Skip to main content
Acta Neuropathologica Communications logoLink to Acta Neuropathologica Communications
. 2013 Sep 23;1:62. doi: 10.1186/2051-5960-1-62

The airbag problem–a potential culprit for bench-to-bedside translational efforts: relevance for Alzheimer’s disease

Dimitrije Krstic 1,✉,#, Irene Knuesel 1,#
PMCID: PMC3893418  PMID: 24252346

Abstract

For the last 20 years, the “amyloid cascade hypothesis” has dominated research aimed at understanding, preventing, and curing Alzheimer’s disease (AD). During that time researchers have acquired an enormous amount of data and have been successful, more than 300 times, in curing the disease in animal model systems by treatments aimed at clearing amyloid deposits. However, to date similar strategies have not been successful in human AD patients. Hence, before rushing into further clinical trials with compounds that aim at lowering amyloid-beta (Aβ) levels in increasingly younger people, it would be of highest priority to re-assess the initial assumption that accumulation of Aβ in the brain is the primary pathological event driving AD. Here we question this assumption by highlighting experimental evidence in support of the alternative hypothesis suggesting that APP and Aβ are part of a neuronal stress/injury system, which is up-regulated to counteract inflammation/oxidative stress-associated neurodegeneration that could be triggered by a brain injury, chronic infections, or a systemic disease. In AD, this protective program may be overridden by genetic and other risk factors, or its maintenance may become dysregulated during aging. Here, we provide a hypothetical example of a hypothesis-driven correlation between car accidents and airbag release in analogy to the evolution of the amyloid focus and as a way to offer a potential explanation for the failure of the AD field to translate the success of amyloid-related therapeutic strategies in experimental models to the clinic.

Keywords: Alzheimer’s disease AD, Amyloid Beta Aβ, Amyloid precursor protein APP, Reelin, Neuroinflammation, Neurodegeneration, Oxidative stress, Synaptic plasticity, Acute-phase reaction

Introduction

At the time of its wording in 1991/92, the “amyloid cascade hypothesis” [1] provided a logical explanation for the distinct amyloid-beta (Aβ) plaque pathology in patients diagnosed with Alzheimer’s Disease (AD). This hypothesis was supported by the direct link between AD and dominant mutations in either the amyloid precursor protein (APP) or enzymes that are involved in the production of Aβ peptides, such as presenilin 1 or 2 (PS1/2) [2-5]. Subsequently, numerous in vitro studies supported a toxic effect of Aβ peptides and by that “approved” its causative role in the disease etiology. However, mice that selectively overproduce Aβ peptides (BRI2-Aβ1-42) and develop plaque pathology display no signs of progressive cognitive impairments or neurodegeneration [6,7]. In addition, knock-in strategies to induce genetic mutations in APP or PS1 in rodents have been proven not to be sufficient to evoke AD-like phenotypes [8,9]. The development of certain features of the disease in animal models depends on transgene overexpression [10,11] and combinations of mutations [12-14]. Only very strong promoters and the presence of multiple mutations in mice, a combination never occuring in AD patients, triggers most of the neuropathological and behavioral phenotypes observed in humans, albeit in very young animals [15]. Nevertheless, transgenic-AD animals became state-of-the art tools in the community, and Aβ remained the proposed principle toxic and causative agent of the disease; first as monomer, then as an insoluble aggregate, followed by a soluble oligomer, and now a combination of all of these forms [16-18].

Review

Aβ–an acute-phase peptide

However, despite its proposed toxic/causative role in AD, physiological/protective roles for Aβ peptides have been described [19-21]. In addition to these neurotrophic effects of Aβ and its neurogenic effect on adult neural stem cells [22], Aβ peptides have been shown to stimulate synaptic function at physiological, picomolar, levels [23]. Increases in synaptic activity, through NMDA receptor activation, induce Aβ production [24], which at high, nanomolar, concentration potently depresses synaptic activity [23]. Hence, Aβ might serve as a suitable synaptic guardian against excitotoxicity. Furthermore, Aβ peptides capture metal ions such as Zn, Fe, and Cu, potentially reducing oxidative stress to neurons [25]. This is consistent with the notion that increased oxidative damage is an early feature in AD development [26-29] and with findings that subsequent increases in Aβ accumulation correlates with the decrease in oxidative stress [27,30]. Similarly, physiological levels of Aβ peptides in plasma and cerebrospinal fluid (CSF) are reported to protect circulating lipoproteins from oxidation [31].

It has been also demonstrated that Aβ possesses anti-inflammatory [32] and anti-microbial functions [33]. This potential immune function of Aβ is in line with studies showing induction of APP and Aβ production in brains of non-transgenic mice infected with C. pneumonia[34] or Herpes simplex virus type 1[35]. One should also keep in mind that although APP production in the brain is most abundant in neurons, astrocytes also produce APP and Aβ and this production is elevated by pro-inflammatory cytokines [36]. Interestingly, as has been noted in the brain, stimulation of the peripheral immune system induces expression of APP/Aβ in both T-lymphocytes [37] and CD14-positive mononuclear phagocytes [38]. Moreover, in humans correlations between neurospirochetosis and AD [39], and between periodontal infections and AD [40] are well established. In addition, young individuals exposed to elevated air pollution levels show neuroinflammatory responses with a significant induction of Aβ1-42 at autopsy [41].

Hence, in the context of Alzheimer’s disease progression, Aβ accumulation and plaque formation at sites of axonal swellings [42] and leakages [43,44] might be interpreted as the brain’s strategy to constrain such inflammation/oxidative stress-inducing hot-spots. This hypothesis is well in line with the observation that Aβ plaques form over 24 hours [45], possibly as an acute reaction to a burst or leakage of an axon as suggested in [44]. In parallel, increases in Aβ or other proteolytic fragments of APP at synaptic sites may act to protect vulnerable neurons from glutamate-mediated excitotoxicity [46].

APP–a guardian of axonal and synapse integrity

Important, away from the spot-light of Aβ-driven research, endogenous functions of APP are also starting to emerge [47]. Of specific interest are observations in APP knock-out mice, which show relatively subtle phenotypes [48] but high vulnerability to mechanical insults and low neuroregenerative capacity [49]. The lack of APP expression in these animals is associated with enhanced susceptibility to kainic acid-induced epilepsy [50] and elevated mortality following cerebral ischemia [51]. These findings are consistent with the idea that the observed elevation of APP expression following traumatic brain injury [52-55], entorhinal cortex lesion [56], induced ischemia [57-59], systemic infection with a bacterial or a viral mimic [60,61], or administration of pro-inflammatory cytokine interleukin 1 [62,63] is a protective neuronal response to stress/injury, rather than a pathological event leading to an overproduction of Aβ. This is also well in line with the notion that while Aβ1-42 levels increase, APP levels decrease with advancing AD pathology [64]. In further support of this idea, head injury in Drosophila induces an increase in expression of Drosophila APP-Like Protein (dAPLP), and mutant flies lacking dAPLP have significantly higher mortality rates after injury than wild-type flies [65], suggesting an evolutionarily conserved role of APP in response to injury/stress. In rodents, double knock-out of APP and APLP-1 induces severe structural and functional synaptic deficits [66], pointing to the crucial role of APP family members in synapse development and maintenance.

Interestingly, a recently discovered “protective” mutation in APP (APPA673T) [67] results not only a delay of the onset of AD, but also in better cognitive performance of AD patients that carry the mutation. Important, this mutation leads not only to decreases in production of Aβx-42, but also to a slight increase in levels of secreted APP ectodomain alpha (sAPPα) [67]. This is especially interesting as APP expression in aged animals is crucial for induction of long-term potentiation (LTP) and proper cognitive performance [68] and sAPPα was shown to reverse the deficits observed in APP knock-out mice [69].

In line with the hypothesis that impairments in axonal integrity and axonal transport represent a key contributing factor in the development of AD [44,70], APP has been shown to play a role in promoting kinesin-mediated fast axonal transport [71]. In addition, both a lack of APP [72] and an increase in proteolytic processing by γ-secretase [73] result in Aβ-independent [74] transport deficits. Together these findings suggest that stressful or injurious conditions in which APP endogenous functions are compromised, e.g. by a mutation in APP or its secretases, axonal transport deficits and subsequent synaptic loss may ensue. This view is supported by positron emission tomography (PET) imaging in human subjects in whom white matter atrophy and synaptic disconnection were observed early in AD pathogenesis [75-78]. Similarly, early white matter atrophy is also observed in women carrying the Apolipoprotein E (ApoE) allele ϵ4 [79], the major genetic risk factor for both familial and sporadic form of AD [80]. This is in agreement with the proposed endogenous function of ApoE in neuronal response to stress and injury [81,82].

Non-demented individuals with AD neuropathology burden

Interestingly, approximately 25% of cognitively healthy elderly people have Aβ plaque and tangle pathology [83], sufficient to meet National Institute on Aging (NIA)-Reagan criteria for AD [84-86]. According to the amyloid cascade hypothesis these individuals would have developed dementia if they had lived longer, hence the name “prodromal Alzheimer patients”. Nevertheless, longitudinal cognitive tests in non-demented individuals above 90 years of age, but with high AD neuropathology, showed no evidence of cognitive decline three years before death [87]. Interestingly, while there was extensive overlap in Aβ peptides profile in AD patients and non-demented high pathology individuals [88], the levels of APP and Aβ42 were higher in the non-demented group [89]. In agreement with an anti-inflammatory role of Aβ peptides [32] there was less neuroinflammation in high-pathology non-demented individuals [90-92]. In addition, when compared to AD patients, these high-pathology non-demented individuals showed higher levels of neuroprotective ApoE and S100B, as well as angiogenic vascular endothelial growth factor (VEGF) [89]. Such non-demented individuals may, therefore, be successful in maintaining or recruiting additional protective pathways to fight age-associated neuroinflammatory stress [93] and its associated neurodegeneration [44]. If so, APP and Aβ may play a significant role in this protection. This idea is in line with a transcriptional profiling study, showing that a decisive point in AD neuropathogenesis is the induction of stress genes as a reaction of the brain to age-associated changes in lipid metabolism and increasing inflammatory stress [94].

While targeting inflammatory processes, as suggested by some studies and epidemiological data [95,96], may prove efficacious in preventing or delaying the development of AD, these treatments failed to show a beneficial effect in patients with symptomatic AD [97]. Hence, in order to design therapeutics that would have efficacy in already diagnosed, late-stage, AD patients we will have to exploit as yet undiscovered factors or pathways employed by the non-demented individuals, who exhibit high levels of neuropatholgical change. In a search for such protective factors, a recent genome-wide association study identified three of the top ten hits as single-nucleotide polymorphisms (SNPs) in the promoter region of a gene encoding for the glycoprotein Reelin [98]. Interestingly, Reelin is an extracellular signaling molecule shown to (i) modulate LTP, (ii) be a strong suppressor of Tau hyperphosphorylation, and (iii) modulate APP processing (reviewed in [99]). Important, compared to age-matched controls, Reelin levels are significantly lower in AD patients already at early stages of the disease [100,101], but in high-pathology non-demented individuals Reelin production is increased [98]. It was also demonstrated that employing histone deacetylase 2 (HDAC) inhibitor following severe experimentally-induced neurodegeneration virtually restored brain functionality [102]. This is specifically interesting since Reelin promoter is known to be sensitive to epigenetic changes [103]. Therefore, exploiting this and similar pathways might be a promising avenue for development of treatments for symptomatic AD patients. However, we are afraid that the pursuit of strategies other than those directly targeting Aβ may have been hindered by the early and prolonged adherence to the Amyloid theory.

The airbag problem–a hypothetical example

The idea that the observed increase in Aβ levels as well as Aβ-containing senile plaques in AD patients is an adaptive response of the brain to an underlying stress/injury [44,94,104] has been put forward by a number of scientists [105-108]. Yet, many researchers continue to treat Aβ as the principle etiological agent in AD. While some might argue that an Aβ protective versus toxic role depends on the local concentration, it’s aggregation or oligomerization status, or the context in which Aβ is produced, we propose here an alternative possibility–namely that the scientific community is being misled by a seemingly overwhelming amount of literature that supports the Aβ hypothesis.

The rationale for making this strong and provocative statement against the amyloid hypothesis is easy to understand if you imagine a scenario in which someone, not knowing how a modern car works, is interpreting the significant association of car accidents with the presence of released airbags. Based on observations that released airbags are consistently observed in crashed cars, but never in intact cars, the airbag became the suspect and possible initiating cause of the accidents. Reinforced by the discovery that in a small percentage of car accidents (<1%) the spontaneous airbag release, due to a manufacturing defect, actually caused the accidence, the engineers design remote-controlled airbags to test the effect of their release in moving cars. Not knowing the real purpose of this protective device, the outcome and interpretation of this experiment–all performed with state-of-the-art technology, well-developed and executed strategies, and proper statistics–is absolutely clear: the airbag must be the cause. This strong evidence would certainly attract many specialists from different fields to investigate the airbag release mechanism in detail. They will eventually succeed, perhaps after years of hard work and accumulation of an overwhelming number of scientific papers, by changing only one important part of the release mechanism or installation of an airbag deployment preventer to hinder the experimentally induced airbag-associated car accidents. Since such intervention would prevent even small percentage of accidents caused by manufacturing defects that lead to unprovoked airbag deployment, the “airbag hypothesis” would become a well-established theory. Nevertheless the production of cars without airbags or removal of the airbags from all existing cars would not decrease the number of accidents in general-an obvious result if the real purpose of the airbag was known.

If we translate this hypothetical scenario to AD research, it might provide the explanation for the lack of translational success despite tremendous work and money invested in the past 20 yearsa. We argue here that the overwhelming data produced through the use of transgenic overexpression models of AD, which are not suitable for the understanding of the disease etiology, led the scientific community down the garden path. We would like to stress here, however, that the aim of this alternative view is not to put down the hard work being done by the AD-research community, but to encourage “outside the box” thinking toward initiating an important discussion and designing of future research directions–beyond Aβ. Finally, although we have focused in this article only on APP and Aβ in the context of Alzheimer’s disease, the implication of a potential “airbag problem” for bench-to-bedside translational efforts is not restricted to research on this protein and its derivatives nor to AD-research only.

Conclusions

Alzheimer’s disease (AD) represents one of the major health-issues of modern society with no promising treatment on the horizon. A potential problem underlying the failure to treat AD in humans is the assumption that senile plaques, the most obvious pathological hallmark of the disease, and especially one of the plaque components–Aβ peptide–represent the major driving force of the disease. Here, we challenge this view (see the airbag problem) and review experimental evidence in support of the hypothesis that APP/Aβ belong to a stress/injury-induced protective system of neurons. Hence, we suggest that AD-associated mutations in APP as well as PS1/2 impair an endogenous–protective–function of these proteins, which will in turn result in elevated neuronal vulnerability, diminution of membrane repair following brain injury, and impairments in axonal transport and axonal integrity when cellular stress becomes chronic. In the context of late-onset AD, chronic inflammatory stress to neurons would, with advancing age, override this protective mechanism leading to the same pathological changes as those observed at earlier age in familial AD cases. These impairments would cause synaptic deterioration, neuronal degeneration, and ultimately dementia. In line with this hypothesis and recognized anti-inflammatory properties of Aβ oligomers, senile plaques may represent an ‘aide de camp’ in the neuronal battle against age-associated and inflammation-driven neurodegeneration rather than the cause of neuronal cell death. Finally, understanding the protective mechanism(s) in cognitively healthy elderly people with high plaque and tangle burden, may hold a key for the development of successful treatments for patients already diagnosed with late-onset Alzheimer’s disease.

Endnote

aEstimate of more than 300 published animal model drug trials is based on: Zahs KR, Ashe KH. ‘Too much good news’-are Alzheimer mouse models trying to tell us how to prevent, not cure, Alzheimer’s disease? Trends Neurosci. 2010; 33(8) 381-9.

Competing interests

Both authors declare that they have no competing interests.

Contributor Information

Dimitrije Krstic, Email: krstic@pharma.uzh.ch.

Irene Knuesel, Email: knuesel@pharma.uzh.ch.

Search strategy and selection criteria

References for this article were identified through searches of PubMed and using Google search engine with the search terms “Alzheimer”, “amyloid-beta / Aβ”, “amyloid precursor protein / APP”, “neuroprotective”, “beneficial”, “adaptive”, without date restriction. Further articles were also identified through searches of the reference lists of selected articles. Only papers published in English were reviewed. The final reference list was generated on the basis of originality and relevance to the scope of the manuscript.

Acknowledgements

We would like to thank Prof. Dr. Sue W Griffin for comments on the manuscript. This study was supported by the Swiss National Science Foundation, grant number 310,030-132,629, the Gottfried und Julia Bangerter-Rhyner Foundation, and the Olga Mayenfisch Foundation.

References

  1. Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;1(5054):184–185. doi: 10.1126/science.1566067. [DOI] [PubMed] [Google Scholar]
  2. Van Broeckhoven C, Haan J, Bakker E, Hardy JA, Van Hul W, Wehnert A, Vegter-Van der Vlis M, Roos RA. Amyloid beta protein precursor gene and hereditary cerebral hemorrhage with amyloidosis (Dutch) Science. 1990;1(4959):1120–1122. doi: 10.1126/science.1971458. [DOI] [PubMed] [Google Scholar]
  3. Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, James L. et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature. 1991;1(6311):704–706. doi: 10.1038/349704a0. [DOI] [PubMed] [Google Scholar]
  4. St George-Hyslop P, Haines J, Rogaev E, Mortilla M, Vaula G, Pericak-Vance M, Foncin JF, Montesi M, Bruni A, Sorbi S. et al. Genetic evidence for a novel familial Alzheimer’s disease locus on chromosome 14. Nat Genet. 1992;1(4):330–334. doi: 10.1038/ng1292-330. [DOI] [PubMed] [Google Scholar]
  5. Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K. et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;1(6534):754–760. doi: 10.1038/375754a0. [DOI] [PubMed] [Google Scholar]
  6. McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, Yu C, Skipper L, Murphy MP, Beard J, Das P. et al. Abeta42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron. 2005;1(2):191–199. doi: 10.1016/j.neuron.2005.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Kim J, Chakrabarty P, Hanna A, March A, Dickson DW, Borchelt DR, Golde T, Janus C. Normal cognition in transgenic BRI2-Abeta mice. Mol Neurodegeneration. 2013;1(1):15. doi: 10.1186/1750-1326-8-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Flood DG, Reaume AG, Dorfman KS, Lin YG, Lang DM, Trusko SP, Savage MJ, Annaert WG, De Strooper B, Siman R. et al. FAD mutant PS-1 gene-targeted mice: increased A beta 42 and A beta deposition without APP overproduction. Neurobiology of aging. 2002;1(3):335–348. doi: 10.1016/S0197-4580(01)00330-X. [DOI] [PubMed] [Google Scholar]
  9. Guo Q, Wang Z, Li H, Wiese M, Zheng H. APP physiological and pathophysiological functions: insights from animal models. Cell research. 2012;1(1):78–89. doi: 10.1038/cr.2011.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Haroutunian V, Zhou Y, Elder G, Li C, Lazzarini RA. Age-dependent spatial memory deficits in transgenic mice expressing the human mid-sized neurofilament gene: I. Brain Res Mol Brain Res. 1996;1(1):62–70. doi: 10.1016/S0169-328X(96)00114-3. [DOI] [PubMed] [Google Scholar]
  11. Simon AM, Schiapparelli L, Salazar-Colocho P, Cuadrado-Tejedor M, Escribano L, Lopez de Maturana R, Del Rio J, Perez-Mediavilla A, Frechilla D. Overexpression of wild-type human APP in mice causes cognitive deficits and pathological features unrelated to Abeta levels. Neurobiol Dis. 2009;1(3):369–378. doi: 10.1016/j.nbd.2008.11.005. [DOI] [PubMed] [Google Scholar]
  12. Schwab C, Hosokawa M, McGeer PL. Transgenic mice overexpressing amyloid beta protein are an incomplete model of Alzheimer disease. Exp Neurol. 2004;1(1):52–64. doi: 10.1016/j.expneurol.2004.03.016. [DOI] [PubMed] [Google Scholar]
  13. Kohler C, Ebert U, Baumann K, Schroder H. Alzheimer’s disease-like neuropathology of gene-targeted APP-SLxPS1mut mice expressing the amyloid precursor protein at endogenous levels. Neurobiol Dis. 2005;1(2):528–540. doi: 10.1016/j.nbd.2005.04.009. [DOI] [PubMed] [Google Scholar]
  14. Wirths O, Bayer TA. Neuron loss in transgenic mouse models of Alzheimer’s disease. Int J Alzheimer’s Dis. 2010;1:723782. doi: 10.4061/2010/723782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L. 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;1(40):10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;1(5580):353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  17. Selkoe DJ. Resolving controversies on the path to Alzheimer’s therapeutics. Nat Med. 2011;1(9):1060–1065. doi: 10.1038/nm.2460. [DOI] [PubMed] [Google Scholar]
  18. Benilova I, Karran E, De Strooper B. The toxic Abeta oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;1(3):349–357. doi: 10.1038/nn.3028. [DOI] [PubMed] [Google Scholar]
  19. Whitson JS, Selkoe DJ, Cotman CW. Amyloid beta protein enhances the survival of hippocampal neurons in vitro. Science. 1989;1(4897):1488–1490. doi: 10.1126/science.2928783. [DOI] [PubMed] [Google Scholar]
  20. Koo EH, Park L, Selkoe DJ. Amyloid beta-protein as a substrate interacts with extracellular matrix to promote neurite outgrowth. Proc Natl Acad Sci U S A. 1993;1(10):4748–4752. doi: 10.1073/pnas.90.10.4748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA. The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J Neurosci. 2003;1(13):5531–5535. doi: 10.1523/JNEUROSCI.23-13-05531.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Lopez-Toledano MA, Shelanski ML. Neurogenic effect of beta-amyloid peptide in the development of neural stem cells. J Neurosci. 2004;1(23):5439–5444. doi: 10.1523/JNEUROSCI.0974-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski A, Palmeri A, Arancio O. Picomolar amyloid-beta positively modulates synaptic plasticity and memory in hippocampus. J Neurosci. 2008;1(53):14537–14545. doi: 10.1523/JNEUROSCI.2692-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Lesne S, Ali C, Gabriel C, Croci N, MacKenzie ET, Glabe CG, Plotkine M, Marchand-Verrecchia C, Vivien D, Buisson A. NMDA receptor activation inhibits alpha-secretase and promotes neuronal amyloid-beta production. J Neurosci. 2005;1(41):9367–9377. doi: 10.1523/JNEUROSCI.0849-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Atwood CS, Obrenovich ME, Liu T, Chan H, Perry G, Smith MA, Martins RN. Amyloid-beta: a chameleon walking in two worlds: a review of the trophic and toxic properties of amyloid-beta. Brain Res Brain Res Rev. 2003;1(1):1–16. doi: 10.1016/S0165-0173(03)00174-7. [DOI] [PubMed] [Google Scholar]
  26. Nunomura A, Perry G, Pappolla MA, Friedland RP, Hirai K, Chiba S, Smith MA. Neuronal oxidative stress precedes amyloid-beta deposition in down syndrome. J Neuropathol Exp Neurol. 2000;1(11):1011–1017. doi: 10.1093/jnen/59.11.1011. [DOI] [PubMed] [Google Scholar]
  27. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S. et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;1(8):759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
  28. Pratico D, Clark CM, Liun F, Rokach J, Lee VY, Trojanowski JQ. Increase of brain oxidative stress in mild cognitive impairment: a possible predictor of Alzheimer disease. Arch Neurol. 2002;1(6):972–976. doi: 10.1001/archneur.59.6.972. [DOI] [PubMed] [Google Scholar]
  29. Nunomura A, Tamaoki T, Motohashi N, Nakamura M, McKeel DW Jr, Tabaton M, Lee HG, Smith MA, Perry G, Zhu X. The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation in vulnerable neurons. J Neuropathol Exp Neurol. 2012;1(3):233–241. doi: 10.1097/NEN.0b013e318248e614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Nunomura A, Tamaoki T, Tanaka K, Motohashi N, Nakamura M, Hayashi T, Yamaguchi H, Shimohama S, Lee HG, Zhu X. et al. Intraneuronal amyloid beta accumulation and oxidative damage to nucleic acids in Alzheimer disease. Neurobiol Dis. 2010;1(3):731–737. doi: 10.1016/j.nbd.2009.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Kontush A, Berndt C, Weber W, Akopyan V, Arlt S, Schippling S, Beisiegel U. Amyloid-beta is an antioxidant for lipoproteins in cerebrospinal fluid and plasma. Free Radic Biol Med. 2001;1(1):119–128. doi: 10.1016/S0891-5849(00)00458-5. [DOI] [PubMed] [Google Scholar]
  32. Kurnellas MP, Adams CM, Sobel RA, Steinman L, Rothbard JB. Amyloid fibrils composed of hexameric peptides attenuate neuroinflammation. Sci Transl Med. 2013;1(179):179ra142. doi: 10.1126/scitranslmed.3005681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, Hyman B, Burton MA, Goldstein LE, Duong S, Tanzi RE. et al. The Alzheimer’s disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS One. 2010;1(3):e9505. doi: 10.1371/journal.pone.0009505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Little CS, Hammond CJ, MacIntyre A, Balin BJ, Appelt DM. Chlamydia pneumoniae induces Alzheimer-like amyloid plaques in brains of BALB/c mice. Neurobiol Aging. 2004;1(4):419–429. doi: 10.1016/S0197-4580(03)00127-1. [DOI] [PubMed] [Google Scholar]
  35. Wozniak MA, Itzhaki RF, Shipley SJ, Dobson CB. Herpes simplex virus infection causes cellular beta-amyloid accumulation and secretase upregulation. Neurosci Lett. 2007;1(2-3):95–100. doi: 10.1016/j.neulet.2007.09.077. [DOI] [PubMed] [Google Scholar]
  36. Zhao J, O’Connor T, Vassar R. The contribution of activated astrocytes to Abeta production: implications for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2011;1:150. doi: 10.1186/1742-2094-8-150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Monning U, Konig G, Banati RB, Mechler H, Czech C, Gehrmann J, Schreiter-Gasser U, Masters CL, Beyreuther K. Alzheimer beta A4-amyloid protein precursor in immunocompetent cells. J Biol Chem. 1992;1(33):23950–23956. [PubMed] [Google Scholar]
  38. Spitzer P, Herrmann M, Klafki HW, Smirnov A, Lewczuk P, Kornhuber J, Wiltfang J, Maler JM. Phagocytosis and LPS alter the maturation state of beta-amyloid precursor protein and induce different Abeta peptide release signatures in human mononuclear phagocytes. J Neuroinflammation. 2010;1:59. doi: 10.1186/1742-2094-7-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Miklossy J. Alzheimer’s disease–a neurospirochetosis: analysis of the evidence following Koch’s and Hill’s criteria. J Neuroinflammation. 2011;1:90. doi: 10.1186/1742-2094-8-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kamer AR, Craig RG, Pirraglia E, Dasanayake AP, Norman RG, Boylan RJ, Nehorayoff A, Glodzik L, Brys M, De Leon MJ. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J Neuroimmunol. 2009;1(1-2):92–97. doi: 10.1016/j.jneuroim.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Calderon-Garciduenas L, Kavanaugh M, Block M, D’Angiulli A, Delgado-Chavez R, Torres-Jardon R, Gonzalez-Maciel A, Reynoso-Robles R, Osnaya N, Villarreal-Calderon R. et al. Neuroinflammation, hyperphosphorylated tau, diffuse amyloid plaques, and down-regulation of the cellular prion protein in air pollution exposed children and young adults. J Alzheimers Dis. 2012;1(1):93–107. doi: 10.3233/JAD-2011-110722. [DOI] [PubMed] [Google Scholar]
  42. Fischer O. Miliare Nekrosen mit drusigen Wucherungen der Neurofibrillen, eine regelmässige Veränderung der Hirnrinde bei seniler Demenz. Monatsschr Psychiatr Neurol. 1907;1:361–372. doi: 10.1159/000211873. [DOI] [Google Scholar]
  43. Xiao AW, He J, Wang Q, Luo Y, Sun Y, Zhou YP, Guan Y, Lucassen PJ, Dai JP. The origin and development of plaques and phosphorylated tau are associated with axonopathy in Alzheimer’s disease. Neurosci Bull. 2011;1(5):287–299. doi: 10.1007/s12264-011-1736-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Krstic D, Knuesel I. Deciphering the mechanism underlying late-onset Alzheimer disease. Nat Rev Neurol. 2013;1(1):25–34. doi: 10.1038/nrneurol.2012.236. [DOI] [PubMed] [Google Scholar]
  45. Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, De Calignon A, Rozkalne A, Koenigsknecht-Talboo J, Holtzman DM, Bacskai BJ, Hyman BT. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature. 2008;1(7179):720–724. doi: 10.1038/nature06616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Novelli A, Reilly JA, Lysko PG, Henneberry RC. Glutamate becomes neurotoxic via the N-methyl-D-aspartate receptor when intracellular energy levels are reduced. Brain research. 1988;1(1-2):205–212. doi: 10.1016/0006-8993(88)90765-2. [DOI] [PubMed] [Google Scholar]
  47. Zheng H, Koo EH. The amyloid precursor protein: beyond amyloid. Mol Neurodegener. 2006;1:5. doi: 10.1186/1750-1326-1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Phinney AL, Calhoun ME, Wolfer DP, Lipp HP, Zheng H, Jucker M. No hippocampal neuron or synaptic bouton loss in learning-impaired aged beta-amyloid precursor protein-null mice. Neuroscience. 1999;1(4):1207–1216. doi: 10.1016/S0306-4522(98)00645-9. [DOI] [PubMed] [Google Scholar]
  49. Corrigan F, Vink R, Blumbergs PC, Masters CL, Cappai R, van den Heuvel C. Characterisation of the effect of knockout of the amyloid precursor protein on outcome following mild traumatic brain injury. Brain Res. 2012;1:87–99. doi: 10.1016/j.brainres.2012.02.045. [DOI] [PubMed] [Google Scholar]
  50. Steinbach JP, Muller U, Leist M, Li ZW, Nicotera P, Aguzzi A. Hypersensitivity to seizures in beta-amyloid precursor protein deficient mice. Cell Death Differ. 1998;1(10):858–866. doi: 10.1038/sj.cdd.4400391. [DOI] [PubMed] [Google Scholar]
  51. Koike MA, Lin AJ, Pham J, Nguyen E, Yeh JJ, Rahimian R, Tromberg BJ, Choi B, Green KN, LaFerla FM. APP knockout mice experience acute mortality as the result of ischemia. PLoS One. 2012;1(8):e42665. doi: 10.1371/journal.pone.0042665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Griffin WS, Sheng JG, Gentleman SM, Graham DI, Mrak RE, Roberts GW. Microglial interleukin-1 alpha expression in human head injury: correlations with neuronal and neuritic beta-amyloid precursor protein expression. Neurosci Lett. 1994;1(2):133–136. doi: 10.1016/0304-3940(94)90066-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Murakami N, Yamaki T, Iwamoto Y, Sakakibara T, Kobori N, Fushiki S, Ueda S. Experimental brain injury induces expression of amyloid precursor protein, which may be related to neuronal loss in the hippocampus. J Neurotrauma. 1998;1(11):993–1003. doi: 10.1089/neu.1998.15.993. [DOI] [PubMed] [Google Scholar]
  54. Chen XH, Siman R, Iwata A, Meaney DF, Trojanowski JQ, Smith DH. Long-term accumulation of amyloid-beta, beta-secretase, presenilin-1, and caspase-3 in damaged axons following brain trauma. Am J Pathol. 2004;1(2):357–371. doi: 10.1016/S0002-9440(10)63303-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Uryu K, Chen XH, Martinez D, Browne KD, Johnson VE, Graham DI, Lee VM, Trojanowski JQ, Smith DH. Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans. Exp Nephrol. 2007;1(2):185–192. doi: 10.1016/j.expneurol.2007.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Ramirez MJ, Heslop KE, Francis PT, Rattray M. Expression of amyloid precursor protein, tau and presenilin RNAs in rat hippocampus following deafferentation lesions. Brain Res. 2001;1(1-2):222–232. doi: 10.1016/S0006-8993(01)02580-X. [DOI] [PubMed] [Google Scholar]
  57. Koistinaho J, Pyykonen I, Keinanen R, Hokfelt T. Expression of beta-amyloid precursor protein mRNAs following transient focal ischaemia. Neuroreport. 1996;1(15-17):2727–2731. doi: 10.1097/00001756-199611040-00064. [DOI] [PubMed] [Google Scholar]
  58. Finnie JW, Manavis J, Blumbergs PC, Kuchel TR. Axonal and neuronal amyloid precursor protein immunoreactivity in the brains of guinea pigs given tunicamycin. Vet Pathol. 2000;1(6):677–680. doi: 10.1354/vp.37-6-677. [DOI] [PubMed] [Google Scholar]
  59. Whitehead SN, Hachinski VC, Cechetto DF. Interaction between a rat model of cerebral ischemia and beta-amyloid toxicity: inflammatory responses. Stroke. 2005;1(1):107–112. doi: 10.1161/01.STR.0000149627.30763.f9. [DOI] [PubMed] [Google Scholar]
  60. Lee JW, Lee YK, Yuk DY, Choi DY, Ban SB, Oh KW, Hong JT. Neuro-inflammation induced by lipopolysaccharide causes cognitive impairment through enhancement of beta-amyloid generation. J Neuroinflammation. 2008;1:37. doi: 10.1186/1742-2094-5-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C, Manalastas A, Hilfiker M, Pfister S, Schwerdel C. et al. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J Neuroinflammation. 2012;1:151. doi: 10.1186/1742-2094-9-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Sheng JG, Ito K, Skinner RD, Mrak RE, Rovnaghi CR, Van Eldik LJ, Griffin WS. In vivo and in vitro evidence supporting a role for the inflammatory cytokine interleukin-1 as a driving force in Alzheimer pathogenesis. Neurobiol Aging. 1996;1(5):761–766. doi: 10.1016/0197-4580(96)00104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Liu L, Aboud O, Jones RA, Mrak RE, Griffin WS, Barger SW. Apolipoprotein E expression is elevated by interleukin 1 and other interleukin 1-induced factors. J Neuroinflammation. 2011;1:175. doi: 10.1186/1742-2094-8-175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Barger SW, DeWall KM, Liu L, Mrak RE, Griffin WS. Relationships between expression of apolipoprotein E and beta-amyloid precursor protein are altered in proximity to Alzheimer beta-amyloid plaques: potential explanations from cell culture studies. J Neuropathol Exp Neurol. 2008;1(8):773–783. doi: 10.1097/NEN.0b013e318180ec47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Leyssen M, Ayaz D, Hebert SS, Reeve S, De Strooper B, Hassan BA. Amyloid precursor protein promotes post-developmental neurite arborization in the Drosophila brain. Embo J. 2005;1(16):2944–2955. doi: 10.1038/sj.emboj.7600757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Wang P, Yang G, Mosier DR, Chang P, Zaidi T, Gong YD, Zhao NM, Dominguez B, Lee KF, Gan WB. et al. Defective neuromuscular synapses in mice lacking amyloid precursor protein (APP) and APP-Like protein 2. J Neurosci. 2005;1(5):1219–1225. doi: 10.1523/JNEUROSCI.4660-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, Stefansson H, Sulem P, Gudbjartsson D, Maloney J. et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;1(7409):96–99. doi: 10.1038/nature11283. [DOI] [PubMed] [Google Scholar]
  68. Dawson GR, Seabrook GR, Zheng H, Smith DW, Graham S, O’Dowd G, Bowery BJ, Boyce S, Trumbauer ME, Chen HY. et al. Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the beta-amyloid precursor protein. Neuroscience. 1999;1(1):1–13. doi: 10.1016/S0306-4522(98)00410-2. [DOI] [PubMed] [Google Scholar]
  69. Ring S, Weyer SW, Kilian SB, Waldron E, Pietrzik CU, Filippov MA, Herms J, Buchholz C, Eckman CB, Korte M. et al. The secreted beta-amyloid precursor protein ectodomain APPs alpha is sufficient to rescue the anatomical, behavioral, and electrophysiological abnormalities of APP-deficient mice. J Neurosci. 2007;1(29):7817–7826. doi: 10.1523/JNEUROSCI.1026-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Terry RD. The pathogenesis of Alzheimer disease: an alternative to the amyloid hypothesis. J Neuropathol Exp Neurol. 1996;1(10):1023–1025. [PubMed] [Google Scholar]
  71. Kamal A, Almenar-Queralt A, LeBlanc JF, Roberts EA, Goldstein LS. Kinesin-mediated axonal transport of a membrane compartment containing beta-secretase and presenilin-1 requires APP. Nature. 2001;1(6864):643–648. doi: 10.1038/414643a. [DOI] [PubMed] [Google Scholar]
  72. Smith KD, Peethumnongsin E, Lin H, Zheng H, Pautler RG. Increased human wildtype tau attenuates axonal transport deficits caused by loss of APP in mouse models. Magn Reson Insights. 2010;1:11–18. doi: 10.4137/mri.s5237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Rodrigues EM, Weissmiller AM, Goldstein LS. Enhanced beta-secretase processing alters APP axonal transport and leads to axonal defects. Hum Mol Genet. 2012;1(21):4587–4601. doi: 10.1093/hmg/dds297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Stokin GB, Almenar-Queralt A, Gunawardena S, Rodrigues EM, Falzone T, Kim J, Lillo C, Mount SL, Roberts EA, McGowan E. et al. Amyloid precursor protein-induced axonopathies are independent of amyloid-beta peptides. Hum Mol Genet. 2008;1(22):3474–3486. doi: 10.1093/hmg/ddn240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Rose SE, Chen F, Chalk JB, Zelaya FO, Strugnell WE, Benson M, Semple J, Doddrell DM. Loss of connectivity in Alzheimer’s disease: an evaluation of white matter tract integrity with colour coded MR diffusion tensor imaging. J Neurol Neurosurg Psychiatry. 2000;1(4):528–530. doi: 10.1136/jnnp.69.4.528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Huang J, Friedland RP, Auchus AP. Diffusion tensor imaging of normal-appearing white matter in mild cognitive impairment and early Alzheimer disease: preliminary evidence of axonal degeneration in the temporal lobe. AJNR Am J Neuroradiol. 2007;1(10):1943–1948. doi: 10.3174/ajnr.A0700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Ringman JM, O’Neill J, Geschwind D, Medina L, Apostolova LG, Rodriguez Y, Schaffer B, Varpetian A, Tseng B, Ortiz F. et al. Diffusion tensor imaging in preclinical and presymptomatic carriers of familial Alzheimer’s disease mutations. Brain. 2007;1(Pt 7):1767–1776. doi: 10.1093/brain/awm102. [DOI] [PubMed] [Google Scholar]
  78. Gili T, Cercignani M, Serra L, Perri R, Giove F, Maraviglia B, Caltagirone C, Bozzali M. Regional brain atrophy and functional disconnection across Alzheimer’s disease evolution. J Neurol Neurosurg Psychiatry. 2011;1(1):58–66. doi: 10.1136/jnnp.2009.199935. [DOI] [PubMed] [Google Scholar]
  79. Gold BT, Powell DK, Andersen AH, Smith CD. Alterations in multiple measures of white matter integrity in normal women at high risk for Alzheimer’s disease. Neuroimage. 2010;1(4):1487–1494. doi: 10.1016/j.neuroimage.2010.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Genin E, Hannequin D, Wallon D, Sleegers K, Hiltunen M, Combarros O, Bullido MJ, Engelborghs S, De Deyn P, Berr C. et al. APOE and Alzheimer disease: a major gene with semi-dominant inheritance. Mol Psychiatry. 2011;1(9):903–907. doi: 10.1038/mp.2011.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Aboud O, Mrak RE, Boop F, Griffin ST. Apolipoprotein epsilon 3 alleles are associated with indicators of neuronal resilience. BMC Med. 2012;1:35. doi: 10.1186/1741-7015-10-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Mahley RW, Huang Y. Apolipoprotein e sets the stage: response to injury triggers neuropathology. Neuron. 2012;1(5):871–885. doi: 10.1016/j.neuron.2012.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Jicha GA, Abner EL, Schmitt FA, Kryscio RJ, Riley KP, Cooper GE, Stiles N, Mendiondo MS, Smith CD, Van Eldik LJ. et al. Preclinical AD workgroup staging: pathological correlates and potential challenges. Neurobiol Aging. 2012;1(3):622 e1–622 e16. doi: 10.1016/j.neurobiolaging.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Crystal H, Dickson D, Fuld P, Masur D, Scott R, Mehler M, Masdeu J, Kawas C, Aronson M, Wolfson L. Clinico-pathologic studies in dementia: nondemented subjects with pathologically confirmed Alzheimer’s disease. Neurology. 1988;1(11):1682–1687. doi: 10.1212/WNL.38.11.1682. [DOI] [PubMed] [Google Scholar]
  85. Snowdon DA. Aging and Alzheimer’s disease: lessons from the nun study. Gerontologist. 1997;1(2):150–156. doi: 10.1093/geront/37.2.150. [DOI] [PubMed] [Google Scholar]
  86. Negash S, Bennett DA, Wilson RS, Schneider JA, Arnold SE. Cognition and neuropathology in aging: multidimensional perspectives from the rush religious orders study and rush memory and aging project. Curr Alzheimer Res. 2011;1(4):336–340. doi: 10.2174/156720511795745302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Balasubramanian AB, Kawas CH, Peltz CB, Brookmeyer R, Corrada MM. Alzheimer disease pathology and longitudinal cognitive performance in the oldest-old with no dementia. Neurology. 2012;1(9):915–921. doi: 10.1212/WNL.0b013e318266fc77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Moore BD, Chakrabarty P, Levites Y, Kukar TL, Baine AM, Moroni T, Ladd TB, Das P, Dickson DW, Golde TE. Overlapping profiles of Abeta peptides in the Alzheimer’s disease and pathological aging brains. Alzheimers Res Ther. 2012;1(3):18. doi: 10.1186/alzrt121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Maarouf CL, Daugs ID, Kokjohn TA, Walker DG, Hunter JM, Kruchowsky JC, Woltjer R, Kaye J, Castano EM, Sabbagh MN. et al. Alzheimer’s disease and non-demented high pathology control nonagenarians: comparing and contrasting the biochemistry of cognitively successful aging. PloS one. 2011;1(11):e27291. doi: 10.1371/journal.pone.0027291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lue LF, Brachova L, Civin WH, Rogers J. Inflammation, A beta deposition, and neurofibrillary tangle formation as correlates of Alzheimer’s disease neurodegeneration. J Neuropathol Exp Neurol. 1996;1(10):1083–1088. [PubMed] [Google Scholar]
  91. Parachikova A, Agadjanyan MG, Cribbs DH, Blurton-Jones M, Perreau V, Rogers J, Beach TG, Cotman CW. Inflammatory changes parallel the early stages of Alzheimer disease. Neurobiol Aging. 2007;1(12):1821–1833. doi: 10.1016/j.neurobiolaging.2006.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Morimoto K, Horio J, Satoh H, Sue L, Beach T, Arita S, Tooyama I, Konishi Y. Expression profiles of cytokines in the brains of Alzheimer’s disease (AD) patients compared to the brains of non-demented patients with and without increasing AD pathology. JAD. 2011;1(1):59–76. doi: 10.3233/JAD-2011-101815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging: an evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;1:244–254. doi: 10.1111/j.1749-6632.2000.tb06651.x. [DOI] [PubMed] [Google Scholar]
  94. Podtelezhnikov AA, Tanis KQ, Nebozhyn M, Ray WJ, Stone DJ, Loboda AP. Molecular insights into the pathogenesis of Alzheimer’s disease and its relationship to normal aging. PloS one. 2011;1(12):e29610. doi: 10.1371/journal.pone.0029610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Vlad SC, Miller DR, Kowall NW, Felson DT. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;1(19):1672–1677. doi: 10.1212/01.wnl.0000311269.57716.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Breitner JC, Baker LD, Montine TJ, Meinert CL, Lyketsos CG, Ashe KH, Brandt J, Craft S, Evans DE, Green RC. et al. Extended results of the Alzheimer’s disease anti-inflammatory prevention trial. Alzheimers Dement. 2011;1(4):402–411. doi: 10.1016/j.jalz.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, Farlow MR, Jin S, Thomas RG, Thal LJ. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;1(21):2819–2826. doi: 10.1001/jama.289.21.2819. [DOI] [PubMed] [Google Scholar]
  98. Kramer PL, Xu H, Woltjer RL, Westaway SK, Clark D, Erten-Lyons D, Kaye JA, Welsh-Bohmer KA, Troncoso JC, Markesbery WR. et al. Alzheimer disease pathology in cognitively healthy elderly: a genome-wide study. Neurobiology of aging. 2011;1(12):2113–2122. doi: 10.1016/j.neurobiolaging.2010.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Krstic D, Pfister S, Notter T, Knuesel I. Decisive role of Reelin signaling during early stages of Alzheimer’s disease. Neuroscience. 2013;1:108–116. doi: 10.1016/j.neuroscience.2013.04.042. [DOI] [PubMed] [Google Scholar]
  100. Chin J, Massaro CM, Palop JJ, Thwin MT, Yu GQ, Bien-Ly N, Bender A, Mucke L. Reelin depletion in the entorhinal cortex of human amyloid precursor protein transgenic mice and humans with Alzheimer’s disease. J Neurosci. 2007;1(11):2727–2733. doi: 10.1523/JNEUROSCI.3758-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Herring A, Donath A, Steiner KM, Widera MP, Hamzehian S, Kanakis D, Kolble K, ElAli A, Hermann DM, Paulus W. et al. Reelin depletion is an early phenomenon of Alzheimer’s pathology. JAD. 2012;1(4):963–979. doi: 10.3233/JAD-2012-112069. [DOI] [PubMed] [Google Scholar]
  102. Graff J, Rei D, Guan JS, Wang WY, Seo J, Hennig KM, Nieland TJ, Fass DM, Kao PF, Kahn M. et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature. 2012;1(7388):222–226. doi: 10.1038/nature10849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Sui L, Wang Y, Ju LH, Chen M. Epigenetic regulation of reelin and brain-derived neurotrophic factor genes in long-term potentiation in rat medial prefrontal cortex. Neurobiol Learn Mem. 2012;1(4):425–440. doi: 10.1016/j.nlm.2012.03.007. [DOI] [PubMed] [Google Scholar]
  104. Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL 3rd, Araoz C. Brain interleukin 1 and S-100 immunoreactivity are elevated in down syndrome and Alzheimer disease. Proc Natl Acad Sci U S A. 1989;1(19):7611–7615. doi: 10.1073/pnas.86.19.7611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Smith MA, Joseph JA, Perry G. Arson: tracking the culprit in Alzheimer’s disease. Ann N Y Acad Sci. 2000;1:35–38. doi: 10.1111/j.1749-6632.2000.tb05557.x. [DOI] [PubMed] [Google Scholar]
  106. Castellani RJ, Lee HG, Siedlak SL, Nunomura A, Hayashi T, Nakamura M, Zhu X, Perry G, Smith MA. Reexamining Alzheimer’s disease: evidence for a protective role for amyloid-beta protein precursor and amyloid-beta. JAD. 2009;1(2):447–452. doi: 10.3233/JAD-2009-1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Herrup K. Reimagining Alzheimer’s disease–an age-based hypothesis. J Neurosci. 2010;1(50):16755–16762. doi: 10.1523/JNEUROSCI.4521-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Stranahan AM, Mattson MP. Recruiting adaptive cellular stress responses for successful brain ageing. Nat Rev Neurosci. 2012;1(3):209–216. doi: 10.1038/nrn3151. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Acta Neuropathologica Communications are provided here courtesy of BMC

RESOURCES