopy

    Neuroscience

    (2005)
  • J. Oberg et al.

    Age related changes in brain metabolites observed by 1H MRS in APP/PS1 mice

    Neurobiol. Aging

    (2008)
  • S. Oddo et al.

    Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer's disease

    Neurobiol. Aging

    (2003)
  • S. Oddo et al.

    Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction

    Neuron

    (2003)
  • J. Pahnke et al.

    Alzheimer's disease and blood-brain barrier function-Why have anti-beta-amyloid therapies failed to prevent dementia progression?

    Neurosci. Biobehav. Rev.

    (2009)
  • J. Penner et al.

    Increased glutamate in the hippocampus after galantamine treatment for Alzheimer disease

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2010)
  • J.F. Poduslo et al.

    Molecular targeting of Alzheimer's amyloid plaques for contrast-enhanced magnetic resonance imaging

    Neurobiol. Dis.

    (2002)
  • R.F. Rosen et al.

    PIB binding in aged primate brain: enrichment of high-affinity sites in humans with Alzheimer's disease

    Neurobiol. Aging

    (2011)
  • D.J. Rowland et al.

    Small-animal preclinical nuclear medicine instrumentation and methodology

    Semin. Nucl. Med.

    (2008)
  • S. Sharma et al.

    SPECT neuroimaging in translational research of CNS disorders

    Neurochem. Int.

    (2008)
  • M. Shidahara et al.

    Functional and structural synergy for resolution recovery and partial volume correction in brain PET

    Neuroimage

    (2009)
  • J. Valla et al.

    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)
  • S. Venneti et al.

    PK11195 labels activated microglia in Alzheimer's disease and in vivo in a mouse model using PET

    Neurobiol. Aging

    (2009)
  • M. von Kienlin et al.

    Altered metabolic profile in the frontal cortex of PS2APP transgenic mice, monitored throughout their life span

    Neurobiol. Dis.

    (2005)
  • T.M. Wengenack et al.

    Regional differences in MRI detection of amyloid plaques in AD transgenic mouse brain

    Neuroimage

    (2011)
  • F. Beekman et al.

    The pinhole: gateway to ultra-high-resolution three-dimensional radionuclide imaging

    Eur. J. Nucl. Med. Mol. Imaging

    (2007)
  • H. Benveniste et al.

    Detection of neuritic plaques in Alzheimer's disease by magnetic resonance microscopy

    Proc. Natl. Acad. Sci. U.S.A.

    (1999)
  • H. Benveniste et al.

    Anatomical and functional phenotyping of mice models of Alzheimer's disease by MR microscopy

    Ann. N.Y. Acad. Sci.

    (2007)
  • V.D. Bianca et al.

    Beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer's disease

    J. Biol. Chem.

    (1999)
  • Biomarkers_Definitions_Working_Group

    Biomarkers and surrogate endpoints: preferred definitions and conceptual framework

    Clin. Pharmacol. Ther.

    (2001)
  • M. Bobinski et al.

    The histological validation of post mortem magnetic resonance imaging-determined hippocampal volume in Alzheimer's disease

    Neuroscience

    (2000)
  • J. Borg et al.

    Differential MRI patterns of brain atrophy in double or single transgenic mice for APP and/or SOD

    J. Neurosci. Res.

    (2008)
  • N. Braakman et al.

    Longitudinal assessment of Alzheimer's beta-amyloid plaque development in transgenic mice monitored by in vivo magnetic resonance microimaging

    J. Magn. Reson. Imaging

    (2006)
  • W. Branderhorst et al.

    Targeted multi-pinhole SPECT

    Eur. J. Nucl. Med. Mol. Imaging.

    (2011)
  • L. Cai et al.

    Synthesis and evaluation of two 18F-labeled 6-iodo-2-(4′-N,N-dimethylamino)phenylimidazo[1,2-a]pyridine derivatives as prospective radioligands for beta-amyloid in Alzheimer's disease

    J. Med. Chem.

    (2004)
  • L. Cai et al.

    Synthesis and evaluation of N-methyl and S-methyl 11C-labeled 6-methylthio-2-(4′-N,N-dimethylamino)phenylimidazo[1,2-a]pyridines as radioligands for imaging beta-amyloid plaques in Alzheimer's disease

    J. Med. Chem.

    (2008)
  • A.P. Carpenter et al.

    The use of the exploratory IND in the evaluation and development of 18F-PET radiopharmaceuticals for amyloid imaging in the brain: a review of one company's experience

    Q. J. Nucl. Med. Mol. Imaging

    (2009)
  • I.H. Cheng et al.

    Aggressive amyloidosis in mice expressing human amyloid peptides with the Arctic mutation

    Nat. Med.

    (2004)
  • S.R. Choi et al.

    Preclinical properties of 18F-AV-45: a PET agent for Abeta plaques in the brain

    J. Nucl. Med.

    (2009)
  • B. Dubois et al.

    Early detection of Alzheimer's disease: new diagnostic criteria

    Dial. Clin. Neurosci.

    (2009)
  • C. Faber et al.

    Gradient-echo and CRAZED imaging for minute detection of Alzheimer plaques in an APPV717I × ADAM10-dn mouse model

    Magn. Reson. Med.

    (2007)
  • M.T. Fodero-Tavoletti et al.

    Bis(thiosemicarbazonato) Cu-64 complexes for positron emission tomography imaging of Alzheimer's disease

    J. Alzheimers Dis.

    (2010)
  • G.B. Fox et al.

    Translational neuroimaging of the CNS: novel pathways to drug development

    Mol. Interv.

    (2009)
  • D. Games et al.

    Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein

    Nature

    (1995)
  • H. Hampel et al.

    Biomarkers for Alzheimer's disease: academic, industry and regulatory perspectives

    Nat. Rev.

    (2010)
  • H. Hampel et al.

    Biomarkers for Alzheimer's disease therapeutic trials

    Prog. Neurobiol.

    (2011)
  • J. Hardy

    The shorter amyloid cascade hypothesis

    Neurobiol. Aging

    (1999)
  • J.A. Helpern et al.

    Quantitative MRI assessment of Alzheimer's disease

    J. Mol. Neurosci.

    (2004)
  • Cited by (27)

    • APPswe/PS1dE9 mice with cortical amyloid pathology show a reduced NAA/Cr ratio without apparent brain atrophy: A MRS and MRI study

      2017, NeuroImage: Clinical
      Citation Excerpt :

      Transgenic animal models of Aβ pathology provide mechanistic insight into some aspects of AD pathology related to Aβ accumulation, but transfer of findings to the AD phenotype in humans is limited (Foley et al., 2015). Using quantitative neuroimaging is a possible aid to improve translation of mechanistic findings in transgenic models to human end phenotypes of brain morphology or function (Teipel et al., 2011). Brain morphometry based on high resolution MRI is considered a proxy of neuronal loss in humans (Bobinski et al., 2000).

    • In vivo DTI tractography of the rat brain: An atlas of the main tracts in Paxinos space with histological comparison

      2015, Magnetic Resonance Imaging
      Citation Excerpt :

      In the last years magnetic resonance imaging (MRI) has been increasingly employed in preclinical studies on rodent models of neurological disorders to probe structural and functional changes throughout the course of disease. MRI inspection introduced additional potential to identify markers for the efficacy of therapeutic strategies [1–6]. Using high-field MRI scanners, it is possible to generate images of the rodent brain with high spatial resolution, giving the opportunity to create MRI-based anatomical atlases, complementary to histology-based atlases.

    • Perspective on future role of biological markers in clinical therapy trials of Alzheimer's disease: A long-range point of view beyond 2020

      2014, Biochemical Pharmacology
      Citation Excerpt :

      Based on mappings of subcortical nuclei from postmortem analyses, MRI scans in cranio will help to identify early changes in cholinergic and noradrenergic projecting nuclei in predementia and dementia stages of AD [175,176]. MRI-based detection of amyloid plaques has been successfully implemented in transgenic animals [177–180]. Further, a recent study indicates that detection is also possible in non-transgenic mouse lemur primates, in which plaques are formed naturally and are more similar to those found in humans [181].

    • Impact of partial volume effect correction on cerebral β-amyloid imaging in APP-Swe mice using [<sup>18</sup>F]-florbetaben PET

      2014, NeuroImage
      Citation Excerpt :

      In general animal models represent an important tool for preclinical testing of disease progression and novel medications in conjunction with molecular imaging (Virdee et al., 2012). A number of rodent AD-models are characterized by cerebral overexpression of ß-amyloid and progressive formation of cortical ß-plaques, emulating this aspect of AD pathology (Teipel et al., 2011). The prospect of monitoring disease progression in individual animals has given rise to a number of rodent positron emission tomography (PET) studies in conjunction with [11C]- or [18F]-labeled ß-amyloid-tracers (Klunk et al., 2005; Maeda et al., 2007; Manook et al., 2012; Poisnel et al., 2012; Rojas et al., 2013; Snellman et al., 2012; Toyama et al., 2005).

    • Risk and resilience: A new perspective on Alzheimer's disease

      2013, Geriatric Mental Health Care
      Citation Excerpt :

      Using measures of fractional anisotropy several studies showed significant decline of fiber tract integrity in corpus callosum, posterior cingulate and other intracortical projecting fiber tracts in Alzheimer's disease and in MCI in small scale single center studies (Fellgiebel et al., 2004; Chen et al., 2007; Teipel et al., 2007a, 2007b, 2007c; Friese et al., 2010; Douaud et al., 2011; Grana et al., 2011). Recently, multicenter DTI have become available within the framework of the European DTI Study in Dementia (EDSD) (Teipel et al., 2011a, 2011b, 2012). Diagnostic accuracy for discriminating Alzheimer's patients from healthy control subjects reached more than 80% accuracy using support vector machine based machine learning with the EDSD data representing 10 different MRI scanners (Dyrba et al., in press).

    View all citing articles on Scopus
    View full text