Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2018 Sep 23;9(4):211–218. doi: 10.1111/cen3.12475

Neuroinflammation in mouse models of Alzheimer's disease

Takashi Saito 1,2,, Takaomi C Saido 1,
PMCID: PMC6282739  PMID: 30546389

Abstract

Alzheimer's disease (AD) is the most common type of neurocognitive disorder. Although both amyloid β peptide deposition and neurofibrillary tangle formation in the AD brain have been established as pathological hallmarks of the disease, many other factors contribute in a complex manner to the pathogenesis of AD before clinical symptoms of the disease become apparent. Longitudinal pathophysiological processes cause patients’ brains to exist in a state of chronic neuroinflammation, with glial cells acting as key regulators of the neuroinflammatory state. However, the detailed molecular and cellular mechanisms of glial function underlying AD pathogenesis remain elusive. Furthermore, recent studies have shown that peripheral inflammatory conditions affect glial cells in the brain through a process of neuroimmune communication. Such disease complexities make it difficult for the pathogenesis of AD to be understood, and impede the development of effective therapeutic strategies to combat the disease. Relevant AD animal models are thus likely to serve as a key resource to overcome many of these issues. Furthermore, as the pathogenesis of AD might be linked to conditions both within the brain as well as peripherally, it might become necessary for AD to be studied as a whole‐body disorder. The present review aimed to summarize insights regarding current AD research, and share perspectives for understanding glial function in the context of the pathogenesis of AD.

Keywords: Alzheimer's disease, glial cell, mouse model, neuroimmune communication, neuroinflammation

Introduction

Neurodegenerative disorders, such as Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease and Alzheimer's disease (AD), are proving to be the most difficult diseases to prevent or treat, and remain as unmet medical needs. AD is the primary cause of neurocognitive disorders in the elderly, and poses a huge socioeconomic burden for modern society. The number of patients with dementia is estimated to be >46 million people worldwide, and is increasing unabated each year. To the present time, only some symptomatic treatments have been found to be effective. To overcome this impasse and to develop effective treatments, elucidation of the molecular and cellular mechanisms underlying the pathogenesis of AD with a view to identifying druggable targets must be a priority.

Senile plaques composed of extracellular amyloid β peptide (Aβ) and neurofibrillary tangles (NFT), which are the aggregates of intracellular hyperphosphorylated tau protein, are hallmarks of the AD brain.1 Aβ deposition and NFT formation in the cortical region of the brain begin appearing 25–30 years and 15 years, respectively, before the clinical onset of AD (Fig. 1a).2 Aβ is generated proteolytically from amyloid precursor protein (APP) to subsequently form oligomeric Aβ, which aggregates into senile plaques. Although microtubule‐associated protein tau stabilizes microtubules in the axon, pathological tau mislocalizes through an unknown mechanism and forms NFT aggregates in the neuronal dendrites and cell body. These protein aggregates in the brain environment induce the activation of microglia and astrocytes, which results in microgliosis and astrocytosis around the pathological structures.

Figure 1.

Figure 1

(a) Time‐course of Alzheimer's disease (AD) progression. Amyloid β peptide (Aβ) deposition begins >25 years before the onset of AD and is followed by neurofibrillary tangles (NFT) formation. This leads to neurodegeneration and neuronal cell death. Both amyloid‐associated and tauopathy‐associated neuroinflammation might facilitate AD pathogenesis. (b) Immunohistochemical staining of gliosis in the human AD brain. 1‐Fluoro‐2,5‐bis(3‐carboxy‐4‐hydroxystyryl)benzene (blue fluorescence) binds to β‐sheet structures, such as dense‐cored Aβ plaques (left panel) and NFTs (right panel), respectively, with ionized calcium binding adaptor molecule 1 (Iba1)‐positive microgliosis shown in red and glial fibrillary acid protein (GFAP)‐positive astrocytosis in green. Scale bar, 20 μm.

Glial cells are thus chronically activated in the brain before the onset of AD,3 with the associated chronic inflammation contributing to the pathogenesis of AD. In the AD brain, microgliosis and astrocytosis as a consequence of the presence of senile plaques and NFTs can be detected immunohistochemically, with these glial cells showing a pathology‐specific morphology (Fig. 1b). Although the extent of gliosis is correlated with cortical thickness and neurodegeneration, the roles of different glial cells in neurodegenerative processes remain unclear.4 Relevant animal models are required for these processes to be investigated in greater detail.

Animal models for AD and neuroinflammation

Animal models representing relevant pathologies with as few artifactual anomalies as possible are necessary. To this end, a number of AD mouse models have been developed,5 with APP overexpressing mice, such as APP transgenic (APP Tg) mice, having been used widely,6, 7 although they are associated with considerable technical and physiological issues. For example, amyloid plaques in some APP Tg mice, particularly Tg2576 and APP23 mice, were found to be very large in size and composed mainly of Aβ40,8 making the plaques decidedly different from those seen in AD patients (Fig. 2). These findings were due to technical limitations associated with the animal models, which were based on an APP overexpression paradigm. To overcome these drawbacks, we created two strains of App knock‐in (KI) mice,9 named App NL‐F KI and App NL‐G‐F KI. App NL‐F KI mice harbor the Swedish mutation (NL) and the Iberian mutation (F), whereas App NL‐G‐F KI mice also harbor the Arctic mutation (G). Both App KI mouse strains showed relevant amyloid deposition composed of pathological Aβ42, similar to that in AD patients (Fig. 2).9 Advantages associated with using the App KI strains have been described,10, 11 with these mouse strains showing fewer artifactual anomalies compared with APP overexpressing mice.12, 13 However, we did not observe NFT in the App KI mice during their lifespan, suggesting that the mice might also be useful as preclinical AD mouse models to investigate the pathological role of amyloidosis and amyloid‐associated neuroinflammation. Hama et al. succeeded in using 3‐D visualization of resting and activated microglia in the brains of App KI mice and AD patients using an optical clearing technique, and showed that microglia are frequently associated with diffuse plaques in the AD brain.14 Zhang et al. further reported that App KI mice show mushroom spine loss,15 which could reflect microglia‐mediated synapse loss in AD.16 Furthermore, Castillo et al. reported amyloidosis‐dependent transcriptomic profiles in 3xTg AD mice17 and App NL‐G‐F KI mice.18 In contrast to 3xTg AD mice, App NL‐G‐F KI mice express genes in common with AD patients, such as neuroinflammation‐related genes (C4a/C4b, Cd74, Ctss, Gfap, Nfe212, Phyhd1, S100b, Tf, Tgfbr2 and Vim) and AD risk factor genes (Abi3, Apoe, Bin2, Cd33, Ctsc, Dock2, Fcer1g, Frmd6, Hck, Inpp5D, Ly86, Plcg2, Trem2 and Tyrobp).18 Thus, App KI mice might overcome some of the previous limitations associated with APP overexpression‐based mouse models, and could thus serve as a useful research tool for further investigations.

Figure 2.

Figure 2

Pathological differences in amyloidosis between mouse models and human brain tissue. Double staining for Aβ (anti‐Aβ antibody: 82E1, red) and ionized calcium binding adaptor molecule 1‐positive microgliosis (green) was carried out using brain sections obtained from amyloid precursor protein (APP)23 mice, App knock‐in mice and post‐mortem brain tissue from an Alzheimer's disease (AD) patient. Scale bar, 25 μm. F, Iberian mutation; G, Arctic mutation; NL, Swedish mutation.

Various tauopathy mouse models have also been generated,19 with most tau transgenic models harboring tau mutations known to be associated with frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP‐17), and not associated with AD.20 The mutations accelerate the self‐aggregation of tau or give rise to an isoform shift from 3‐repeat to 4‐repeat tau. In any case, tau transgenic mice containing FTDP‐17 mutations form NFT without amyloid plaque deposition, meaning that these mouse models might contribute to elucidation of the role of tauopathy‐associated neuroinflammation. The microglial phenotype changes from a ramified type to an amoeboid type during the development of tauopathy in rTg4510 mice.21 P301S‐tau Tg mice show reactive gliosis before tau aggregation, whereas immunosuppression of P301S‐tau Tg mice with FK506 attenuates tau pathology.22 In another scenario, amyloid deposition exacerbates NFT formation in JNPL3 transgenic mice.23 Although the pathomolecular mechanisms linking Aβ deposition to NFT formation, or NFT formation to neurodegeneration and neuronal cell death remain unclear, these outcomes suggest that neuroinflammation could link amyloid pathology and NFT formation as an important pathophysiological event in the development of AD. To further elucidate a pathological role of neuroinflammation in AD, different strategies using immune challenge‐based models and neurotoxin‐induced AD models have been used.24 In virtually all scenarios considered, animal models will be indispensable for elucidating the molecular and cellular mechanisms of AD pathogenesis, and for developing effective strategies to prevent and treat the disease.

Neuroinflammatory glial responses (relevance to the brain's microenvironment)

Microglia, the principal innate immune cells in the brain, carry out macrophage‐like phagocytic actions to remove pathogens and to protect neurons from toxic species. However, microglia produce and release molecules, such as reactive oxygen species and nitric oxide, that are neurotoxic.25 They also generate pro‐inflammatory cytokines and chemokines in response to danger signals, and communicate with astrocytes.26 To this end, the dysregulation of microglial activity has been associated with AD pathogenesis in the aged brain.27 Recently, it was reported that TREM2, as well as CD36 and the receptor for advanced glycation end‐products, work as Aβ sensor molecules and activate microglia.28 A consistent and elevated expression of TREM2 was described in App NL‐G‐F KI mice.18 Activated microglia also produce the pro‐inflammatory cytokines CCL3/MIP‐1α and interleukin (IL)‐6,28 the latter being a key component of the senescence‐associated secretory phenotype,29 which might provide a pathophysiological connection between cellular/tissue senescence and age‐related chronic diseases in the brain, including AD. Furthermore, pro‐inflammatory gene polymorphisms, including CCL3/MIP‐1α and IL‐6, have been identified as risk factors for AD.30

The genetic modulation of inflammation‐related factors has been investigated in various transgenic models of AD.31 These studies commonly showed that the modulation of inflammatory factors alters amyloid pathology and tau phosphorylation in the mouse models used. Interestingly, the inflammasome, a key inflammatory signaling platform in immune cells that activates IL‐1β and IL‐18 through NLRP3/ASC/Caspase1 activation,32 in microglia might contribute to AD pathogenesis in APP/PS1 mice.33, 34 To this end, microglia‐derived ASC (a constituent of the inflammasome) has been shown to regulate amyloidosis in APPswe/PSEN1dE9 mice.35 Modulation of glial function through the manipulation of cytokines/chemokines and their receptors has also been investigated.31, 36 These studies provided evidence that the microglial fractalkine receptor (CX3CR1) could potentially exacerbate tau pathology and neuronal cell death,37, 38 and that microglia also expand tau propagation through the exosome.27, 39 These findings support the notion that reactive microglial neuroinflammation accelerates AD pathogenesis, particularly by linking tau pathology with neurodegeneration.

Astrocytes serve multiple functions, including providing support to endothelial cells that form neurovascular units in the blood–brain barrier, supplying nutrients to the central nervous system, maintaining the extracellular balance of electrolytes and water, and repairing or remodeling tissue during the process of traumatic brain injury or neuroinflammation. As shown in Figure 1b, reactive astrocytes can be typically observed in the vicinity of amyloid plaque (plaque‐associated astrocytes). Although the primary function of the plaque‐associated astrocytic response remains unclear, deletion of the glial filament proteins glial fibrillary acid protein and vimentin in APP/PS1 mice increased the number of dystrophic neurites,40, 41 whereas astrocyte‐producing kallikrein‐related peptidase 7 contributed to Aβ degradation.42, 43 Astrocytes also contribute to the clearance of Aβ and other debris from the brain through astrocytic transport, the so‐called “glymphatic system”.44 As the glymphatic system is comparable with the lymphatic system in peripheral organs, astrocytes might act as a gateway from the brain to the blood vessels. Recently, phagocytic astrocytes were observed at ischemic sites of the brain.45 Astrocytic phagocytosis has been suggested to engulf and degrade plaque‐associated synaptic dystrophies in APP/PS‐1 mice and AD brain.46 Furthermore, neurotoxic A1 astrocytes are induced by activated microglia;47 to this end, a glucagon‐like peptide‐1 receptor agonist was postulated to act as a potential neuroprotective agent through the suppression of A1 astrocytes in a mouse model of Parkinson's disease.48 Although further classification of astrocyte cell types is required, plaque‐associated reactive astrocytes could protect neurons surrounding amyloid plaques in the early stages of AD pathogenesis. While these glial cells thus serve as “guardians” of the brain microenvironment, any dysregulation of glial communication could lead to a neurotoxic state.

The receptivity and responsiveness of glial cells are different with respect to amyloid plaques compared with NFT in the AD brain. Plaque‐associated astrocytes surround amyloid plaques, whereas microglia attack the inside of the plaques. However, microglia are unable to reach the inside of amyloid plaques in APP23 mouse brains due to the unphysiologically large size of these plaques (Fig. 2). Consequently, the microglial response in APP Tg mice is likely to be different from that in App KI mice and in the AD brain. The morphological pattern of reactive astrocytes and microglia in response to FSB‐positive NFT (ghost tangles) was different from that in response to Aβ deposition (Fig. 1b). Regulation of the glial response might therefore change markedly depending on the pathological stage of tauopathy.21, 49 However, it is still unclear which glial cells, astrocytes or microglia, recognize these abnormal protein aggregates as danger signals, and how this is achieved, especially considering that the communication between astrocytes and microglia is regulated by pathology‐associated cytokines/chemokines.

In summary of the above literature, the cytokines induced by amyloidosis are different from those induced by tauopathy in the different mouse models, meaning that it might be possible to distinguish amyloidosis‐associated glial responses (neuroinflammation) from tauopathy‐associated neuroinflammation, as shown in Figure 1a.18, 31, 36, 37, 38 Indeed, in a manner similar to that describing amyloidosis and tauopathy in the AD brain, glial pathology in amyloidosis mouse models is different from that in tauopathy mouse models. Furthermore, at the stage when both amyloid and tau pathologies exist, dysregulation of glial communication in the brain microenvironment along with the long‐lasting abnormal activation of glial cells might facilitate the pathogenesis of AD.

Neuroinflammation and the interaction between brain and peripheral tissue (whole‐body macroenvironment)

Evidence from epidemiological studies pointed to a possible link between the use of non‐steroidal anti‐inflammatory drugs (NSAIDs) and a decreased risk of AD in people with rheumatoid arthritis.50 Subsequently, the long‐term use of NSAIDs was found to potentially protect individuals against AD, but not against vascular dementia.51 A number of studies using different AD mouse models also suggested that NSAIDs improve Aβ‐mediated brain dysfunction. Although the protective mechanisms by which NSAIDs exert their effects remain unclear, inflammation in the brain and/or periphery could be involved in the pathogenesis of AD. In contrast to the epidemiological evidence, a recent meta‐analysis on the effects of NSAID treatment reported no beneficial effect on AD.52 These conflicting results suggest that NSAIDs do not improve AD pathogenesis directly in the brain, but that systemic inflammation, such as that seen with rheumatoid arthritis, might affect the brain pathologically. Recent evidence also suggests that inflammatory diseases, such as osteoporosis,53 diabetes,54 cancer55 and infection,56 are possibly implicated in brain disorders, and that these diseases could affect brain function through immune responses elicited in the periphery; that is, through neuroimmune communication. Furthermore, treatment strategies against such diseases might influence brain function and the macroenvironment, as reported for cancer‐related cognitive impairment57 or HIV‐associated neurocognitive disorder,58 possibly leading to the onset of brain disorders. Recently, accumulating evidence has suggested a role of peripheral immune cells, particularly CD4+ T cells, in the central nervous system. CD4+ T cells have been suggested to affect the activation of microglia, and to alter the Aβ burden in APP Tg mice and AD patients.59, 60, 61 Thus, future investigations need to clarify the role of both tissue resident immune cells and circulating immune cells in the pathogenesis of AD.

Interestingly, gut microbiota and associated metabolites have been described to influence brain dysfunction62, 63 and to modulate the host immune system.64 This “gut–brain axis” has consequently received attention in several research fields. While it was reported that factors, such as obesity,65 exercise,65 diet/nutrition,65 circadian rhythm,66 sleep,67 stress68 and aging,69 modulate gut microbiotic conditions, such factors might also affect brain function and brain disorders. The body's macroenvironment, particularly the role played by inflammatory factors and immune cells including microglia and astrocytes, almost certainly contributes to the correct physiological functioning of the brain, as well as to the pathogenesis of brain disorders when this environment is disturbed. Taken together, these observations shed new light on the notion that the pathogenesis of AD might be linked not only to conditions within the central nervous system, but also to peripheral conditions, thus making it a whole‐body disorder. To further investigate such whole‐body interactions, studies using relevant animal models and the in vivo imaging of neuroinflammation will be critical to understanding the mechanisms underlying AD and to predicting therapeutic outcomes.70

Conclusion

Numerous AD studies showing the time‐course of disease development and the complex nature of pathological processes in the brain attest to the difficulty of elucidating the molecular and cellular mechanisms underlying AD pathogenesis. To promote further investigation, animal models will be critical if progress is to be made. However, some AD mouse models do not accurately or reproducibly reflect AD in humans, and might thus need to be re‐evaluated as suitable models. As highlighted here, neuroinflammation is an important process in the pathogenesis of AD, and contributes a rate‐limiting component that potentially links Aβ amyloidosis with neurodegeneration and neuronal cell death through tauopathy. However, the pathological roles of neuroinflammation based on the brain microenvironment, as well as that contributed by the whole‐body macroenvironment, remain elusive. Understanding glial cell interactions associated with AD and other neurodegenerative disorders according to the newly coined term “gliostasis” (homeostasis of glial cells) might serve as a promising starting point. Relevant animal models will again be necessary to initiate such studies, and will serve as a fundamental research tool to elucidate the pathogenetic mechanisms underlying AD development and to develop preventive or therapeutic interventions to combat the disease.

Conflict of interest

T.S. and T.C.S. serve as the advisor and CEO, respectively, for RIKEN BIO Co. Ltd., which sublicenses animal models to for‐profit organizations, the profits from which are used for the identification of disease biomarkers.

Acknowledgments

We thank Naomi Mihira and Yukio Matsuba for technical assistance, and Yukiko Watanabe‐Nagai for secretarial assistance. This work was partially supported by AMED, under Grant Number JP18dm0107070h0003 (the Strategic Research Program for Brain Sciences; T.S.) and JP18dm027001 (Brain Mapping by Integrated Neurotechnologies for Disease Studies [Brain/MINDS] T.C.S.); a grant‐in‐aid for scientific research (B) (grant number: 26290019) of the Ministry of Education, Culture, Sports, Science and Technology (T.S.); JST PRESTO (T.S); the Cell Science Research Foundation (T.S.); the Aging Project of RIKEN (T.C.S.); and research grants from RIKEN Center for Brain Science (T.C.S.). We also thank Michael Patterson, Sciencedit, for editing the English in our manuscript.

Contributor Information

Takashi Saito, Email: takashi.saito.aa@riken.jp.

Takaomi C. Saido, Email: takaomi.saido@riken.jp.

References

  • 1. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med. 2016; 8: 595–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Bateman RJ, Xiong C, Benzinger TLS, et al. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N Engl J Med. 2012; 367: 795–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Maeda J, Zhang MR, Okauchi T, et al. 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: 4720–4730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Serrano‐Pozo A, Mielke ML, Gómez‐Iska T, et al. Reactive glia not only associated with plaques by also parallels tangles in Alzheimer's disease. Am J Pathol. 2011; 179: 1373–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Drummond E, Wisniewski T. Alzheimer's disease: experimental models and reality. Acta Neuropathol. 2017; 133: 155–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Benzing WC, Wujek JR, Ward EK, et al. Evidence for glial‐mediated inflammation in aged APP(SW) transgenic mice. Neurobiol Aging. 1999; 20: 581–589. [DOI] [PubMed] [Google Scholar]
  • 7. Bornemann KD, Wiederhold KH, Pauli C, et al. Abeta‐induced inflammatory processes in microglia cells of APP23 transgenic mice. Am J Pathol. 2001; 158: 63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Saito T, Suemoto T, Brouwers N, et al. Potent amyloidogenicity and pathogenicity of Aβ43. Nat Neurosci. 2011; 14: 1023–1032. [DOI] [PubMed] [Google Scholar]
  • 9. Saito T, Matsuba Y, Mihira N, et al. Single App knock‐in mouse models of Alzheimer's disease. Nat Neurosci. 2014; 17: 661–663. [DOI] [PubMed] [Google Scholar]
  • 10. Saito T. Chronic neuroinflammation underlying pathogenesis of Alzheimer's disease. Chron Inflamm. 2016; Chapter 50: 661–671. [Google Scholar]
  • 11. Sasaguri H, Nilsson P, Hashimoto K, et al. APP mouse models for Alzheimer's disease preclinical studies. EMBO J. 2017; 36: 2473–2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Saito T, Matsuba Y, Yamazaki N, et al. Calpain activation in Alzheimer's model mice is an artifact of APP and Presenilin overexpression. J Neurosci. 2016; 36: 9933–9936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Hashimoto S, Ishii A, Kamano N, et al. Endoplasmic reticulum stress responses in mouse models of Alzheimer's disease: Overexpression paradigm versus knockin paradigm. J Biol Chem. 2018; 293: 3118–3125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Hama H, Hioki H, Namiki K, et al. ScaleS: an optical clearing palette for biological imaging. Nat Neurosci. 2015; 10: 1518–1529. [DOI] [PubMed] [Google Scholar]
  • 15. Zhang H, Wu L, Pchitskaya E, et al. Neuronal store‐operated calcium entry and mushroom spine loss in amyloid precursor protein knock‐in mouse model of Alzheimer's disease. J Neurosci. 2015; 35: 113275–113286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Rajendran L, Paolicelli RC. Microglia‐mediated synapse loss in Alzheimer's disease. J Neurosci. 2018; 38: 2911–2919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Oddo S, Caccamo A, Kitazawa M, et al. Triple‐transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron. 2003; 39: 409–421. [DOI] [PubMed] [Google Scholar]
  • 18. Castillo E, Leon J, Mazzei G, et al. Comparative profiling of cortical gene expression in Alzheimer's disease patients and mouse models demonstrates a link between amyloidosis and neuroinflammation. Sci Rep. 2017; 7: 17762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Jankowsky JL, Zheng H. Practical considerations for choosing a mouse model of Alzheimer's disease. Mol Neurodegener. 2017; 12: 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Ghetti B, Oblak AL, Boeve BF, et al. Invited review: frontotemporal dementia caused by microtubule‐associated protein tau gene (MAPT) mutations: a chameleon for neuropathology and neuroimaging. Neuropathol Appl Neurobiol. 2015; 41: 24–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sahara N, Maeda J, Ishikawa A, et al. Microglial activation during pathogenesis of tauopathy in rTg4510 mice: implications for the early diagnosis of tauopathy. J Alzheimers Dis. 2018; 64(s1): S353–S359. [DOI] [PubMed] [Google Scholar]
  • 22. Yoshiyama Y, Higuchi M, Zhang B, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy model mouse. Neuron. 2007; 53: 337–351. [DOI] [PubMed] [Google Scholar]
  • 23. Lewis J, Dickson DW, Lin WL, et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science. 2001; 293: 1487–1491. [DOI] [PubMed] [Google Scholar]
  • 24. Nazem A, Sankowski R, Bacher M, et al. Rodent models of neuroinflammation for Alzheimer's disease. J Neuroinflammation. 2015; 12: 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Block ML, Zecca L, Hong JS, et al. Microglia‐mediated neurotoxicity: uncovering the molecular mechasims. Nat Rev Neurosci. 2007; 8: 57–69. [DOI] [PubMed] [Google Scholar]
  • 26. Burda JE, Sofroniew M. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014; 81: 229–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Clayton KA, Van Enoo AA, Ikezu T. Alzheimer's disease: the role of microglia in brain homeostasis and proteopathy. Front Neurosci. 2017; 11: 680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zhao Y, Wu X, Li X, et al. TREM2 is a receptor for β‐amyloid that mediates microglial function. Neuron. 2018; 97: 1023–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Zhu Y, Armstrong JL, Tchkonia T, et al. Cellular senescence and the senescent secretory phenotype in age‐related chronic diseases. Curr Opin Clin Nutr Metab Care. 2014; 17: 324–328. [DOI] [PubMed] [Google Scholar]
  • 30. Flex A, Giovannini S, Biscetti F, et al. Effect of proinflammatory gene polymorphisms on the risk of Alzheimer's disease. Neurodegener Dis. 2014; 13: 230–236. [DOI] [PubMed] [Google Scholar]
  • 31. Birch AM, Katsouri L, Sastre M. Modulation of inflammation in transgenic models of Alzheimer's disease. J Neuroinflam. 2014; 11: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev. 2013; 13: 397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013; 493: 674–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Liu L, Chan C. The role of inflammasome in Alzheimer's disease. Ageing Res Rev. 2014; 15: 6–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Venegas C, Kumar S, Franklin BS, et al. Microglia‐derived ASC specks cross‐seed amyloid‐β in Alzheimer's disease. Nature. 2017; 552: 355–361. [DOI] [PubMed] [Google Scholar]
  • 36. Liu C, Cui G, Zhu M, et al. Neuroinflammation in Alzheimer's disease: chemokines produced by astrocytes and chemokine receptors. Int J Clin Exp Pathol. 2014; 7: 8342–8355. [PMC free article] [PubMed] [Google Scholar]
  • 37. Fuhrmann M, Bittner T, Jung CKE, et al. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer's disease. Nat Neurosci. 2010; 13: 411–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Maphis N, Xu G, Kokiko‐Cochran ON, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015; 138: 1738–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015; 18: 1584–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Serrano‐Pozo A, Muzikansky A, Gómez‐Iska T, et al. Differential relationships of reactive astrocytes and microglia to fibrillar amyloid deposits in Alzheimer disease. J Neuropathol Exp Neurol. 2013; 72: 462–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kraft AW, Hu X, Yoon H, et al. Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice. FASEB J. 2013; 27: 187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kidana K, Tatebe T, Ito K, et al. Loss of kallikrein‐related peptidase 7 exacerbates amyloid pathology in Alzheimer's disease. EMBO Mol Med. 2018; 10: e8184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Ries M, Sastre M. Mechanisms of Aβ clearance and degradation by glial cells. Front Aging Neurosci. 2016; 8: 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Benveniste H, Liu X, Koundal S, et al. The glymphatic system and waste clearance with brain aging: a review. Gerontology. 2018; 11: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Morizawa YM, Hirayama Y, Ohno N, et al. Reactive astrocytes function as phagocytes after brain ischemia via ABCA1‐mediated pathway. Nat Commun. 2017; 8: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Gomez‐Arboledas A, Davila JC, Sanchez‐Mejias E, et al. Phagocytic clearance of presynaptic dystrophies by reactive astrocytes in Alzheimer's disease. Glia. 2018; 66: 637–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Liddelow SA, Guttenplan KA, Clarke LE, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017; 541: 481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Yun SP, Kam TI, Panicker N, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease. Nat Med. 2018; 24: 931–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Laurent C, Buée L, Blum D. Tau and neuroinflammation: what impact for Alzheimer's disease and tauopathies? Biomed J. 2018; 41: 21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. McGeer PL, Schulzer M, McGeer EG. Arthritis and anti‐inflammatory agents as possible protective factors for Alzheimer's disease: a review of 17 epidemiologic studies. Neurology. 1996; 47: 425–432. [DOI] [PubMed] [Google Scholar]
  • 51. in't Veld BA, Ruitenberg A, Hofman A, et al. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer's disease. N Engl J Med. 2001; 345: 1515–1521. [DOI] [PubMed] [Google Scholar]
  • 52. Miguel‐Álvarez M, Santos‐Lozano A, Sanchis‐Gomar F, et al. Non‐steroidal anti‐inflammatory drugs as a treatment for Alzheimer's disease: a systematic review and meta‐analysis of treatment effect. Drugs Aging. 2015; 32: 139–147. [DOI] [PubMed] [Google Scholar]
  • 53. Chen YH, Lo RY. Alzheimer's disease and osteoporosis. Ci Ji Yi Xue Za Zhi. 2017; 29: 138–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. de Nazareth AM. Type 2 diabetes mellitus in the pathophysiology of Alzheimer's disease. Dement Neuropsychol. 2017; 11: 105–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Guo J, Cheng J, North BJ, Wei W. Functional analyses of major cancer‐related signaling pathways in Alzheimer's disease etiology. Biochim Biophys Acta. 2017; 1868: 341–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Sochocka M, Zwolińska K, Leszek J. The infectious etiology of Alzheimer's disease. Curr Neuropharmacol. 2017; 15: 996–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Horowitz TS, Suls J, Treviño M. A call for a neuroscience approach to cancer‐related cognitive impairment. Trends Neurosci. 2018; 41: 493–496. [DOI] [PubMed] [Google Scholar]
  • 58. Sanmarti M, Ibáñez L, Huertas S. HIV‐associated neurocognitive disorders. J. Mol Psychiatry. 2014; 2: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Fisher Y, Nemirovsky A, Baron R, et al. T cells specifically targeted to amyloid plaques enhance plaque clearance in a mouse model of Alzheimer's disease. PLoS ONE. 2010; 5: e10830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Browne TC, McQuillan K, McManus RM, et al. IFN‐γ production by amyloid β‐specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer's disease. J Immunol. 2013; 190: 2241–2251. [DOI] [PubMed] [Google Scholar]
  • 61. Goldeck D, Larbi A, Pellicanó M, et al. Enhanced chemokine receptor expression on leukocytes of patients with Alzheimer's disease. PLoS ONE. 2013; 8: e66664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Hsiao EY, McBride SW, Hsien S, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013; 155: 1451–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015; 125: 926–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a rationally selected mixture of chostridia strains from the human microbiota. Nature. 2013; 500: 232–236. [DOI] [PubMed] [Google Scholar]
  • 65. Solas M, Milagro FI, Ramírez MJ. Inflammation and gut‐brain axis link obesity to cognitive dysfunction: plausible pharmacological interventions. Curr Opin Pharmacol. 2017; 37: 87–92. [DOI] [PubMed] [Google Scholar]
  • 66. Deaver JA, Eum SY, Toborek M. Circadian disruption changes gut microbiome taxa and functional gene composition. Front Microbiol. 2018; 9: 737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Krueger JM, Opp MR. Sleep and microbes. Int Rev Neurobiol. 2016; 131: 207–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Konturek PC, Brzozowski T, Konturek SJ. Stress and the gut: pathophysiology, clinical consequences, diagnostic approach and treatment options. J Physiol Pharmacol. 2011; 62: 591–599. [PubMed] [Google Scholar]
  • 69. Choi J, Hur TY, Hong Y. Influence of altered gut microbiota composition on aging and aging‐related diseases. J Lifestyle Med. 2018; 8: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Higuchi M, Ji B, Maeda J, et al. In vivo imaging of neuroinflammation in Alzheimer's disease. Clin Exp Neuroimmunol. 2016; 7: 139–144. [Google Scholar]

Articles from Clinical & Experimental Neuroimmunology are provided here courtesy of Wiley

RESOURCES