Chapter One - Relevance of transgenic mouse models for Alzheimer's disease

https://doi.org/10.1016/bs.pmbts.2020.07.007Get rights and content

Highlights

  • Alzheimer's disease (AD) is a uniquely human condition. However, transgenic mouse models that mimic specific aspects of AD pathology are necessary to evaluate hypotheses and molecular mechanism not accessible in humans.

  • Recognizing the complexity of AD is important if expectations from any form of research, whether it involves human subjects or mouse models, are to be managed.

  • A number of models of AD-relevant pathology have been developed, each with strengths and limitations that should be fully understood before use.

  • Transgenic mice are available that incorporate universal biological variables that drive AD risk and pathology (age, sex, APOE), and AD-related gene mutations (TREM2, ABCA7, PICALM) that affect a limited cohort of AD patients.

  • The frequently expressed sentiment “We have cured AD mice hundreds of times, but the cures never work in humans, so we need better models” is false. No mouse model fully replicates AD and no treatment has fully “cured” even the limited pathology of any mouse model.

  • The most relevant question for the use of a model should be: Is the choice of this rodent model appropriate for the question being addressed? While transgenic mice do not in fact have AD, they can tell us a great deal about specific aspects of AD disease mechanisms if the question is properly framed.

  • After selecting the model, studies must be planned and executed with an understanding of what information is provided by the readouts.

  • Current funding mechanisms and career development requirements are impediments to full hypothesis testing, either mechanistic or therapeutic.

  • Sensationalization of data, promulgation of dogma, and copycat science are hindering the optimal use of transgenic models of AD. Developing new models will not solve this problem.

  • Answers to fundamental questions about AD that remain elusive may be addressed using current models, or through careful consideration of what a new model should replicate.

  • As always, the best science comes from asking the right questions, not from following the latest trends in either technology or posited causality.

Abstract

Over the last several decades, a number of mouse models have been generated for mechanistic and preclinical therapeutic research on Alzheimer's disease (AD)-like behavioral impairments and pathology. Acceptance or rejection of these models by the scientific community is playing a prominent role in how research findings are viewed and whether grants get funded and manuscripts published. The question of whether models are useful has become an exceptionally contentious issue. Much time and effort have gone into investigators debating comments such as “there are no mouse models of AD,” “…nice work but needs to be tested in another mouse model,” or “only data from humans is valid.” This leads to extensive written justifications for the choice of a model in grant applications, to the point of almost apologizing for the use of models. These debates also lead to initiatives to create new, better models of AD without consideration of what “better” may mean in this context. On the “other side,” an argument supporting the use of mouse models is one cannot dissect a biological mechanism in postmortem human tissue. In this chapter, we examine issues that we believe must be addressed if in vivo AD research is to progress. We opine that it is not the models that are the issue, but rather a lack of understanding the aspects of AD-like pathology the models were designed to mimic. The goal here is to improve the utilization of models to address critical issues, not to offer a critique of existing models or make endorsements.

Introduction

There are diseases that are uniquely human, since no orthologous disease has been identified in other species. Many adult-onset neurodegenerative diseases fall into this category, including Alzheimer's disease (AD). A corollary of this absence of equivalent diseases is that there are few, if any, effective treatments for this category of disease. Recent history suggests that we need to understand the molecular basis of pathogenesis in these diseases if we are to develop effective therapies. However, many kinds of analysis cannot be done in human patients. Therefore, we are often left with descriptions of postmortem patient tissues that retain few indications of early stage disease and often are confounded by generic neuronal damage (e.g., mitochondrial and other organelle dysfunction, markers of reactive oxygen species stress, lower neuronal marker levels, changes in neuron structure) that is not disease specific and has not been useful in developing treatments.

Sequencing of the human genome and identification of mutations that produce AD in humans led to hope that a genetic solution might be found, but this approach has failed to produce clinically useful therapies. This may reflect (at least in part) the delayed onset of these diseases, which in turn indicates a phenotypic complexity that is not simply a result of gene expression but instead reflects interactions with the environment and posttranslational effects. However, identification of gene mutations that result in AD has led to the generation of animal models expressing mutant human proteins as models. Typically, these models produce pathological changes that partially replicate changes seen in human patients but fail to capture the full characteristics of the disease. Undeterred, a wide range of therapeutic approaches have been considered and many that showed limited success in animal models have been taken to clinical trials and failed. Investigators have reacted by blaming the animal models for the failure to develop therapies, leading to ever more complex models or complete dismissal of animal models as a means to better understand the disease process. This creates a dilemma—if animal models are dismissed then how do we model AD? The time is right to consider the use of models in the study of AD and other adult-onset neurodegenerative diseases. We propose that animal models should not be dismissed but recognized for their strengths in modeling specific aspects of AD-relevant pathology.

Section snippets

AD is complex, therefore realistic research expectations are important

The first challenge is to define pathology and pathogenesis in a disease as complicated as AD. Not only are there multiple pathogenic elements involved, but there are extensive co-morbidities leading to multiple types of dementia with overlapping pathology. Here we take a simplistic view of AD but one that still reflects the complexity of the disease and research challenges faced by investigators. AD is an adult-onset neurodegenerative disease preferentially affecting specific brain regions

Why do we need AD animal models?

The failure of multiple clinical trials that were based on approaches reported to be effective in mouse models have led some investigators to question the validity of mouse models. AD is rightly characterized as a uniquely human disease, so why do we need a mouse model of AD? Undoubtedly, analysis of human patients and tissue is a critical part of AD research. Analysis of patient materials defines the disease, characterizes pathological changes (mostly late stage changes) and identifies likely

Modeling risk factors

Extensive epidemiological and genetic studies have identified a number of risk factors that increase (or in some cases decrease) the likelihood of developing SAD. These factors are distinct from the FAD mutations in APP and PS1/2. FAD mutations are typically autosomal dominant mutations with high penetrance. Essentially, individuals with one of these FAD mutations or with Down syndrome will develop AD pathology, although the age of onset may vary. In contrast, many patients with increased risk

Study designs for therapeutic interventions

In a room of researchers discussing AD models, one is likely to hear comments such as “we have cured AD mice hundreds of times, but they never work in humans, so we need better models.” As common as this sentiment may be, it fails to hold up under inspection. First, there are no mice with AD, so we can't cure AD in mice. Second, none of the available treatments have eliminated AD pathology, even in mice. At best, reductions in amyloid or tangle burden have been achieved with a modest

Do scientists and the press need to sensationalize data?

Another challenge facing the field is the question of how to present results of studies that identify potential treatments. The understandable desperate need to find causes and treatments for AD has often resulted in sensationalization of scientific findings. How often have we read articles where the complexity of science is distilled down to “scientists find that x causes AD” or “miracle drug cures AD in mice.” The intensity of overselling becomes even more extreme in press releases from the

Acknowledgments

LMT is supported by NIH/NIA R01AG061114, R21AG061715 and R21AG053876, and the College of Medicine at the University of Illinois at Chicago institutional start-up funds. JMW is supported by NIH/NIA R01 AG057008-03S1. MJL is supported by NIH/NIA R01 AG056472, R01 AG057008, UH2/3 NS10012, R56 AG058655, 1R44 AG060826, RF1 AG058068, Institutional funds from the College of Medicine at the University of Illinois at Chicago, and anonymous philanthropic contributions. STB is supported by NIH/NINDS

References (54)

  • F. Grueninger et al.

    Phosphorylation of tau at S422 is enhanced by Abeta in TauPS2APP triple transgenic mice

    Neurobiol Dis

    (2010)
  • J.L. Wittnam et al.

    Pyroglutamate amyloid beta (Abeta) aggravates behavioral deficits in transgenic amyloid mouse model for Alzheimer disease

    J Biol Chem

    (2012)
  • A. Lord et al.

    The Arctic Alzheimer mutation facilitates early intraneuronal Abeta aggregation and senile plaque formation in transgenic mice

    Neurobiol Aging

    (2006)
  • M.A. Chishti et al.

    Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695

    J Biol Chem

    (2001)
  • J. Davis et al.

    Early-onset and robust cerebral microvascular accumulation of amyloid beta-protein in transgenic mice expressing low levels of a vasculotropic Dutch/Iowa mutant form of amyloid beta-protein precursor

    J Biol Chem

    (2004)
  • S. Oddo et al.

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

    Neuron

    (2003)
  • P.M. Sullivan et al.

    Targeted replacement of the mouse apolipoprotein E gene with the common human APOE3 allele enhances diet-induced hypercholesterolemia and atherosclerosis

    J Biol Chem

    (1997)
  • K.L. Youmans et al.

    APOE4-specific changes in Abeta accumulation in a new transgenic mouse model of Alzheimer disease

    J Biol Chem

    (2012)
  • S.J. Webster et al.

    Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models

    Front Genet

    (2014)
  • R. Roychaudhuri et al.

    Role of species-specific primary structure differences in Abeta42 assembly and neurotoxicity

    ACS Chem Nerosci

    (2015)
  • H. Oakley et al.

    Intraneuronal beta-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)
  • C. Sturchler-Pierrat et al.

    Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology

    Proc Natl Acad Sci U S A

    (1997)
  • K. Fukuchi et al.

    High levels of circulating beta-amyloid peptide do not cause cerebral beta-amyloidosis in transgenic mice

    Am J Pathol

    (1996)
  • T. Tomiyama et al.

    A mouse model of amyloid beta oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo

    J Neurosci

    (2010)
  • R. Radde et al.

    Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology

    EMBO Rep

    (2006)
  • J.G. Richards et al.

    PS2APP transgenic mice, coexpressing hPS2mut and hAPPswe, show age-related cognitive deficits associated with discrete brain amyloid deposition and inflammation

    J Neurosci

    (2003)
  • B.T. Lamb et al.

    Altered metabolism of familial Alzheimer's disease-linked amyloid precursor protein variants in yeast artificial chromosome transgenic mice

    Hum Mol Genet

    (1997)
  • Cited by (12)

    • Mechanisms of action by sulphur compounds in Allium sativum. A review

      2023, Pharmacological Research - Modern Chinese Medicine
    • Perspectives of ozone induced neuropathology and memory decline in Alzheimer's disease: A systematic review of preclinical evidences

      2022, Environmental Pollution
      Citation Excerpt :

      Expanding imaging and biomarkers in clinical practice allows physicians to apprehend heterogeneity in diagnosing and treating patients with dementia and mild cognitive impairment, enabling Tau PET radiotracers to add to clinical practice (Serrano-Pozo and Growdon, 2019). Various pathologies have developed other than plaques and tangles, including activated astrocytes and microglia and changes in cytokine profile (Tai et al., 2021). EOAD shows distinct non-memory symptoms and a shorter survival time.

    • Anthocyanin as a therapeutic in Alzheimer's disease: A systematic review of preclinical evidences

      2022, Ageing Research Reviews
      Citation Excerpt :

      It is also notable that cognitive decline is seen after decades of neurological alteration in the human brain. Moreover, transgenic models fail to limit the AD risk factor in mice (Tai et al., 2021). Furthermore, developing transgenic rodent models is time-consuming and uneconomical.

    • Remembering your A, B, C's: Alzheimer's disease and ABCA1

      2022, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      FAD-Tg mice are generally studied as young adults (at 3–12 months of age) and do not reproduce NFT pathology, nor do the FAD-Tg mice that also express mutations in the human gene responsible for tau protein expression (e.g., 3xTg mice) faithfully model human AD45–50. This inability to reproduce the complexity of human AD in mouse models has been discussed extensively in other recent reviews51,52. The heavy focus on amyloid mutations in these models naturally biased AD drug development toward agents (e.g., aducanumab and several others) that could reverse the development of amyloid pathology in these mice, which, as discussed above, have not shown beneficial clinical effects.

    • New Insights into Alzheimer's Drug Discovery Targeting NF-кB

      2024, Biointerface Research in Applied Chemistry
    View all citing articles on Scopus
    View full text