Abstract
Microglia are the predominant immune cells of the central nervous system (CNS) that exert key physiological roles required for maintaining CNS homeostasis, notably in response to chronic stress, as well as mediating synaptic plasticity, learning and memory. The repeated exposure to stress confers a higher risk of developing neurodegenerative diseases including sporadic Alzheimer's disease (AD). While microglia have been causally linked to amyloid beta (Aβ) accumulation, tau pathology, neurodegeneration, and synaptic loss in AD, they were also attributed beneficial roles, notably in the phagocytic elimination of Aβ. In this review, we discuss the interactions between chronic stress and AD pathology, overview the roles played by microglia in AD, especially focusing on chronic stress as an environmental risk factor modulating their function, and present recently-described microglial phenotypes associated with neuroprotection in AD. These microglial phenotypes observed under both chronic stress and AD pathology may provide novel opportunities for the development of better-targeted therapeutic interventions.
Keywords: Microglia, Stress, Alzheimer's disease, Neurodegeneration, Neuroinflammation, Synaptic remodeling, Microglial phenotypes, Dark microglia
Abbreviations: Aβ, Amyloid beta; ABCA7, ATP-binding cassette transporter A7; AD, Alzheimer's disease; APOE, Apolipoprotein E; APP, amyloid precursor protein; BDNF, brain derived neurotrophic factor; CD11b, cluster of differentiation molecule 11B; CD33, cluster of differentiation 33; CNS, central nervous system; CR, complement receptor; CRF, corticotropin releasing factor; DAM, disease associated microglia; DAP12, DNAX-activation protein 12; FAD, Familial Alzheimer's disease; FCRLS, Fc receptor-like S scavenger receptor; GR, glucocorticoid receptor; HPA axis, hypothalamic pituitary adrenocortical axis; IBA1, ionized calcium-binding adapter molecule 1; IL, interleukin; LTP, long-term potentiation; MGnD, microglia with a neurodegenerative phenotype; mPFC, medial prefrontal cortex; MR, mineralocorticoid receptor; NADPH, nicotinamide adenine dinucleotide phosphate; NFT, neurofibrillary tangles; PS, presenilin; ROS, reactive oxygen species; TGFβ, transforming growth factor β; TLR, Toll-like receptors; TMEM119, transmembrane protein 119; TNFα, tumor necrosis factor-α; TREM2, triggering receptor expressed in myeloid cells 2; TYROBP, TYRO protein tyrosine kinase binding protein
1. Introduction: microglia and Alzheimer's disease
Microglia are known to rapidly respond to alterations in brain homeostasis during stress, trauma, disease or pathology (Matcovitch-Natan et al., 2016; Tian et al., 2017). They are the resident, predominant immune cells of the CNS, which although initially thought to be responsible for orchestrating neuroinflammation and causing neurodegeneration (Cartier et al., 2014), were also recently attributed important roles in the maintenance of CNS homeostasis and remodeling of neuronal circuits across development and experience-dependent plasticity (Derecki et al., 2014; Ransohoff and El Khoury, 2015; Tay et al., 2017). Microglial ability to sense neuronal activity allows them to regulate synaptic plasticity, learning and memory mechanisms, and hence determine cognitive abilities (Morris et al., 2013; Sipe et al., 2016). For instance, brain derived neurotrophic factor (BDNF) produced by microglia was shown to modulate motor learning-dependent synapse formation in mice (Parkhurst et al., 2013). In addition, CX3CR1CreER/ROSA26iDTR-flox mice treated with tamoxifen to induce expression of the diphtheria toxin receptor selectively in microglia, and receiving diphtheria toxin to deplete these cells, showed decreased motor learning-induced dendritic spine formation, and reduced levels of synaptic proteins VGlut1 and GluA2 from glutamatergic synapses in the motor cortex (Parkhurst et al., 2013).
Reactive ‘microgliosis’ in terms of abnormal morphology (e.g. a shift from a ramified to an amoeboid morphology), exacerbated immunoreactivity for ionized calcium-binding adapter molecule 1 (IBA1), and increased proliferation is a prominent feature of AD pathology, especially observed around amyloid plaques in postmortem brain tissue. Similarly, positron emission tomography scans of AD patients using the TSPO tracer [11C]-PK11195 revealed exacerbated glial activity (Edison et al., 2008; Perlmutter et al., 1992; Rozemuller et al., 1992; Serrano-Pozo et al., 2011b). Investigating microglial implication in AD onset and progression is becoming increasingly important given the various roles that these immune cells can play. While microglia were causally linked to neuroinflammation, Aβ accumulation (see Box 1), and tau pathology, as well as implicated in neuronal and synaptic loss (Fuhrmann et al., 2010; Morris et al., 2013), they were also shown to actively participate in the phagocytic clearance of Aβ and protection of neuronal tissue (Ard et al., 1996; Grathwohl et al., 2009; Majumdar et al., 2011; Maphis et al., 2015).
AD is a debilitating, progressive neurodegenerative disease and the most common cause of dementia worldwide (van der Flier and Scheltens, 2005). It is characterized by a slow, progressive cognitive dysfunction, memory decline, and gradual disturbances in reasoning, language, and physical abilities (Holtzman et al., 2011). In AD, the presence of extracellular Aβ deposits and intracellular neurofibrillary tangles (NFTs) are main hallmarks that lead to neuronal dysfunction, cell death, and loss of synaptic connections, notably due to ensuing inflammation and oxidative stress (Zhao and Zhao, 2013). The imbalance in Aβ homeostasis, i.e. production versus clearance of Aβ peptides, leads to their accumulation, aggregation –in either fibrillary, oligomeric, or β-sheet conformations– and deposition within the extracellular space (Sadigh-Eteghad et al., 2015). The extracellular Aβ deposits trigger active ‘gliosis’ that primarily consists in hyperactive astrocytes and microglia among the surrounding parenchyma (Hardy and Higgins, 1992; Kurz and Perneczky, 2011; Wildsmith et al., 2013). The hyperphosphorylation of tau –a tubulin binding protein that functions to stabilize microtubules in the neurons (Kadavath et al., 2015)– results in its dissociation from microtubules, and aggregation in the form of NFTs inside of neuronal cell bodies and neurites, which also triggers gliosis (Grundke-Iqbal et al., 1986; Kadavath et al., 2015; Serrano-Pozo et al., 2011a). Synaptic and neuronal loss as a result of tau pathology and Aβ deposition have been shown to best correlate with the cognitive impairment seen in AD (Spires-Jones and Hyman, 2014). These pathological features may appear early in subcortical nuclei such as the locus coeruleus, then affect the hippocampus and entorhinal cortex, and eventually spread to other cortical areas as the disease progresses (Braak and Del Tredici, 2011; Glenner and Wong, 1984; Grundke-Iqbal et al., 1986; Hardy and Higgins, 1992; Holtzman et al., 2011; Raber et al., 2004).
AD is a multifactorial disease driven by a combination of genetic and environmental factors, and can therefore be classified into familial (FAD) and sporadic forms (Johns, 2014). FAD comprising about 5% of AD cases is due to mutations in APP, presenilin-1, or presenilin-2 genes that result in the production of aggregation-prone Aβ peptides (Johns, 2014; Lanoiselée et al., 2017). Sporadic forms have no familial history of AD, but may be related to the expression of apolipoprotein E4 (APOE4) involved with the transport of lipids, cholesterol, and other hydrophobic molecules into the brain (Raber et al., 2004). In addition, environmental factors that comprise psychosocial stress confer a high risk of developing neurodegenerative diseases and may play an important role in the etiology of sporadic AD (Alkadhi, 2012; Aznar and Knudsen, 2011; Nation et al., 2011). Genetic risk factors for sporadic AD also include mutations or polymorphisms in several genes expressed by microglia and other myeloid cells (Dos Santos et al., 2017; Efthymiou and Goate, 2017; Jonsson et al., 2013; Malik et al., 2015; Satoh et al., 2017; Sims et al., 2017), stressing the importance of these immune cells. While the brain is home to perivascular macrophages, as well as monocytes derived from the bone marrow that enter the CNS from the circulation, especially during brain injury or systemic inflammation (Guillemin and Brew, 2004; Schwartz et al., 2013; Wohleb et al., 2015), microglia are the predominant immune cell population within the CNS.
The genes associated with sporadic AD include TREM2 (Guerreiro et al., 2013), which codes for a cell surface protein (Prokop et al., 2015; Sessa et al., 2004). The particular R47H variant of TREM2 leads to a loss-of-function that impairs TREM2-mediated neuroprotection and increases the risk of developing AD by threefold (Guerreiro et al., 2013; Jonsson et al., 2013; Melchior et al., 2010). Knockdown of TREM2 in the brain of a senescence-accelerated mouse prone 8 (SAMP8) mouse model resulted in cognitive impairment, accompanied by increased expression levels of pro-inflammatory tumor necrosis factor (TNF)α and interleukin (IL)-6, and decreased expression levels of anti-inflammatory IL-10 (Jiang et al., 2014). The association of TREM2 with its binding partner DAP12, an activating adaptor protein, is important for microglial survival and proliferation in mice, especially through regulation of the Wnt/β-Catenin pathway (Poliani et al., 2015; Zheng et al., 2017). TREM2/DAP12 activity is essential for microglial innate immune responses, including chemotaxis and phagocytosis (Kleinberger et al., 2014; Takahashi et al., 2005; Zhong et al., 2017). TREM2 knockdown using a lentiviral strategy reduced microglial ‘efferocytosis’ or phagocytosis of apoptotic neurons, while its overexpression increased microglial phagocytic activity in a mouse neuron-microglia co-culture system (Takahashi et al., 2005).
Other genes expressed by microglia that were implicated in sporadic AD include the ABCA7 gene and its variants rs3764650, rs4147929, and rs3752246 (Cuyvers et al., 2015). The gene encodes a protein thought to regulate lipid homeostasis (as well as Aβ homeostasis possibly) and mediate the formation of phagocytic cups, critical for microglial phagocytosis of apoptotic cells and Aβ (Abe-Dohmae et al., 2004; Jehle et al., 2006; Kim et al., 2013). Increased ABCA7 levels were measured in AD postmortem brains, and also in an AD mouse model knockout for ABCA7 (J20/Abca7−/− mice) showing significantly increased insoluble Aβ levels and plaque burden in the brain, thus suggesting that ABCA7 exerts a neuroprotective role in AD (Kim et al., 2013; Li et al., 2015).
Through this review, we aim to discuss the consequences of chronic stress on the onset and progression of AD pathology, overview the roles played by microglia in AD, by focusing on chronic stress as an environmental risk factor modulating their function, and present the recently-described microglial phenotypes associated with neuroprotection in AD. These microglial phenotypes observed upon chronic stress and AD pathology may provide novel opportunities for the development of therapeutic applications across different contexts of neurodegeneration.
2. Chronic stress as an environmental risk factor
Stress is our body's natural response to adverse or demanding changes in our environment, developed as a measure to deal and overcome these challenges to our well-being (McEwen, 2005). Allostasis refers to such internal adaptive mechanisms that attempt to restore homeostasis to meet the perceived and anticipated demands posed by the stressful situations (McEwen and Seeman, 1999). Stress, therefore, is crucial for survival, but its response can become maladaptive (Ellis and Del Giudice, 2014). Depending on the actual stressor and the severity of its effects, stress can be either beneficial or detrimental, and the duration and chronicity of the stressors exposure also plays an important role in determining its outcome (McCormick and Hodges, 2017).
While the body can quickly resolve and normalize the effects of acute stress, when it becomes chronic, stress may seriously perturb the physiological and psychological balance of an individual. Repetitive recruitment of the neuroendocrine response to chronic stress can lead to a cumulative disturbance of the internal homeostatic mechanisms. Chronic unpredictable and uncontrollable stressors are generally associated with a high risk of developing cardiovascular diseases, depression, and neurodegenerative disorders that include AD, especially in individuals susceptible to stress and its detrimental effects (Leonard, 2010; Liu et al., 2017).
In individuals that are susceptible to stress, due to the high allostatic load, the once protective coping mechanisms become pathological, whereas in resilient individuals, the adaptive response is successfully used to maintain homeostasis (McEwen, 2000; McEwen and Wingfield, 2007). The biological mechanisms that determine the individual response to stress is the subject of intense research with the aim of promoting stress resilience, given the severe impact that chronic stress has on human health and the serious burden that it imposes on individuals as well as to the society (Schneiderman et al., 2005; Seib et al., 2014).
Stress is also a consequence of disease, especially when the normal day-to-day activities of the affected individuals are being significantly disrupted as a result of their underlying medical condition, thus causing psychological stress (Donovan et al., 2018). The resulting psychological stress may lead to exacerbated cellular stress due to inflammation and oxidative damage (Hayashi, 2015; Miller and Sadeh, 2014). The molecular mechanisms by which stress leads to altered homeostasis and pathology will be discussed further in this review.
A key physiological system that responds to psychological stress is the hypothalamic-pituitary-adrenocortical (HPA) axis, which comprises a tightly regulated pathway of communication between the paraventricular nucleus of the hypothalamus, the pituitary gland, and the adrenal gland. Stressors exposure results in HPA axis activation that eventually leads to the secretion into the blood stream of glucocorticoid stress hormones: cortisol in humans or corticosterone in rodents (Smith and Vale, 2006). Blood glucocorticoids cross the blood-brain barrier and enter the brain to activate glucocorticoid receptors (GR) and mineralocorticoid receptors (MR), two important signaling pathways having essential roles in mediating the brain response to stress (Scheuer, 2010). While GR is ubiquitously expressed by neurons, microglia, CNS-infiltrating monocytes, astrocytes, and oligodendrocytes, MR expression is restricted to mainly neurons showing limited expression by microglia and astrocytes (Madalena and Lerch, 2017). MR is active in basal conditions, due to its high binding affinity and sensitivity toward the low resting levels of corticosteroids (de Kloet et al., 2000). It mediates the tonic functions of glucocorticoids, including their circadian regulation of learning and memory (de Kloet et al., 2000). GR is mainly active during stressful conditions, due to its low binding affinity for corticosteroids, allowing its response to high corticosterone/cortisol levels (de Kloet et al., 2000; Groeneweg et al., 2012). GR and MR also differ in their relative distribution across the brain; while MR is restricted to the prefrontal cortex (PFC), amygdala, and hippocampus, GR shows an abundant brain-wide expression especially in the PFC, amygdala, hypothalamic paraventricular nucleus, locus coeruleus, hippocampus, and hindbrain (Fuxe et al., 1987; Meaney et al., 1985; Reul and de Kloet, 1986, 1985).
Glucocorticoid signaling through GR and MR ensures a tight control over the HPA axis during an acute stress response, requiring an integrative and coordinated activity of neuroendocrine circuits among the forebrain, hypothalamic, and hindbrain systems in order to mediate adaptive changes at both the physiological and behavioral levels (Myers et al., 2014). However, in conditions of prolonged psychological stress, when the adaptive changes are no longer sufficient to meet the environmental challenges, the HPA axis is chronically activated due to a loss of inhibitory feedback causing a dysregulation of glucocorticoid signaling (Ellis and Del Giudice, 2014; Nicolaides et al., 2015; O'Connor et al., 2017, 2016; Vyas et al., 2016). For instance, rats receiving chronic subcutaneous administration of corticosterone showed a permanent depletion of hippocampal corticosterone receptors, accompanied by loss of hippocampal neurons, similar to that seen during aging (Sapolsky et al., 1985).
The sustained increase in glucocorticoid levels was also proposed to initiate Aβ accumulation and tau pathology in AD (Dong and Csernansky, 2009; Green et al., 2006). The glucocorticoid cascade hypothesis postulates that HPA axis dysfunction may represent a contributing factor in the pathogenesis of AD and other neurodegenerative diseases, but strong clinical evidence is currently lacking (Bao et al., 2008; Franceschi et al., 1991; Swanwick et al., 1998). In support of this hypothesis, AD patients present a ∼83% increase in cortisol levels in their cerebrospinal fluid compared to healthy age-matched controls, thus indicating an association between stress and AD (Sapolsky et al., 1985; Swaab et al., 1994; Woolley et al., 1990). Several other human studies revealed that individuals with a life-long history of stress or trauma are at higher risk of developing brain atrophy, dementia, and AD in late-life stages (Johansson, 2014; Johansson et al., 2012, 2010; Wang et al., 2012). These evidence indicate that chronic stressors confer an increased vulnerability to developing neurodegenerative diseases across the lifespan.
Similarly, chronic stress was shown to ‘prime’ or sensitize microglia to generate heightened, exaggerated responses to a second stimulus occurring later in life (Santos et al., 2016). Studies by our group and others have detailed the effects of chronic stress on microglia using a number of paradigms in rodents, including maternal separation, physical restraint, tail suspension, forced swim or water immersion, social defeat or isolation, and unpredictable modifications of the housing conditions (Calcia et al., 2016; Lehmann et al., 2016; Roque et al., 2016; Sugama et al., 2011, 2007; Wang et al., 2017; Winkler et al., 2017; Wohleb et al., 2018). Overall, these studies revealed that microglia are integral partners in mediating the response of the brain and behavior to stress. In the absence of proper neuron-microglial crosstalk (as observed in Cx3cr1 knockout mice, which are deficient in fractalkine signaling) (see Box 1), chronic unpredictable stress failed to alter microglial morphology and phagocytic activity, as well as short- and long-term synaptic plasticity, including long-term potentiation (LTP, a cellular mechanism proposed to underlie memory formation) (Milior et al., 2016). Cx3cr1 knockout mice also failed to develop anhedonia, which is a measure of depressive-like behavior (Milior et al., 2016). Similar findings of an increased resistance to stress in the Cx3cr1 knockout mice were reported by other groups using different stress paradigms (Hellwig et al., 2016; Rimmerman et al., 2017; Winkler et al., 2017). In addition, microglial depletion, using systemic administration of a colony stimulating factor 1 receptor antagonist, in mice exposed to repeated social defeat stress, prevented anxiety (McKim et al., 2017). Similarly, treatment of rats with minocycline, a well-known modulator of microglial activity, rescued working memory upon exposure to chronic restraint stress (Hinwood et al., 2012). These findings and those from other studies indicate that microglial response to stress may be critical in determining its outcome on resilience or susceptibility, with consequences on the overall brain homeostasis.
3. Consequences of chronic stress on AD pathology
A complex interaction of genetic and environmental factors has been proposed to modulate the etiology and pathogenesis of AD. Meta-analysis studies conducted by Ownby et al. provided strong correlations between individuals’ histories of early-life depression and their risk of developing AD later in life (Ownby et al., 2006).
In animal studies, the exposure of male and female 5xFAD mice (in pre-pathological stages; see Fig. 1 for additional information on the AD pathology mouse models mentioned in this review) to chronic restraint stress resulted in increased neurotoxic Aβ42 (see Box 1) levels and plaque deposition in the hippocampus of females only, without changes in the cerebral cortex of both males and females (Devi et al., 2010). This finding suggests a sex- and brain region-specific vulnerability to the detrimental effects of stress. The elevation of blood glucocorticoid levels upon stress could adversely influence Aβ processing, by targeting the amyloid precursor and amyloid precursor-like proteins as shown in rats in vivo (Budas et al., 1999). Similarly, exposure of APP-PS1 mice (see Fig. 1) to chronic isolation stress resulted in increased Aβ42/Aβ40 ratios (see Box 1) in the hippocampus while worsening spatial working memory (Huang et al., 2011). Chronic immobilization stress to APPV717I-CT100 mice (overexpressing human APP-CT100 bearing the London mutation (V717I), see Fig. 1) also resulted in impaired performance under the passive avoidance task, increased extracellular Aβ deposits, elevated tau phosphorylation levels, and increased Aβ and APP carboxyl-terminal fragments (APP-CTFs; see Box 1) in the hippocampus, entorhinal, and piriform cortex (Jeong et al., 2006). Also, 3xTg mice (see Fig. 1) subjected to social stress showed increased plasma corticosterone levels accompanied by heightened anxiety compared to age-matched wild-types (Rothman et al., 2012). The 3xTg mice additionally displayed increased levels of Aβ oligomers, intraneuronal Aβ, and decreased levels of BDNF in the hippocampus, but no change in hyperphosphorylated tau (Rothman et al., 2012). Tg2576 mice showed increased Aβ levels and reduced IBA1 immunoreactivity, as well as deficits in spatial and fear memories, while the PS19 mice had elevated levels of hyperphosphorylated tau in the form of insoluble aggregates, with an impairment of fear memory upon chronic restraint and isolation stress (Carroll et al., 2011). These detrimental effects were prevented by pre-stress treatment with an antagonist of the corticotropin releasing factor (CRF) (Carroll et al., 2011). Mice overexpressing CRF (without bearing any mutations associated with AD) similarly displayed increased levels of hyperphosphorylated tau in this study (Carroll et al., 2011) and others (Campbell et al., 2015), supporting the detrimental consequences of chronic stress-mediated HPA axis dysregulation on AD pathogenesis.
Fig. 1.
Overview of the AD pathology mouse models mentioned in the review. Data compiled from Alzforum Alzheimer's disease research model database (https://www.alzforum.org/research-models/alzheimers-disease).
Aside from affecting the hallmark features of AD, the exposure of rodents to stress resulted in impaired adult neurogenesis and synaptic plasticity. Tg2576 mice (see Fig. 1) undergoing chronic isolation stress showed an accelerated age-dependent deposition of Aβ, accompanied by reduced hippocampal neurogenesis and decreased contextual memory, compared with their unstressed littermates (Dong et al., 2004). Tran et al. revealed significant alterations of LTP and spatial memory in rats chronically receiving subpathogenic doses of Aβ (subAβ) that were also exposed to repeated social defeat stress. Unstressed rats receiving subAβ did not differ from the controls receiving vehicle (Tran et al., 2011). This effect of social stress on LTP and spatial memory may partly result from the decreased expression levels of different plasticity-related proteins, including calcium calmodulin kinase II and protein kinase C, and increased expression levels of protein phosphatase 2B (calcineurin), which are known to inhibit LTP in the rat hippocampus (Gerges et al., 2004). Cytokines secreted by microglia such as anti-inflammatory IL-10 and TGFβ, and pro-inflammatory TNFα, IL-1β, IL-6, IL-12 and IL-18, could also be implicated (Piirainen et al., 2017). For example, IL-1 regulates learning and memory mechanisms notably through its influence on LTP. The induction of LTP promotes IL-1 expression while blocking IL-1 impairs maintenance of LTP in the rat hippocampus (Balschun et al., 2003; Schneider et al., 1998). Similarly, IL-18 positively regulates LTP maintenance, while IL-6 negatively regulates LTP maintenance in the rat hippocampus (del Rey et al., 2013). The contribution of inflammatory microglia to AD pathology (Sarlus and Heneka, 2017) will be discussed below.
Chronic stress additionally regulates the synthesis and secretion of neurotrophins that play key roles in synaptic plasticity, learning and memory. A decline in BDNF expression was measured in the hippocampus of wild-type rats in response to chronic restraint stress (Chiba et al., 2012; Smith et al., 1995) and of rodent depression models upon predator stress or repeated social defeat (Roth et al., 2011; Tsankova et al., 2006). A similar decline in blood BDNF levels was detected in patients suffering from depression or showing predisposition to depression, which could be reversed by antidepressant treatment (Karege et al., 2005; Sen et al., 2008; Shimizu et al., 2003; Trajkovska et al., 2008). A reduction in BDNF levels was also observed in AD patients at preclinical and clinical stages, within the hippocampus and temporal cortex, where the diminished BDNF levels correlated with the pathology severity (Connor et al., 1997; Michalski and Fahnestock, 2003; Peng et al., 2005; Phillips et al., 1991). These data suggest that alterations in neurotrophin signaling, particularly of BDNF, may underlie some of the deleterious effects of chronic stress, and hence influence AD onset and progression. BDNF expression is among other mechanisms regulated by glucocorticoid signaling onto both GR and MR (Suri and Vaidya, 2013). Since microglia express GR and MR, they were proposed to play important roles in facilitating the adaptive response to stress and AD pathology, notably through their remodeling of neuronal circuits (Carrillo-de Sauvage et al., 2013; Chantong et al., 2012; Parkhurst et al., 2013; Sierra et al., 2008; Tanaka et al., 1997; Vyas et al., 2016).
4. Roles of microglia in AD pathology
While microglia exert neuroprotection in AD, especially through their phagocytic clearance of Aβ (ElAli and Rivest, 2016), a loss of their beneficial functions can exacerbate amyloidosis, leading to subsequent inflammation, synaptic loss, and neuronal damage (Gandy and Heppner, 2013; Heppner et al., 2015; Perry and Holmes, 2014; Rivest, 2015). Vice versa, microglial inability to clear abnormally aggregated Aβ results in the activation of pro-inflammatory signaling pathways, furthering inflammation, oxidative stress, and neurodegeneration in AD (Lee and Landreth, 2010; Piirainen et al., 2017; Solito and Sastre, 2012). The following section aims at providing an overview of these various roles of microglia in AD, emphasizing the relationship to chronic stress as an environmental risk factor.
4.1. Loss of beneficial physiological functions
The classical complement pathway is normally utilized by microglia to eliminate apoptotic or damaged neurons, microbial load and cellular debris, as well as synaptic elements (Boulanger, 2009; Chung et al., 2015). The cascade involves tagging of the targeted elements by C1q, their opsonization and subsequent phagocytosis via C3b complement-CR3 microglial complement receptor (also known as CD11b) interaction. Its contribution to microglial pruning of synapses is now well accepted during normal physiological conditions (Sasaki et al., 2014; Schafer et al., 2012; Stevens et al., 2007) and was also proposed to underlie the pathological or traumatic remodeling of neuronal circuits during stress by ‘dark’ microglia, a newly-defined microglial phenotype discussed below (Bisht et al., 2016). Complement pathway-mediated microglial phagocytosis is a mechanism also recruited in diseases, implicated in AD and in a number of neurodegenerative disorders such as Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis (Loeffler et al., 2006; Stevens et al., 2007). Zanjani et al. documented a progressive activation of complement proteins in human AD brain tissue, specifically of C1q, C3 and C4 around the plaques in both preclinical and clinical stages of the disease (Zanjani et al., 2005). This study revealed staining of NFTs and dystrophic neurites for the membrane attack complex C5b, which results from the cleavage of C5 upon binding of Cb3b to Aβ, in more advanced stages of pathology (Zanjani et al., 2005). Elevated C1q expression levels were also measured during aging in the human postmortem hippocampus, as well as in the hippocampus and frontal cortex of J20 and APP-PS1 mouse models of AD (see Fig. 1), showing co-localization with synapses before the plaques could be detected (Hong et al., 2016; Stephan et al., 2013). Synaptic loss was further prevented in the hippocampus of J20 and Tg2576 mice with either C1q neutralizing antibodies or knocking out of C1q (Fonseca et al., 2004; Hong et al., 2016). The knockout or inhibition of C3 similarly prevented synaptic loss in the hippocampus and cognitive decline in APPswe/PS1dE9 mice (see Fig. 1) still devoid of plaques, as well as during normal aging in wild-type mice (Shi et al., 2017). This detrimental role of microglial complement-mediated phagocytosis is supported by the fact that microglia are the main cellular source of complement C1q protein in the brain (Fonseca et al., 2017).
The exacerbated activity of microglial signaling through their CD33 receptors was also linked to their deleterious effects in AD. Sialic acid is an important glycan typically found on the outer membrane of most cell types. The interaction of sialylated residues on neurons with Siglec receptors on microglia is essential for a healthy neuron-microglial communication in the brain (Varki, 2008; Wielgat and Braszko, 2012). CD33 (or siglec-3), a sialic acid-binding transmembrane receptor protein, is largely expressed by microglia, and functions to inhibit autoimmune activation through its binding to sialylated self-association molecular patterns (Griciuc et al., 2013; Malik et al., 2013; Schwarz et al., 2015; Varki and Angata, 2006). A modest increase in CD33 mRNA and protein levels detected in postmortem human AD frontal cortex was associated with a decrease in Aβ phagocytosis, and consequently, knockout mice lacking CD33 showed a reduced Aβ burden (Griciuc et al., 2013). Sialic acid binding (from sialylated agents found in the vicinity of plaques, such as ApoE and ApoJ) to CD33 results in the activation of SHP1 and SHP2 phosphatases inhibiting the TREM2/DAP12 pathway. This mechanism was proposed to conceal the plaques from microglial immune surveillance and phagocytic clearance (Linnartz and Neumann, 2013). TREM2 binding to its ligands such as ApoE, clusterin (CLU, or ApoJ), and low density lipoproteins, allows its association with the plaques (Atagi et al., 2015; Bailey et al., 2015). TREM2-ApoE interaction is essential for the phagocytosis of apoptotic neurons and extracellular debris, from dying or dead neurons, as will be discussed below when presenting novel microglial phenotypes. It also prevents escalation of neuroinflammation in mice treated with cuprizone, a model of demyelination (Cantoni et al., 2015). TREM2 overexpression in mouse primary microglial cells results in their cytoskeletal reorganization through the activation of DAP12 and phosphorylation of extracellular signal-regulated kinases, and this step is crucial for the phagocytosis of microsphere beads in vitro (Takahashi et al., 2005). Furthermore, TREM2 is expressed by microglial processes in the immediate vicinity of the plaques. Plaque-associated microglia surround amyloid deposits in postmortem human AD brain tissue, as a mechanism proposed to compact the deposits and inhibit any additional deposition of Aβ peptides into the existing plaques, in order to prevent further neurotoxicity and axonal dystrophy (Yuan et al., 2016). This neuroprotective barrier was not observed in Trem2 knockout mice, or in human AD postmortem brain bearing the TREM2 variant R47H, thus supporting the implication of TREM2 in mediating microglial ability to form neuroprotective barriers (Condello et al., 2017, 2015; Yuan et al., 2016).
4.2. Gain of detrimental inflammatory functions
A characteristic hallmark of stress, especially chronic, is the induction of pro-inflammatory mechanisms leading to oxidative stress (Hayashi, 2015; Miller and Sadeh, 2014), a condition resulting from the imbalance between antioxidant mechanisms and production of reactive oxygen species (such as H2O2, O2−, NO, ONOO−/ONOOH), eventually triggering lipid peroxidation and DNA damage (Aschbacher et al., 2013; Bergamini et al., 2004; Epel, 2009). Oxidative stress induced neuronal damage is also a characteristic feature seen in Aβ and tau pathology, and often mediated by microglia and peripheral myeloid cells (Ghribi et al., 2003; Giraldo et al., 2014; Kempuraj et al., 2016; Martin et al., 2017). Microglia engaged in inflammation are considered a major source of reactive oxygen species in the brain (Block and Hong, 2007).
Aβ aggregates can mimic the damage-associated molecular patterns and stimulate Toll-like receptors (TLR; see Box 1) on microglia thereby mounting an inflammatory response by the NRLP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome complex (see Box 1) (Heneka et al., 2013). As a consequence of the inflammasome activation, caspase-1 is recruited to the inflammasome, and a number of pro-inflammatory mediators such as TNFα and IL-1β are released to induce further pro-inflammatory responses by microglia and other brain immune cells like astrocytes (Heneka et al., 2013). Genetic deletion of NLRP3, caspase-1 and TLRs was shown to be neuroprotective by preventing the synaptic loss and cognitive decline, thereby confirming the pro-inflammatory contributions of CD11b-positive microglia in AD (Heneka et al., 2013). Also, microglia were demonstrated to increase the formation of Aβ oligomers and promote further their aggregation due to the ability of Aβ to rapidly bind to ASC specks (see Box 1) released by microglia, seeding further Aβ aggregation and spreading of Aβ pathology, as observed in APPSwePSEN1dE9 mice (see Fig. 1) given intra-hippocampal injection of ASC specks (Venegas et al., 2017). Similarly, co-application of an anti-ASC antibody did not exacerbate Aβ pathology, as was also the case in ASC-deficient APPSwePSEN1dE9 mice treated with brain homogenates derived from APPSwePSEN1dE9 mice (Venegas et al., 2017). These findings provide a direct evidence for the involvement of microglial inflammasome in the seeding and spreading of Aβ pathology in AD patients.
The pro-inflammatory C5a, which also results from the binding of C3b to Aβ, and is recognized by microglial CD88 receptors, leads to the production of various pro-inflammatory cytokines, reactive oxygen species (ROS), reactive nitrogen intermediates, and bioactive amines (Wilkinson and Landreth, 2006; Woodruff et al., 2010). Microglial ROS can be formed by the activities of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, mitochondrial respiratory chain, xanthine oxidase, microsomal enzymes, cycloxygenase and lipoxygenase (Wilkinson and Landreth, 2006). NADPH oxidase forms part of the integral innate phagocytic immune system that utilizes superoxide (O2-) to mediate the destruction of invading microorganisms (Bergendi et al., 1999; Wilkinson and Landreth, 2006). Other sources of ROS include hydrogen peroxide, hydroxyl radical, peroxynitrite and other oxidants (Wilkinson and Landreth, 2006). In rat primary microglia, exposure to Aβ activates NADPH oxidase and causes the release of ROS, which can be terminated through the inhibition of upstream signaling partners such as the src-family tyrosine kinases or phosphatidylinositol-3 kinase (Bianca et al., 1999; McDonald et al., 1997). Aβ induced NADPH oxidase activation in the rat hippocampus results in the formation of peroxynitrite, a potent oxidant that oxidizes proteins, lipids, and DNA (Nam et al., 2012). Peroxynitrite mediated tyrosine nitration and nitrosylation of cysteines is extremely detrimental resulting in protein and enzyme dysfunction, leading ultimately to neuronal death in vitro (Bonfoco et al., 1995; Mander and Brown, 2005). The accumulation of peroxynitrite in AD is supported by the presence of elevated levels of nitrated proteins in postmortem AD brain tissue, particularly within regions containing NFTs (Castegna et al., 2003). Lipid peroxidation and DNA oxidative damage was also demonstrated in postmortem human AD brains (Matsuoka et al., 2001; Nunomura et al., 1999; Sayre et al., 1997; Wang et al., 2005). These findings indicate that pro-inflammatory microglial activities may be detrimental in AD due to the formation of reactive oxygen and nitrogen intermediates resulting in oxidative stress-induced neuronal damage, which could be further exacerbated by chronic stress.
5. Newly-described microglial phenotypes
The recent technical advancements, including in high-throughput gene expression analyses distinguishing microglia from bone marrow-derived myeloid cells, have allowed to start characterizing the diversity of microglial phenotypes in the CNS, especially in the context of altered brain homeostasis. Electron microscopy with immunostaining against these recently identified markers was also used by our group to describe ‘dark’ microglia, a newly-defined phenotype associated with both chronic stress and AD pathology.
5.1. Dark microglia
In 2016, we described the ‘dark’ microglia, which are strikingly distinct from typical IBA1-positive microglia in their ultrastructural details (Bisht et al., 2016). In electron microscopy, these cells showed a condensed, electron-dense cytoplasm and nucleoplasm, a characteristic giving them a ‘dark’ appearance. Dark microglia also displayed endoplasmic reticulum and Golgi apparatus dilation, as well as mitochondrial alterations, considered the best-characterized signs of oxidative stress at the ultrastructural level. These cells seemed to be extremely active at synapses, by means of their thin, elongated, ramified processes strongly expressing complement receptor CR3 when contacting synaptic clefts and encircling synaptic elements. In mouse hippocampus, dark microglia were highly prevalent in conditions where normal homeostasis is disturbed: chronic stress, aging, AD pathology (APP-PS1 mice; see Fig. 1), and where fractalkine signaling is disrupted (Cx3cr1 knockout mice; see Box 1). Especially in APP-PS1 mice, dark microglia were frequently encountered around the plaques, encircling dystrophic neurons, containing amyloid depositions in their processes, and showing a strong expression of TREM2, involved as discussed above in mediating microglial phagocytosis (Colonna and Wang, 2016; Numasawa et al., 2011; Painter et al., 2015; Zheng et al., 2017). Dark microglia showed a downregulated expression of homeostatic markers, CX3CR1, IBA1 and P2RY12, but strongly expressed the microglia-specific 4D4 in their processes (Bisht et al., 2016). They were proposed to represent a subset of hyperactive microglia that become stressed as a result of their hyperactivity under adaptive pressure, leading to exacerbated interactions with synapses. The increased phagocytic activity of dark microglia also suggested that these cells might contribute to restricting the Aβ burden.
5.2. Disease associated microglia
Keren-Shaul et al. identified a unique microglial population, the disease associated microglia (DAM), using single-cell RNA-sequencing, by comparing microglia from wild-type versus 5xFAD mice (see Fig. 1) (Keren-Shaul et al., 2017). Using the immune marker CD45+, myeloid cells were isolated from the brain and grouped into different clusters based on single-cell RNA-sequencing data. Three main microglial clusters emerged. Cluster I comprised cells expressing markers of homeostatic microglia, which were present in both control and AD pathology animals, while clusters II and III microglia were only observed in AD pathology. The clusters II and III microglia showed an upregulated expression of genes related to lipid metabolism, phagocytosis, and AD pathology, such as CST7, APOE, LPL, CTSD, TYROBP, and TREM2, and downregulated expression of homeostatic markers such as transmembrane protein 119 (TMEM119, selective to microglia) and CX3CR1, without changes in expression of inflammatory cytokines. They differed in their expression levels of phagocytic markers, suggesting an intermediate phenotype between homeostatic and cluster III (DAM) microglia. This transition was proposed to proceed in two steps, with the complete transition into DAM requiring the expression of TREM2, highlighting again the importance of this receptor in mediating microglial response to AD pathology. The DAM phenotype was also described in human postmortem AD brains, where it was found to contain intracellular Aβ, suggesting a neuroprotective role for the DAM (Keren-Shaul et al., 2017).
5.3. Neurodegenerative microglial phenotype
The complexity of the roles played by microglia in homeostasis as well as diseased conditions is portrayed in another study by Krasemann et al. where the authors described regulatory mechanisms underlying the switch of microglia from a homeostatic to a neurodegenerative phenotype: MGnD (microglia with a neurodegenerative phenotype) (Krasemann et al., 2017). Microglia were isolated from the brain and spinal cord using a specific Fc receptor-like S scavenger receptor (FCRLS) antibody, in mouse models of AD (APP-PS1), multiple sclerosis (experimental autoimmune encephalomyelitis), and amyotrophic lateral sclerosis (SOD1-G93A). Under normal physiological conditions, homeostatic microglia expressed Olfml3, P2ry12, Tmem119, Mef2a, Jun, and Sall1, among other genes regulated by TGFβ, all of which were downregulated by the MGnD phenotype. Similarly, these cells showed an upregulated expression of APOE, Axl, and Clec7a genes. Microglial transformation into MGnD, identified by gene expression signature, was only observed in contexts where neuronal apoptosis occurred. In APP-PS1 mice, MGnD primarily encircled amyloid plaques and dystrophic neurites, blanketed by homeostatic microglia in the periphery. They also expressed TMEM119, providing support to their microglial origin. In APOE knockout mice, microglia still phagocytosed dying neurons, but did not show the expression patterns of MGnD, indicating the importance of APOE for maintaining this phenotype. Similar findings were observed in TREM2 knockout animals, proposing that an APOE-TREM2 switch mediates the transformation of homeostatic microglia into phagocytic MGnD during neurodegeneration.
The above three studies stress the importance of studying the variety of microglial phenotypes in the CNS. Brain resident microglia and peripheral macrophages should not be combined into one category, neither should the brain resident microglia considered as a single cell entity, since they appear to comprise heterogeneous populations having distinct gene expression signatures, functions, and morphologies depending on the homeostasis status. Future studies aiming to address the roles of microglia in stressed and pathological states should take these new findings into consideration.
6. Conclusion and perspective
The way the brain microglia react to homeostatic changes as in stress and neurodegeneration determines the overall outcome of the brain's capacity to respond to these alterations. These responses contribute to what is known as allostasis or the adaptive measures taken to adapt the organism to the adverse challenges. An imbalance or overload in the adaptive responses to conditions of prolonged stress especially by microglia may lead to altered physiological functions, becoming either maladaptive or pro-inflammatory, thus predisposing the brain to secondary insults in the form of trauma or disease. Hence, microglia were thus proposed to play an important role in linking the adverse effects of chronic stress with the onset and progression of AD (Piirainen et al., 2017). Therefore, a better understanding of the roles and underlying mechanisms by which microglia may contribute to regulating brain functions and behavior in response to changes in the environment under both homeostatic and altered states is required.
While microglia can exacerbate pathological damage during chronic stress and AD pathology via dysregulated complement-mediated phagocytosis, release of inflammatory cytokines, and their mediation of oxidative stress, they may also contribute positively to the restoration of neuronal homeostasis (see Fig. 2). The recent description of novel microglial phenotypes associated with chronic stress and neurodegenerative diseases further supports the view that microglia represent a heterogeneous cell population in which different phenotypes behave differently depending on the context. Through this review, we have attempted to highlight the importance of studying individual microglial phenotypes and their distinct contributions to brain homeostasis and disease, considering that their activities could be selectively targeted to promote neuroprotection in various neurological and neurodegenerative contexts.
Fig. 2.
Overview of microglial functions in response to stress and AD pathology.
Acknowledgements
We are grateful to Marie-Kim St-Pierre, Katherine Picard, and Mathilde S. Henry for their critical revision of the manuscript. This work was supported by a CIHR Foundation Scheme grant to MET and an excellence scholarship from Fondation du CHU de Québec to KB. MET is a Canada Research Chair (Tier 2) in Neuroimmune plasticity in health and therapy.
Box 1: Glossary
- Aβ peptides
Proteolytic cleavage product of amyloid precursor protein or APP, formed as a result of sequential cleavage by β-secretase 1, and γ-secretase enzymes
- Aβ40 and Aβ42
40- and 42-residue long isoforms of Aβ respectively, derived from APP proteolysis, with Aβ42 forming a major component of amyloid plaques
- Aβ42/Aβ40
This ratio plays a critical role in AD, with higher ratios often associated with greater neurotoxicity, and correlating with early-onset familial AD cases
- APP carboxyl-terminal (APP-CTF) fragments
A 99 amino acid long C-terminal fragment generated by the cleavage of APP by the β-site APP cleaving enzyme, which on further cleavage by γ-secretase generates Aβ that gets deposited in AD
- Toll-like receptors (TLRs)
A class of protein receptors expressed on immune cells that recognize microbe derived structurally conserved molecules, and thus form an important part of the innate immune system
- Inflammasome
A group of inducible, high molecular weight, cytosolic multiprotein complexes and an essential component of the innate immune response important for the clearance of pathogens or damaged cells
- ASC Speck
Apoptosis-associated Speck-like protein containing Caspase Activation and Recruitment Domain. It functions as the adaptor protein in the inflammasome complex that after release in response to specific stimuli can form a discrete “speck,” within the activated cell. It functions to recruit caspase-1 to eventually result in the production of inflammatory cytokines, IL-1β and IL-18
- Fractalkine signaling
Involves the interaction between fractalkine ligand CX3CL1 expressed by neurons that interacts with its unique receptor, CX3CR1, expressed by microglia and macrophages. It is essential for proper neuron-microglia communication in the brain across homeostatic and diseased conditions
References
- Abe-Dohmae S., Ikeda Y., Matsuo M., Hayashi M., Okuhira K., Ueda K., Yokoyama S. Human ABCA7 supports apolipoprotein-mediated release of cellular cholesterol and phospholipid to generate high density lipoprotein. J. Biol. Chem. 2004;279:604–611. doi: 10.1074/jbc.M309888200. [DOI] [PubMed] [Google Scholar]
- Alkadhi K.A. Chronic psychosocial stress exposes Alzheimer's disease phenotype in a novel at-risk model. Front. Biosci. 2012;4:214–229. doi: 10.2741/371. [DOI] [PubMed] [Google Scholar]
- Ard M.D., Cole G.M., Wei J., Mehrle A.P., Fratkin J.D. Scavenging of Alzheimer's amyloid beta-protein by microglia in culture. J. Neurosci. Res. 1996;43:190–202. doi: 10.1002/(SICI)1097-4547(19960115)43:2<190::AID-JNR7>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
- Aschbacher K., O'Donovan A., Wolkowitz O.M., Dhabhar F.S., Su Y., Epel E. Good stress, bad stress and oxidative stress: insights from anticipatory cortisol reactivity. Psychoneuroendocrinology. 2013;38:1698–1708. doi: 10.1016/j.psyneuen.2013.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atagi Y., Liu C.-C., Painter M.M., Chen X.-F., Verbeeck C., Zheng H., Li X., Rademakers R., Kang S.S., Xu H., Younkin S., Das P., Fryer J.D., Bu G. Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2) J. Biol. Chem. 2015;290:26043–26050. doi: 10.1074/jbc.M115.679043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aznar S., Knudsen G.M. Depression and Alzheimer's disease: is stress the initiating factor in a common neuropathological cascade? J. Alzheimers. Dis. 2011;23:177–193. doi: 10.3233/JAD-2010-100390. [DOI] [PubMed] [Google Scholar]
- Bailey C.C., DeVaux L.B., Farzan M. The triggering receptor expressed on myeloid cells 2 binds apolipoprotein E. J. Biol. Chem. 2015;290:26033–26042. doi: 10.1074/jbc.M115.677286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balschun D., Randolf A., Pitossi F., Schneider H., Del Rey A., Besedovsky H.O. Hippocampal interleukin-1 beta gene expression during long-term potentiation decays with age. Ann. N. Y. Acad. Sci. 2003;992:1–8. doi: 10.1111/j.1749-6632.2003.tb03132.x. [DOI] [PubMed] [Google Scholar]
- Bao A.-M., Meynen G., Swaab D.F. The stress system in depression and neurodegeneration: focus on the human hypothalamus. Brain Res. Rev. 2008;57:531–553. doi: 10.1016/j.brainresrev.2007.04.005. [DOI] [PubMed] [Google Scholar]
- Bergamini C.M., Gambetti S., Dondi A., Cervellati C. Oxygen, reactive oxygen species and tissue damage. Curr. Pharmaceut. Des. 2004;10:1611–1626. doi: 10.2174/1381612043384664. [DOI] [PubMed] [Google Scholar]
- Bergendi L., Benes L., Duracková Z., Ferencik M. Chemistry, physiology and pathology of free radicals. Life Sci. 1999;65:1865–1874. doi: 10.1016/s0024-3205(99)00439-7. [DOI] [PubMed] [Google Scholar]
- Bianca V.D., Dusi S., Bianchini E., Dal Prà I., Rossi F. beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer's disease. J. Biol. Chem. 1999;274:15493–15499. doi: 10.1074/jbc.274.22.15493. [DOI] [PubMed] [Google Scholar]
- Bisht K., Sharma K.P., Lecours C., Sánchez M.G., El Hajj H., Milior G., Olmos-Alonso A., Gómez-Nicola D., Luheshi G., Vallières L., Branchi I., Maggi L., Limatola C., Butovsky O., Tremblay M.-È.È., Gabriela Sánchez M., El Hajj H., Milior G., Olmos-Alonso A., Gómez-Nicola D., Luheshi G., Vallières L., Branchi I., Maggi L., Limatola C., Butovsky O., Tremblay M.-È.È. Dark microglia: a new phenotype predominantly associated with pathological states. Glia. 2016;64:826–839. doi: 10.1002/glia.22966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Block M.L., Hong J.-S. Chronic microglial activation and progressive dopaminergic neurotoxicity. Biochem. Soc. Trans. 2007;35:1127–1132. doi: 10.1042/BST0351127. [DOI] [PubMed] [Google Scholar]
- Bonfoco E., Krainc D., Ankarcrona M., Nicotera P., Lipton S.A. Apoptosis and necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-D-aspartate or nitric oxide/superoxide in cortical cell cultures. Proc. Natl. Acad. Sci. U. S. A. 1995;92:7162–7166. doi: 10.1073/pnas.92.16.7162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boulanger L.M. Immune proteins in brain development and synaptic plasticity. Neuron. 2009;64:93–109. doi: 10.1016/j.neuron.2009.09.001. [DOI] [PubMed] [Google Scholar]
- Braak H., Del Tredici K. The pathological process underlying Alzheimer's disease in individuals under thirty. Acta Neuropathol. 2011;121:171–181. doi: 10.1007/s00401-010-0789-4. [DOI] [PubMed] [Google Scholar]
- Budas G., Coughlan C.M., Seckl J.R., Breen K.C. The effect of corticosteroids on amyloid beta precursor protein/amyloid precursor-like protein expression and processing in vivo. Neurosci. Lett. 1999;276:61–64. doi: 10.1016/s0304-3940(99)00790-9. [DOI] [PubMed] [Google Scholar]
- Calcia M.A., Bonsall D.R., Bloomfield P.S., Selvaraj S., Barichello T., Howes O.D. Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology (Berlin) 2016;233:1637–1650. doi: 10.1007/s00213-016-4218-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell S.N., Zhang C., Monte L., Roe A.D., Rice K.C., Taché Y., Masliah E., Rissman R.A. Increased tau phosphorylation and aggregation in the hippocampus of mice overexpressing corticotropin-releasing factor. J. Alzheimers. Dis. 2015;43:967–976. doi: 10.3233/JAD-141281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cantoni C., Bollman B., Licastro D., Xie M., Mikesell R., Schmidt R., Yuede C.M., Galimberti D., Olivecrona G., Klein R.S., Cross A.H., Otero K., Piccio L. TREM2 regulates microglial cell activation in response to demyelination in vivo. Acta Neuropathol. 2015;129:429–447. doi: 10.1007/s00401-015-1388-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carrillo-de Sauvage M.Á., Maatouk L., Arnoux I., Pasco M., Sanz Diez A., Delahaye M., Herrero M.T., Newman T.A., Calvo C.F., Audinat E., Tronche F., Vyas S. Potent and multiple regulatory actions of microglial glucocorticoid receptors during CNS inflammation. Cell Death Differ. 2013;20:1546–1557. doi: 10.1038/cdd.2013.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carroll J.C., Iba M., Bangasser D.A., Valentino R.J., James M.J., Brunden K.R., Lee V.M.-Y., Trojanowski J.Q. Chronic stress exacerbates tau pathology, neurodegeneration, and cognitive performance through a corticotropin-releasing factor receptor-dependent mechanism in a transgenic mouse model of tauopathy. J. Neurosci. 2011;31:14436–14449. doi: 10.1523/JNEUROSCI.3836-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cartier N., Lewis C.-A., Zhang R., Rossi F.M.V. The role of microglia in human disease: therapeutic tool or target? Acta Neuropathol. 2014;128:363–380. doi: 10.1007/s00401-014-1330-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castegna A., Thongboonkerd V., Klein J.B., Lynn B., Markesbery W.R., Butterfield D.A. Proteomic identification of nitrated proteins in Alzheimer's disease brain. J. Neurochem. 2003;85:1394–1401. doi: 10.1046/j.1471-4159.2003.01786.x. [DOI] [PubMed] [Google Scholar]
- Chantong B., Kratschmar D.V., Nashev L.G., Balazs Z., Odermatt A. Mineralocorticoid and glucocorticoid receptors differentially regulate NF-kappaB activity and pro-inflammatory cytokine production in murine BV-2 microglial cells. J. Neuroinflammation. 2012;9:260. doi: 10.1186/1742-2094-9-260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiba S., Numakawa T., Ninomiya M., Richards M.C., Wakabayashi C., Kunugi H. Chronic restraint stress causes anxiety- and depression-like behaviors, downregulates glucocorticoid receptor expression, and attenuates glutamate release induced by brain-derived neurotrophic factor in the prefrontal cortex. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2012;39:112–119. doi: 10.1016/j.pnpbp.2012.05.018. [DOI] [PubMed] [Google Scholar]
- Chung W.-S., Welsh C.A., Barres B.A., Stevens B. Do glia drive synaptic and cognitive impairment in disease? Nat. Neurosci. 2015;18:1539–1545. doi: 10.1038/nn.4142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colonna M., Wang Y. TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nat. Rev. Neurosci. 2016;17:201–207. doi: 10.1038/nrn.2016.7. [DOI] [PubMed] [Google Scholar]
- Condello C., Yuan P., Grutzendler J. Microglia-mediated neuroprotection, TREM2, and Alzheimer's disease: evidence from optical imaging. Biol. Psychiatr. 2017;1–11 doi: 10.1016/j.biopsych.2017.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Condello C., Yuan P., Schain A., Grutzendler J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat. Commun. 2015;6:6176. doi: 10.1038/ncomms7176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connor B., Young D., Yan Q., Faull R.L., Synek B., Dragunow M. Brain-derived neurotrophic factor is reduced in Alzheimer's disease. Brain Res. Mol. Brain Res. 1997;49:71–81. doi: 10.1016/s0169-328x(97)00125-3. [DOI] [PubMed] [Google Scholar]
- Cuyvers E., De Roeck A., Van den Bossche T., Van Cauwenberghe C., Bettens K., Vermeulen S., Mattheijssens M., Peeters K., Engelborghs S., Vandenbulcke M., Vandenberghe R., De Deyn P.P., Van Broeckhoven C., Sleegers K. Mutations in ABCA7 in a Belgian cohort of Alzheimer's disease patients: a targeted resequencing study. Lancet Neurol. 2015;14:814–822. doi: 10.1016/S1474-4422(15)00133-7. [DOI] [PubMed] [Google Scholar]
- de Kloet E.R., Van Acker S.A., Sibug R.M., Oitzl M.S., Meijer O.C., Rahmouni K., de Jong W. Brain mineralocorticoid receptors and centrally regulated functions. Kidney Int. 2000;57:1329–1336. doi: 10.1046/j.1523-1755.2000.00971.x. [DOI] [PubMed] [Google Scholar]
- del Rey A., Balschun D., Wetzel W., Randolf A., Besedovsky H.O. A cytokine network involving brain-borne IL-1β, IL-1ra, IL-18, IL-6, and TNFα operates during long-term potentiation and learning. Brain Behav. Immun. 2013;33:15–23. doi: 10.1016/j.bbi.2013.05.011. [DOI] [PubMed] [Google Scholar]
- Derecki N.C., Katzmarski N., Kipnis J., Meyer-Luehmann M. Microglia as a critical player in both developmental and late-life CNS pathologies. Acta Neuropathol. 2014;128:333–345. doi: 10.1007/s00401-014-1321-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devi L., Alldred M.J., Ginsberg S.D., Ohno M. Sex- and brain region-specific acceleration of β-amyloidogenesis following behavioral stress in a mouse model of Alzheimer's disease. Mol. Brain. 2010;3:34. doi: 10.1186/1756-6606-3-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong H., Csernansky J.G. Effects of stress and stress hormones on amyloid-beta protein and plaque deposition. J. Alzheimers. Dis. 2009;18:459–469. doi: 10.3233/JAD-2009-1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong H., Goico B., Martin M., Csernansky C.A., Bertchume A., Csernansky J.G. Modulation of hippocampal cell proliferation, memory, and amyloid plaque deposition in APPsw (Tg2576) mutant mice by isolation stress. Neuroscience. 2004;127:601–609. doi: 10.1016/j.neuroscience.2004.05.040. [DOI] [PubMed] [Google Scholar]
- Donovan N.J., Locascio J.J., Marshall G.A., Gatchel J., Hanseeuw B.J., Rentz D.M., Johnson K.A., Sperling R.A., Harvard Aging Brain Study Longitudinal association of amyloid beta and anxious-depressive symptoms in cognitively normal older adults. Am. J. Psychiatr. 2018 doi: 10.1176/appi.ajp.2017.17040442. appiajp201717040442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dos Santos L.R., Pimassoni L.H.S., Sena G.G.S., Camporez D., Belcavello L., Trancozo M., Morelato R.L., Errera F.I.V., Bueno M.R.P., de Paula F. Validating GWAS variants from microglial genes implicated in Alzheimer's disease. J. Mol. Neurosci. 2017;62:215–221. doi: 10.1007/s12031-017-0928-7. [DOI] [PubMed] [Google Scholar]
- Edison P., Archer H.A., Gerhard A., Hinz R., Pavese N., Turkheimer F.E., Hammers A., Tai Y.F., Fox N., Kennedy A., Rossor M., Brooks D.J. Microglia, amyloid, and cognition in Alzheimer's disease: an [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol. Dis. 2008;32:412–419. doi: 10.1016/j.nbd.2008.08.001. [DOI] [PubMed] [Google Scholar]
- Efthymiou A.G., Goate A.M. Late onset Alzheimer's disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener. 2017;12:43. doi: 10.1186/s13024-017-0184-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ElAli A., Rivest S. Microglia in Alzheimer's disease: a multifaceted relationship. Brain. Behav. Immun. 2016;55:138–150. doi: 10.1016/j.bbi.2015.07.021. [DOI] [PubMed] [Google Scholar]
- Ellis B.J., Del Giudice M. Beyond allostatic load: rethinking the role of stress in regulating human development. Dev. Psychopathol. 2014;26:1–20. doi: 10.1017/S0954579413000849. [DOI] [PubMed] [Google Scholar]
- Epel E.S. Psychological and metabolic stress: a recipe for accelerated cellular aging? Hormones (Basel) 2009;8:7–22. doi: 10.14310/horm.2002.1217. [DOI] [PubMed] [Google Scholar]
- Fonseca M.I., Chu S.-H., Hernandez M.X., Fang M.J., Modarresi L., Selvan P., MacGregor G.R., Tenner A.J. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J. Neuroinflammation. 2017;14:48. doi: 10.1186/s12974-017-0814-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonseca M.I., Zhou J., Botto M., Tenner A.J. Absence of C1q leads to less neuropathology in transgenic mouse models of Alzheimer's disease. J. Neurosci. 2004;24:6457–6465. doi: 10.1523/JNEUROSCI.0901-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franceschi M., Airaghi L., Gramigna C., Truci G., Manfredi M.G., Canal N., Catania A. ACTH and cortisol secretion in patients with Alzheimer's disease. J. Neurol. Neurosurg. Psychiatry. 1991;54:836–837. doi: 10.1136/jnnp.54.9.836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuhrmann M., Bittner T., Jung C.K.E., Burgold S., Page R.M., Mitteregger G., Haass C., LaFerla F.M., Kretzschmar H., Herms J. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer's disease. Nat. Neurosci. 2010;13:411–413. doi: 10.1038/nn.2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuxe K., Cintra A., Agnati L.F., Härfstrand A., Wikstrom A.C., Okret S., Zoli M., Miller L.S., Greene J.L., Gustafsson J.A. Studies on the cellular localization and distribution of glucocorticoid receptor and estrogen receptor immunoreactivity in the central nervous system of the rat and their relationship to the monoaminergic and peptidergic neurons of the brain. J. Steroid Biochem. 1987;27:159–170. doi: 10.1016/0022-4731(87)90306-2. [DOI] [PubMed] [Google Scholar]
- Gandy S., Heppner F.L. Microglia as dynamic and essential components of the amyloid hypothesis. Neuron. 2013;78:575–577. doi: 10.1016/j.neuron.2013.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerges N.Z., Aleisa A.M., Schwarz L.A., Alkadhi K.A. Reduced basal CaMKII levels in hippocampal CA1 region: possible cause of stress-induced impairment of LTP in chronically stressed rats. Hippocampus. 2004;14:402–410. doi: 10.1002/hipo.10193. [DOI] [PubMed] [Google Scholar]
- Ghribi O., Herman M.M., Savory J. Lithium inhibits Abeta-induced stress in endoplasmic reticulum of rabbit hippocampus but does not prevent oxidative damage and tau phosphorylation. J. Neurosci. Res. 2003;71:853–862. doi: 10.1002/jnr.10511. [DOI] [PubMed] [Google Scholar]
- Giraldo E., Lloret A., Fuchsberger T., Viña J. Aβ and tau toxicities in Alzheimer's are linked via oxidative stress-induced p38 activation: protective role of vitamin E. Redox Biol. 2014;2:873–877. doi: 10.1016/j.redox.2014.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glenner G.G., Wong C.W. Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem. Biophys. Res. Commun. 1984;122:1131–1135. doi: 10.1016/0006-291x(84)91209-9. [DOI] [PubMed] [Google Scholar]
- Grathwohl S.A., Kälin R.E., Bolmont T., Prokop S., Winkelmann G., Kaeser S.A., Odenthal J., Radde R., Eldh T., Gandy S., Aguzzi A., Staufenbiel M., Mathews P.M., Wolburg H., Heppner F.L., Jucker M. Formation and maintenance of Alzheimer's disease beta-amyloid plaques in the absence of microglia. Nat. Neurosci. 2009;12:1361–1363. doi: 10.1038/nn.2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green K.N., Billings L.M., Roozendaal B., McGaugh J.L., LaFerla F.M. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer's disease. J. Neurosci. 2006;26:9047–9056. doi: 10.1523/JNEUROSCI.2797-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griciuc A., Serrano-Pozo A., Parrado A.R., Lesinski A.N., Asselin C.N., Mullin K., Hooli B., Choi S.H., Hyman B.T., Tanzi R.E. Alzheimer's disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631–643. doi: 10.1016/j.neuron.2013.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groeneweg F.L., Karst H., de Kloet E.R., Joëls M. Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling. Mol. Cell. Endocrinol. 2012;350:299–309. doi: 10.1016/j.mce.2011.06.020. [DOI] [PubMed] [Google Scholar]
- Grundke-Iqbal I., Iqbal K., Tung Y.C., Quinlan M., Wisniewski H.M., Binder L.I. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. U. S. A. 1986;83:4913–4917. doi: 10.1073/pnas.83.13.4913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guerreiro R., Wojtas A., Bras J., Carrasquillo M., Rogaeva E., Majounie E., Cruchaga C., Sassi C., Kauwe J.S.K., Younkin S., Hazrati L., Collinge J., Pocock J., Lashley T., Williams J., Lambert J.-C., Amouyel P., Goate A., Rademakers R., Morgan K., Powell J., St George-Hyslop P., Singleton A., Hardy J., Alzheimer Genetic Analysis Group TREM2 variants in Alzheimer's disease. N. Engl. J. Med. 2013;368:117–127. doi: 10.1056/NEJMoa1211851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guillemin G.J., Brew B.J. Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. J. Leukoc. Biol. 2004;75:388–397. doi: 10.1189/jlb.0303114. [DOI] [PubMed] [Google Scholar]
- Hardy J.A., Higgins G.A. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992;256:184–185. doi: 10.1126/science.1566067. [DOI] [PubMed] [Google Scholar]
- Hayashi T. Conversion of psychological stress into cellular stress response: roles of the sigma-1 receptor in the process. Psychiatr. Clin. Neurosci. 2015;69:179–191. doi: 10.1111/pcn.12262. [DOI] [PubMed] [Google Scholar]
- Hellwig S., Brioschi S., Dieni S., Frings L., Masuch A., Blank T., Biber K. Altered microglia morphology and higher resilience to stress-induced depression-like behavior in CX3CR1-deficient mice. Brain Behav. Immun. 2016;55:126–137. doi: 10.1016/j.bbi.2015.11.008. [DOI] [PubMed] [Google Scholar]
- Heneka M.T., Kummer M.P., Stutz A., Delekate A., Schwartz S., Vieira-Saecker A., Griep A., Axt D., Remus A., Tzeng T.-C., Gelpi E., Halle A., Korte M., Latz E., Golenbock D.T. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674–678. doi: 10.1038/nature11729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heppner F.L., Ransohoff R.M., Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015;16:358–372. doi: 10.1038/nrn3880. [DOI] [PubMed] [Google Scholar]
- Hinwood M., Morandini J., Day T.A., Walker F.R. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cerebr. Cortex. 2012;22:1442–1454. doi: 10.1093/cercor/bhr229. [DOI] [PubMed] [Google Scholar]
- Holtzman D.M., Morris J.C., Goate A.M. Alzheimer's disease: the challenge of the second century. Sci. Transl. Med. 2011;3:77sr1. doi: 10.1126/scitranslmed.3002369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong S., Beja-Glasser V.F., Nfonoyim B.M., Frouin A., Li S., Ramakrishnan S., Merry K.M., Shi Q., Rosenthal A., Barres B.A., Lemere C.A., Selkoe D.J., Stevens B. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352:712–716. doi: 10.1126/science.aad8373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H.-J., Liang K.-C., Ke H.-C., Chang Y.-Y., Hsieh-Li H.M. Long-term social isolation exacerbates the impairment of spatial working memory in APP/PS1 transgenic mice. Brain Res. 2011;1371:150–160. doi: 10.1016/j.brainres.2010.11.043. [DOI] [PubMed] [Google Scholar]
- Jehle A.W., Gardai S.J., Li S., Linsel-Nitschke P., Morimoto K., Janssen W.J., Vandivier R.W., Wang N., Greenberg S., Dale B.M., Qin C., Henson P.M., Tall A.R. ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J. Cell Biol. 2006;174:547–556. doi: 10.1083/jcb.200601030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeong Y.H., Park C.H., Yoo J., Shin K.Y., Ahn S.-M., Kim H.-S., Lee S.H., Emson P.C., Suh Y.-H. Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV717I-CT100 transgenic mice, an Alzheimer's disease model. Faseb. J. 2006;20:729–731. doi: 10.1096/fj.05-4265fje. [DOI] [PubMed] [Google Scholar]
- Jiang T., Yu J.-T., Zhu X.-C., Tan M.-S., Gu L.-Z., Zhang Y.-D., Tan L. Triggering receptor expressed on myeloid cells 2 knockdown exacerbates aging-related neuroinflammation and cognitive deficiency in senescence-accelerated mouse prone 8 mice. Neurobiol. Aging. 2014;35:1243–1251. doi: 10.1016/j.neurobiolaging.2013.11.026. [DOI] [PubMed] [Google Scholar]
- Johansson L. Can stress increase Alzheimer's disease risk in women? Expert Rev. Neurother. 2014;14:123–125. doi: 10.1586/14737175.2014.878651. [DOI] [PubMed] [Google Scholar]
- Johansson L., Guo X., Waern M., Ostling S., Gustafson D., Bengtsson C., Skoog I. Midlife psychological stress and risk of dementia: a 35-year longitudinal population study. Brain. 2010;133:2217–2224. doi: 10.1093/brain/awq116. [DOI] [PubMed] [Google Scholar]
- Johansson L., Skoog I., Gustafson D.R., Olesen P.J., Waern M., Bengtsson C., Björkelund C., Pantoni L., Simoni M., Lissner L., Guo X. Midlife psychological distress associated with late-life brain atrophy and white matter lesions. Psychosom. Med. 2012;74:120–125. doi: 10.1097/PSY.0b013e318246eb10. [DOI] [PubMed] [Google Scholar]
- Johns P. Clinical Neuroscience. Elsevier; 2014. Dementia; pp. 145–162. [Google Scholar]
- Jonsson T., Stefansson H., Steinberg S., Jonsdottir I., Jonsson P.V., Snaedal J., Bjornsson S., Huttenlocher J., Levey A.I., Lah J.J., Rujescu D., Hampel H., Giegling I., Andreassen O.A., Engedal K., Ulstein I., Djurovic S., Ibrahim-Verbaas C., Hofman A., Ikram M.A., van Duijn C.M., Thorsteinsdottir U., Kong A., Stefansson K. Variant of TREM2 associated with the risk of Alzheimer's disease. N. Engl. J. Med. 2013;368:107–116. doi: 10.1056/NEJMoa1211103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadavath H., Hofele R.V., Biernat J., Kumar S., Tepper K., Urlaub H., Mandelkow E., Zweckstetter M. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc. Natl. Acad. Sci. U. S. A. 2015;112:7501–7506. doi: 10.1073/pnas.1504081112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karege F., Bondolfi G., Gervasoni N., Schwald M., Aubry J.-M., Bertschy G. Low brain-derived neurotrophic factor (BDNF) levels in serum of depressed patients probably results from lowered platelet BDNF release unrelated to platelet reactivity. Biol. Psychiatr. 2005;57:1068–1072. doi: 10.1016/j.biopsych.2005.01.008. [DOI] [PubMed] [Google Scholar]
- Kempuraj D., Thangavel R., Natteru P.A., Selvakumar G.P., Saeed D., Zahoor H., Zaheer S., Iyer S.S., Zaheer A. Neuroinflammation induces neurodegeneration. J. Neurol. Neurosurg. Spine. 2016;1 [PMC free article] [PubMed] [Google Scholar]
- Keren-Shaul H., Spinrad A., Weiner A., Matcovitch-Natan O., Dvir-Szternfeld R., Ulland T.K., David E., Baruch K., Lara-Astaiso D., Toth B., Itzkovitz S., Colonna M., Schwartz M., Amit I. A unique microglia type associated with restricting development of Alzheimer's disease. Cell. 2017;169 doi: 10.1016/j.cell.2017.05.018. 1276–1290.e17. [DOI] [PubMed] [Google Scholar]
- Kim W.S., Li H., Ruberu K., Chan S., Elliott D.A., Low J.K., Cheng D., Karl T., Garner B. Deletion of Abca7 increases cerebral amyloid-β accumulation in the J20 mouse model of Alzheimer's disease. J. Neurosci. 2013;33:4387–4394. doi: 10.1523/JNEUROSCI.4165-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinberger G., Yamanishi Y., Suárez-Calvet M., Czirr E., Lohmann E., Cuyvers E., Struyfs H., Pettkus N., Wenninger-Weinzierl A., Mazaheri F., Tahirovic S., Lleó A., Alcolea D., Fortea J., Willem M., Lammich S., Molinuevo J.L., Sánchez-Valle R., Antonell A., Ramirez A., Heneka M.T., Sleegers K., van der Zee J., Martin J.-J., Engelborghs S., Demirtas-Tatlidede A., Zetterberg H., Van Broeckhoven C., Gurvit H., Wyss-Coray T., Hardy J., Colonna M., Haass C. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci. Transl. Med. 2014;6 doi: 10.1126/scitranslmed.3009093. 243ra86. [DOI] [PubMed] [Google Scholar]
- Krasemann S., Madore C., Cialic R., Baufeld C., Calcagno N., El Fatimy R., Beckers L., O'Loughlin E., Xu Y., Fanek Z., Greco D.J., Smith S.T., Tweet G., Humulock Z., Zrzavy T., Conde-Sanroman P., Gacias M., Weng Z., Chen H., Tjon E., Mazaheri F., Hartmann K., Madi A., Ulrich J.D., Glatzel M., Worthmann A., Heeren J., Budnik B., Lemere C., Ikezu T., Heppner F.L., Litvak V., Holtzman D.M., Lassmann H., Weiner H.L., Ochando J., Haass C., Butovsky O., O'Loughlin E., Xu Y., Fanek Z., Greco D.J., Smith S.T., Tweet G., Humulock Z., Zrzavy T., Conde-Sanroman P., Gacias M., Weng Z., Chen H., Tjon E., Mazaheri F., Hartmann K., Madi A., Ulrich J.D., Glatzel M., Worthmann A., Heeren J., Budnik B., Lemere C., Ikezu T., Heppner F.L., Litvak V., Holtzman D.M., Lassmann H., Weiner H.L., Ochando J., Haass C., Butovsky O. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 2017;47 doi: 10.1016/j.immuni.2017.08.008. 566–581.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurz A., Perneczky R. Amyloid clearance as a treatment target against Alzheimer's disease. J. Alzheimers. Dis. 2011;24(Suppl. 2):61–73. doi: 10.3233/JAD-2011-102139. [DOI] [PubMed] [Google Scholar]
- Lanoiselée H.M., Nicolas G., Wallon D., Rovelet-Lecrux A., Lacour M., Rousseau S., Richard A.C., Pasquier F., Rollin-Sillaire A., Martinaud O., Quillard-Muraine M., de la Sayette V., Boutoleau-Bretonniere C., Etcharry-Bouyx F., Chauviré V., Sarazin M., le Ber I., Epelbaum S., Jonveaux T., Rouaud O., Ceccaldi M., Félician O., Godefroy O., Formaglio M., Croisile B., Auriacombe S., Chamard L., Vincent J.L., Sauvée M., Marelli-Tosi C., Gabelle A., Ozsancak C., Pariente J., Paquet C., Hannequin D., Campion D. APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: a genetic screening study of familial and sporadic cases. PLoS Med. 2017;14:1–16. doi: 10.1371/journal.pmed.1002270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee C.Y.D., Landreth G.E. The role of microglia in amyloid clearance from the AD brain. J. Neural. Transm. 2010;117:949–960. doi: 10.1007/s00702-010-0433-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lehmann M.L., Cooper H.A., Maric D., Herkenham M. Social defeat induces depressive-like states and microglial activation without involvement of peripheral macrophages. J. Neuroinflammation. 2016;13:1–19. doi: 10.1186/s12974-016-0672-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leonard B.E. The concept of depression as a dysfunction of the immune system. Curr. Immunol. Rev. 2010;6:205–212. doi: 10.2174/157339510791823835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H., Karl T., Garner B. Understanding the function of ABCA7 in Alzheimer's disease. Biochem. Soc. Trans. 2015;43:920–923. doi: 10.1042/BST20150105. [DOI] [PubMed] [Google Scholar]
- Linnartz B., Neumann H. Microglial activatory (immunoreceptor tyrosine-based activation motif)- and inhibitory (immunoreceptor tyrosine-based inhibition motif)-signaling receptors for recognition of the neuronal glycocalyx. Glia. 2013;61:37–46. doi: 10.1002/glia.22359. [DOI] [PubMed] [Google Scholar]
- Liu Y.-Z., Wang Y.-X., Jiang C.-L. Inflammation: the common pathway of stress-related diseases. Front. Hum. Neurosci. 2017;11:316. doi: 10.3389/fnhum.2017.00316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loeffler D.A., Camp D.M., Conant S.B. Complement activation in the Parkinson's disease substantia nigra: an immunocytochemical study. J. Neuroinflammation. 2006;3:29. doi: 10.1186/1742-2094-3-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madalena K.M., Lerch J.K. The effect of glucocorticoid and glucocorticoid receptor interactions on brain, spinal cord, and glial cell plasticity. Neural Plast. 2017;2017 doi: 10.1155/2017/8640970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Majumdar A., Capetillo-Zarate E., Cruz D., Gouras G.K., Maxfield F.R. Degradation of Alzheimer's amyloid fibrils by microglia requires delivery of ClC-7 to lysosomes. Mol. Biol. Cell. 2011;22:1664–1676. doi: 10.1091/mbc.E10-09-0745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malik M., Parikh I., Vasquez J.B., Smith C., Tai L., Bu G., LaDu M.J., Fardo D.W., Rebeck G.W., Estus S. Genetics ignite focus on microglial inflammation in Alzheimer's disease. Mol. Neurodegener. 2015;10:52. doi: 10.1186/s13024-015-0048-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malik M., Simpson J.F., Parikh I., Wilfred B.R., Fardo D.W., Nelson P.T., Estus S. CD33 Alzheimer's risk-altering polymorphism, CD33 expression, and exon 2 splicing. J. Neurosci. 2013;33:13320–13325. doi: 10.1523/JNEUROSCI.1224-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mander P., Brown G.C. Activation of microglial NADPH oxidase is synergistic with glial iNOS expression in inducing neuronal death: a dual-key mechanism of inflammatory neurodegeneration. J. Neuroinflammation. 2005;2:20. doi: 10.1186/1742-2094-2-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maphis N., Xu G., Kokiko-Cochran O.N., Jiang S., Cardona A., Ransohoff R.M., Lamb B.T., Bhaskar K. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–1755. doi: 10.1093/brain/awv081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin E., Boucher C., Fontaine B., Delarasse C. Distinct inflammatory phenotypes of microglia and monocyte-derived macrophages in Alzheimer's disease models: effects of aging and amyloid pathology. Aging Cell. 2017;16:27–38. doi: 10.1111/acel.12522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matcovitch-Natan O., Winter D.R., Giladi A., Vargas Aguilar S., Spinrad A., Sarrazin S., Ben-Yehuda H., David E., Zelada González F., Perrin P., Keren-Shaul H., Gury M., Lara-Astaiso D., Thaiss C.A., Cohen M., Bahar Halpern K., Baruch K., Deczkowska A., Lorenzo-Vivas E., Itzkovitz S., Elinav E., Sieweke M.H., Schwartz M., Amit I. Microglia development follows a stepwise program to regulate brain homeostasis. Science. 2016;353 doi: 10.1126/science.aad8670. aad8670. [DOI] [PubMed] [Google Scholar]
- Matsuoka Y., Picciano M., La Francois J., Duff K. Fibrillar beta-amyloid evokes oxidative damage in a transgenic mouse model of Alzheimer's disease. Neuroscience. 2001;104:609–613. doi: 10.1016/s0306-4522(01)00115-4. [DOI] [PubMed] [Google Scholar]
- McCormick C.M., Hodges T.E. Stress, glucocorticoids, and brain development in rodent models. In: Fink G., editor. Stress: Neuroendocrinology and Neurobiology. Academic Press; San Diego: 2017. pp. 197–206. [DOI] [Google Scholar]
- McDonald D.R., Brunden K.R., Landreth G.E. Amyloid fibrils activate tyrosine kinase-dependent signaling and superoxide production in microglia. J. Neurosci. 1997;17:2284–2294. doi: 10.1523/JNEUROSCI.17-07-02284.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McEwen B.S. Stressed or stressed out: what is the difference? J. Psychiatry Neurosci. 2005;30:315–318. [PMC free article] [PubMed] [Google Scholar]
- McEwen B.S. Allostasis and allostatic load: implications for neuropsychopharmacology. Neuropsychopharmacology. 2000;22:108–124. doi: 10.1016/S0893-133X(99)00129-3. [DOI] [PubMed] [Google Scholar]
- McEwen B.S., Seeman T. Protective and damaging effects of mediators of stress. Elaborating and testing the concepts of allostasis and allostatic load. Ann. N. Y. Acad. Sci. 1999;896:30–47. doi: 10.1111/j.1749-6632.1999.tb08103.x. [DOI] [PubMed] [Google Scholar]
- McEwen B.S., Wingfield J.C. Encyclopedia of Stress. Elsevier; 2007. Allostasis and allostatic load*; pp. 135–141. [Google Scholar]
- McKim D.B., Weber M.D., Niraula A., Sawicki C.M., Liu X., Jarrett B.L., Ramirez-Chan K., Wang Y., Roeth R.M., Sucaldito A.D., Sobol C.G., Quan N., Sheridan J.F., Godbout J.P. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol. Psychiatr. 2017 doi: 10.1038/mp.2017.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meaney M.J., Sapolsky R.M., McEwen B.S. The development of the glucocorticoid receptor system in the rat limbic brain. II. An autoradiographic study. Brain Res. 1985;350:165–168. doi: 10.1016/0165-3806(85)90260-3. [DOI] [PubMed] [Google Scholar]
- Melchior B., Garcia A.E., Hsiung B.-K., Lo K.M., Doose J.M., Thrash J.C., Stalder A.K., Staufenbiel M., Neumann H., Carson M.J. Dual induction of TREM2 and tolerance-related transcript, Tmem176b, in amyloid transgenic mice: implications for vaccine-based therapies for Alzheimer's disease. ASN Neuro. 2010;2 doi: 10.1042/AN20100010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michalski B., Fahnestock M. Pro-brain-derived neurotrophic factor is decreased in parietal cortex in Alzheimer's disease. Brain Res. Mol. Brain Res. 2003;111:148–154. doi: 10.1016/s0169-328x(03)00003-2. [DOI] [PubMed] [Google Scholar]
- Milior G., Lecours C., Samson L., Bisht K., Poggini S., Pagani F., Deflorio C., Lauro C., Alboni S., Limatola C., Branchi I., Tremblay M.E., Maggi L. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain Behav. Immun. 2016;55:114–125. doi: 10.1016/j.bbi.2015.07.024. [DOI] [PubMed] [Google Scholar]
- Miller M.W., Sadeh N. Traumatic stress, oxidative stress and post-traumatic stress disorder: neurodegeneration and the accelerated-aging hypothesis. Mol. Psychiatr. 2014;19:1156–1162. doi: 10.1038/mp.2014.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris G.P., Clark I.A., Zinn R., Vissel B. Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol. Learn. Mem. 2013;105:40–53. doi: 10.1016/j.nlm.2013.07.002. [DOI] [PubMed] [Google Scholar]
- Myers B., McKlveen J.M., Herman J.P. Glucocorticoid actions on synapses, circuits, and behavior: implications for the energetics of stress. Front. Neuroendocrinol. 2014;35:180–196. doi: 10.1016/j.yfrne.2013.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nam J.H., Park K.W., Park E.S., Lee Y.B., Lee H.G., Baik H.H., Kim Y.-S., Maeng S., Park J., Jin B.K. Interleukin-13/-4-induced oxidative stress contributes to death of hippocampal neurons in aβ1-42-treated hippocampus in vivo. Antioxidants Redox Signal. 2012;16:1369–1383. doi: 10.1089/ars.2011.4175. [DOI] [PubMed] [Google Scholar]
- Nation D.A., Hong S., Jak A.J., Delano-Wood L., Mills P.J., Bondi M.W., Dimsdale J.E. Stress, exercise, and Alzheimer's disease: a neurovascular pathway. Med. Hypotheses. 2011;76:847–854. doi: 10.1016/j.mehy.2011.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolaides N.C., Kyratzi E., Lamprokostopoulou A., Chrousos G.P., Charmandari E. Stress, the stress system and the role of glucocorticoids. Neuroimmunomodulation. 2015;22:6–19. doi: 10.1159/000362736. [DOI] [PubMed] [Google Scholar]
- Numasawa Y., Yamaura C., Ishihara S., Shintani S., Yamazaki M., Tabunoki H., Satoh J.-I. Nasu-Hakola disease with a splicing mutation of TREM2 in a Japanese family. Eur. J. Neurol. 2011;18:1179–1183. doi: 10.1111/j.1468-1331.2010.03311.x. [DOI] [PubMed] [Google Scholar]
- Nunomura A., Perry G., Pappolla M.A., Wade R., Hirai K., Chiba S., Smith M.A. RNA oxidation is a prominent feature of vulnerable neurons in Alzheimer's disease. J. Neurosci. 1999;19:1959–1964. doi: 10.1523/JNEUROSCI.19-06-01959.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O'Connor D.B., Ferguson E., Green J.A., O'Carroll R.E., O'Connor R.C. Cortisol levels and suicidal behavior: a meta-analysis. Psychoneuroendocrinology. 2016;63:370–379. doi: 10.1016/j.psyneuen.2015.10.011. [DOI] [PubMed] [Google Scholar]
- O'Connor D.B., Green J.A., Ferguson E., O'Carroll R.E., O'Connor R.C. Cortisol reactivity and suicidal behavior: investigating the role of hypothalamic-pituitary-adrenal axis responses to stress in suicide attempters and ideators. Psychoneuroendocrinology. 2017;75:183–191. doi: 10.1016/j.psyneuen.2016.10.019. [DOI] [PubMed] [Google Scholar]
- Ownby R.L., Crocco E., Acevedo A., John V., Loewenstein D. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch. Gen. Psychiatr. 2006;63:530–538. doi: 10.1001/archpsyc.63.5.530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Painter M.M., Atagi Y., Liu C.-C., Rademakers R., Xu H., Fryer J.D., Bu G. TREM2 in CNS homeostasis and neurodegenerative disease. Mol. Neurodegener. 2015;10:43. doi: 10.1186/s13024-015-0040-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parkhurst C.N., Yang G., Ninan I., Savas J.N., Yates J.R., Lafaille J.J., Hempstead B.L., Littman D.R., Gan W.-B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell. 2013;155:1596–1609. doi: 10.1016/j.cell.2013.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng S., Wuu J., Mufson E.J., Fahnestock M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer's disease. J. Neurochem. 2005;93:1412–1421. doi: 10.1111/j.1471-4159.2005.03135.x. [DOI] [PubMed] [Google Scholar]
- Perlmutter L.S., Scott S.A., Barrón E., Chui H.C. MHC class II-positive microglia in human brain: association with Alzheimer lesions. J. Neurosci. Res. 1992;33:549–558. doi: 10.1002/jnr.490330407. [DOI] [PubMed] [Google Scholar]
- Perry V.H., Holmes C. Microglial priming in neurodegenerative disease. Nat. Rev. Neurol. 2014;10:217–224. doi: 10.1038/nrneurol.2014.38. [DOI] [PubMed] [Google Scholar]
- Phillips H.S., Hains J.M., Armanini M., Laramee G.R., Johnson S.A., Winslow J.W. BDNF mRNA is decreased in the hippocampus of individuals with Alzheimer's disease. Neuron. 1991;7:695–702. doi: 10.1016/0896-6273(91)90273-3. [DOI] [PubMed] [Google Scholar]
- Piirainen S., Youssef A., Song C., Kalueff A.V., Landreth G.E., Malm T., Tian L. Psychosocial stress on neuroinflammation and cognitive dysfunctions in Alzheimer's disease: the emerging role for microglia? Neurosci. Biobehav. Rev. 2017;77:148–164. doi: 10.1016/j.neubiorev.2017.01.046. [DOI] [PubMed] [Google Scholar]
- Poliani P.L., Wang Y., Fontana E., Robinette M.L., Yamanishi Y., Gilfillan S., Colonna M. TREM2 sustains microglial expansion during aging and response to demyelination. J. Clin. Invest. 2015;125:2161–2170. doi: 10.1172/JCI77983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prokop S., Miller K.R., Drost N., Handrick S., Mathur V., Luo J., Wegner A., Wyss-Coray T., Heppner F.L. Impact of peripheral myeloid cells on amyloid-β pathology in Alzheimer's disease-like mice. J. Exp. Med. 2015;212:1811–1818. doi: 10.1084/jem.20150479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raber J., Huang Y., Ashford J.W. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol. Aging. 2004;25:641–650. doi: 10.1016/j.neurobiolaging.2003.12.023. [DOI] [PubMed] [Google Scholar]
- Ransohoff R.M., El Khoury J. Microglia in health and disease. Cold Spring Harb. Perspect. Biol. 2015;8 doi: 10.1101/cshperspect.a020560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reul J.M., de Kloet E.R. Anatomical resolution of two types of corticosterone receptor sites in rat brain with in vitro autoradiography and computerized image analysis. J. Steroid Biochem. 1986;24:269–272. doi: 10.1016/0022-4731(86)90063-4. [DOI] [PubMed] [Google Scholar]
- Reul J.M., de Kloet E.R. Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation. Endocrinology. 1985;117:2505–2511. doi: 10.1210/endo-117-6-2505. [DOI] [PubMed] [Google Scholar]
- Rimmerman N., Schottlender N., Reshef R., Dan-Goor N., Yirmiya R. The hippocampal transcriptomic signature of stress resilience in mice with microglial fractalkine receptor (CX3CR1) deficiency. Brain Behav. Immun. 2017;61:184–196. doi: 10.1016/j.bbi.2016.11.023. [DOI] [PubMed] [Google Scholar]
- Rivest S. TREM2 enables amyloid β clearance by microglia. Cell Res. 2015;25:535–536. doi: 10.1038/cr.2015.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roque A., Ochoa-Zarzosa A., Torner L. Maternal separation activates microglial cells and induces an inflammatory response in the hippocampus of male rat pups, independently of hypothalamic and peripheral cytokine levels. Brain Behav. Immun. 2016;55:39–48. doi: 10.1016/j.bbi.2015.09.017. [DOI] [PubMed] [Google Scholar]
- Roth T.L., Zoladz P.R., Sweatt J.D., Diamond D.M. Epigenetic modification of hippocampal Bdnf DNA in adult rats in an animal model of post-traumatic stress disorder. J. Psychiatr. Res. 2011;45:919–926. doi: 10.1016/j.jpsychires.2011.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothman S.M., Herdener N., Camandola S., Texel S.J., Mughal M.R., Cong W.-N., Martin B., Mattson M.P. 3xTgAD mice exhibit altered behavior and elevated Aβ after chronic mild social stress. Neurobiol. Aging. 2012;33 doi: 10.1016/j.neurobiolaging.2011.07.005. 830.e1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rozemuller J.M., van der Valk P., Eikelenboom P. Activated microglia and cerebral amyloid deposits in Alzheimer's disease. Res. Immunol. 1992;143:646–649. doi: 10.1016/0923-2494(92)80050-u. [DOI] [PubMed] [Google Scholar]
- Sadigh-Eteghad S., Sabermarouf B., Majdi A., Talebi M., Farhoudi M., Mahmoudi J. Amyloid-beta: a crucial factor in Alzheimer's disease. Med. Princ. Pract. 2015;24:1–10. doi: 10.1159/000369101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santos L.E., Beckman D., Ferreira S.T. Microglial dysfunction connects depression and Alzheimer's disease. Brain Behav. Immun. 2016;55:151–165. doi: 10.1016/j.bbi.2015.11.011. [DOI] [PubMed] [Google Scholar]
- Sapolsky R.M., Krey L.C., McEwen B.S. Prolonged glucocorticoid exposure reduces hippocampal neuron number: implications for aging. J. Neurosci. 1985;5:1222–1227. doi: 10.1523/JNEUROSCI.05-05-01222.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarlus H., Heneka M.T. Microglia in Alzheimer's disease. J. Clin. Invest. 2017;127:3240–3249. doi: 10.1172/JCI90606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sasaki T., Oga T., Nakagaki K., Sakai K., Sumida K., Hoshino K., Miyawaki I., Saito K., Suto F., Ichinohe N. Developmental expression profiles of axon guidance signaling and the immune system in the marmoset cortex: potential molecular mechanisms of pruning of dendritic spines during primate synapse formation in late infancy and prepuberty (I) Biochem. Biophys. Res. Commun. 2014;444:302–306. doi: 10.1016/j.bbrc.2014.01.024. [DOI] [PubMed] [Google Scholar]
- Satoh J.-I., Kino Y., Yanaizu M., Tosaki Y., Sakai K., Ishida T., Saito Y. Microglia express ABI3 in the brains of Alzheimer's disease and Nasu-Hakola disease. Intractable rare Dis. Res. 2017;6:262–268. doi: 10.5582/irdr.2017.01073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sayre L.M., Zelasko D.A., Harris P.L., Perry G., Salomon R.G., Smith M.A. 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer's disease. J. Neurochem. 1997;68:2092–2097. doi: 10.1046/j.1471-4159.1997.68052092.x. [DOI] [PubMed] [Google Scholar]
- Schafer D.P., Lehrman E.K., Kautzman A.G., Koyama R., Mardinly A.R., Yamasaki R., Ransohoff R.M., Greenberg M.E., Barres B.A., Stevens B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74:691–705. doi: 10.1016/j.neuron.2012.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheuer D.A. Adrenal corticosteroid effects in the central nervous system on long-term control of blood pressure. Exp. Physiol. 2010;95:10–12. doi: 10.1113/expphysiol.2008.045484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider H., Pitossi F., Balschun D., Wagner A., del Rey A., Besedovsky H.O. A neuromodulatory role of interleukin-1beta in the hippocampus. Proc. Natl. Acad. Sci. U. S. A. 1998;95:7778–7783. doi: 10.1073/pnas.95.13.7778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneiderman N., Ironson G., Siegel S.D. Stress and health: psychological, behavioral, and biological determinants. Annu. Rev. Clin. Psychol. 2005;1:607–628. doi: 10.1146/annurev.clinpsy.1.102803.144141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwartz M., Kipnis J., Rivest S., Prat A. How do immune cells support and shape the brain in health, disease, and aging? J. Neurosci. 2013;33:17587–17596. doi: 10.1523/JNEUROSCI.3241-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwarz F., Fong J.J., Varki A. Human-specific evolutionary changes in the biology of siglecs. Adv. Exp. Med. Biol. 2015;842:1–16. doi: 10.1007/978-3-319-11280-0_1. [DOI] [PubMed] [Google Scholar]
- Seib C., Whiteside E., Humphreys J., Lee K., Thomas P., Chopin L., Crisp G., O'Keeffe A., Kimlin M., Stacey A., Anderson D. A longitudinal study of the impact of chronic psychological stress on health-related quality of life and clinical biomarkers: protocol for the Australian Healthy Aging of Women Study. BMC Publ. Health. 2014;14:9. doi: 10.1186/1471-2458-14-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sen S., Duman R., Sanacora G. Serum brain-derived neurotrophic factor, depression, and antidepressant medications: meta-analyses and implications. Biol. Psychiatr. 2008;64:527–532. doi: 10.1016/j.biopsych.2008.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serrano-Pozo A., Frosch M.P., Masliah E., Hyman B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2011;1 doi: 10.1101/cshperspect.a006189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serrano-Pozo A., Mielke M.L., Gómez-Isla T., Betensky R.A., Growdon J.H., Frosch M.P., Hyman B.T. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer's disease. Am. J. Pathol. 2011;179:1373–1384. doi: 10.1016/j.ajpath.2011.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sessa G., Podini P., Mariani M., Meroni A., Spreafico R., Sinigaglia F., Colonna M., Panina P., Meldolesi J. Distribution and signaling of TREM2/DAP12, the receptor system mutated in human polycystic lipomembraneous osteodysplasia with sclerosing leukoencephalopathy dementia. Eur. J. Neurosci. 2004;20:2617–2628. doi: 10.1111/j.1460-9568.2004.03729.x. [DOI] [PubMed] [Google Scholar]
- Shi Q., Chowdhury S., Ma R., Le K.X., Hong S., Caldarone B.J., Stevens B., Lemere C.A. Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice. Sci. Transl. Med. 2017;9 doi: 10.1126/scitranslmed.aaf6295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimizu E., Hashimoto K., Okamura N., Koike K., Komatsu N., Kumakiri C., Nakazato M., Watanabe H., Shinoda N., Okada S., Iyo M. Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol. Psychiatr. 2003;54:70–75. doi: 10.1016/s0006-3223(03)00181-1. [DOI] [PubMed] [Google Scholar]
- Sierra A., Gottfried-Blackmore A., Milner T.A., McEwen B.S., Bulloch K. Steroid hormone receptor expression and function in microglia. Glia. 2008;56:659–674. doi: 10.1002/glia.20644. [DOI] [PubMed] [Google Scholar]
- Sims R., van der Lee S.J., Naj A.C., Bellenguez C., Badarinarayan N., Jakobsdottir J., Kunkle B.W., Boland A., Raybould R., Bis J.C., Martin E.R., Grenier-Boley B., Heilmann-Heimbach S., Chouraki V., Kuzma A.B., Sleegers K., Vronskaya M., Ruiz A., Graham R.R., Olaso R., Hoffmann P., Grove M.L., Vardarajan B.N., Hiltunen M., Nöthen M.M., White C.C., Hamilton-Nelson K.L., Epelbaum J., Maier W., Choi S.-H., Beecham G.W., Dulary C., Herms S., Smith A.A.V.V., Funk C.C., Derbois C., Forstner A.J., Ahmad S., Li H., Bacq D., Harold D., Satizabal C.L., Valladares O., Squassina A., Thomas R., Brody J.A., Qu L., Sánchez-Juan P., Morgan T., Wolters F.J., Zhao Y., Garcia F.S., Denning N., Fornage M., Malamon J., Naranjo M.C.D., Majounie E., Mosley T.H., Dombroski B., Wallon D., Lupton M.K., Dupuis J., Whitehead P., Fratiglioni L., Medway C., Jian X., Mukherjee S., Keller L., Brown K., Lin H., Cantwell L.B., Panza F., McGuinness B., Moreno-Grau S., Burgess J.D., Solfrizzi V., Proitsi P., Adams H.H., Allen M., Seripa D., Pastor P., Cupples L.A., Price N.D., Hannequin D., Frank-García A., Levy D., Chakrabarty P., Caffarra P., Giegling I., Beiser A.S., Giedraitis V., Hampel H., Garcia M.E., Wang X., Lannfelt L., Mecocci P., Eiriksdottir G., Crane P.K., Pasquier F., Boccardi V., Henández I., Barber R.C., Scherer M., Tarraga L., Adams P.M., Leber M., Chen Y., Albert M.S., Riedel-Heller S., Emilsson V., Beekly D., Braae A., Schmidt R., Blacker D., Masullo C., Schmidt H., Doody R.S., Spalletta G., Jr., W T.L., Fairchild T.J., Bossù P., Lopez O.L., Frosch M.P., Sacchinelli E., Ghetti B., Yang Q., Huebinger R.M., Jessen F., Li S., Kamboh M.I., Morris J., Sotolongo-Grau O., Katz M.J., Corcoran C., Dunstan M., Braddel A., Thomas C., Meggy A., Marshall R., Gerrish A., Chapman J., Aguilar M., Taylor S., Hill M., Fairén M.D., Hodges A., Vellas B., Soininen H., Kloszewska I., Daniilidou M., Uphill J., Patel Y., Hughes J.T., Lord J., Turton J., Hartmann A.M., Cecchetti R., Fenoglio C., Serpente M., Arcaro M., Caltagirone C., Orfei M.D., Ciaramella A., Pichler S.S., Mayhaus M., Gu W., Lleó A., Fortea J., Blesa R., Barber I.S., Brookes K., Cupidi C., Maletta R.G., Carrell D., Sorbi S., Moebus S., Urbano M., Pilotto A., Kornhuber J., Bosco P., Todd S., Craig D., Johnston J., Gill M., Lawlor B., Lynch A., Fox N.C., Hardy J.J., ARUK Consortium, Albin R.L., Apostolova L.G., Arnold S.E., Asthana S., Atwood C.S., Baldwin C.T., Barnes L.L., Barral S., Beach T.G., Becker J.T., Bigio E.H., Bird T.D., Boeve B.F., Bowen J.D., Boxer A., Burke J.R., Burns J.M., Buxbaum J.D., Cairns N.J., Cao C., Carlson C.S., Carlsson C.M., Carney R.M., Carrasquillo M.M., Carroll S.L., Diaz C.C., Chui H.C., Clark D.G., Cribbs D.H., Crocco E.A., DeCarli C., Dick M., Duara R., Evans D.A., Faber K.M., Fallon K.B., Fardo D.W., Farlow M.R., Ferris S., Foroud T.M., Galasko D.R., Gearing M., Geschwind D.H.H., Gilbert J.R., Graff-Radford N.R., Green R.C., Growdon J.H., Hamilton R.L., Harrell L.E., Honig L.S., Huentelman M.J., Hulette C.M., Hyman B.T., Jarvik G.P., Abner E., Jin L.-W., Jun G., Karydas A., Kaye J.A., Kim R., Kowall N.W., Kramer J.H., LaFerla F.M., Lah J.J., Leverenz J.B., Levey A.I., Li G., Lieberman A.P., Lunetta K.L., Lyketsos C.G., Marson D.C., Martiniuk F., Mash D.C., Masliah E., McCormick W.C., McCurry S.M., McDavid A.N., McKee A.C., Mesulam M., Miller B.L., Miller C.A., Miller J.W., Morris J.C., Murrell J.R., Myers A.J., O'Bryant S., Olichney J.M., Pankratz V.S., Parisi J.E., Paulson H.L., Perry W., Peskind E., Pierce A., Poon W.W., Potter H., Quinn J.F., Raj A., Raskind M., Reisberg B., Reitz C.C., Ringman J.M., Roberson E.D., Rogaeva E., Rosen H.J., Rosenberg R.N., Sager M.A., Saykin A.J., Schneider J.A., Schneider L.S., Seeley W.W., Smith A.G., Sonnen J.A., Spina S., Stern R.A., Swerdlow R.H., Tanzi R.E., Thornton-Wells T.A., Trojanowski J.Q., Troncoso J.C., Van Deerlin V.M., Van Eldik L.J., Vinters H.H.V.V., Vonsattel J.P., Weintraub S., Welsh-Bohmer K.A., Wilhelmsen K.C., Williamson J., Wingo T.S., Woltjer R.L., Wright C.B., Yu C.-E., Yu L., Garzia F., Golamaully F., Septier G., Engelborghs S., Vandenberghe R., De Deyn P.P., Fernadez C.M., Benito Y.A., Thonberg H., Forsell C., Lilius L., Kinhult-Stählbom A., Kilander L., Brundin R., Concari L., Helisalmi S., Koivisto A.M., Haapasalo A., Dermecourt V., Fievet N., Hanon O., Dufouil C., Brice A., Ritchie K., Dubois B., Himali J.J., Keene C.D., Tschanz J., Fitzpatrick A.L., Kukull W.A., Norton M., Aspelund T., Larson E.B., Munger R., Rotter J.I., Lipton R.B., Bullido M.J., Hofman A., Montine T.J., Coto E., Boerwinkle E., Petersen R.C., Alvarez V., Rivadeneira F., Reiman E.M., Gallo M., O'Donnell C.J., Reisch J.S., Bruni A.C., Royall D.R., Dichgans M., Sano M., Galimberti D.D., St George-Hyslop P., Scarpini E., Tsuang D.W., Mancuso M., Bonuccelli U., Winslow A.R., Daniele A., Wu C.-K., GERAD/PERADES, CHARGE, ADGC E., Peters O., Nacmias B., Riemenschneider M., Heun R., Brayne C., Rubinsztein D.C., Bras J., Guerreiro R., Al-Chalabi A., Shaw C.E., Collinge J., Mann D., Tsolaki M., Clarimón J., Sussams R., Lovestone S., O'Donovan M.C., Owen M.J., Behrens T.W., Mead S., Goate A.M., Uitterlinden A.G., Holmes C., Cruchaga C., Ingelsson M., Bennett D.A., Powell J., Golde T.E., Graff C., De Jager P.L., Morgan K., Ertekin-Taner N., Combarros O., Psaty B.M., Passmore P., Younkin S.G., Berr C., Gudnason V., Rujescu D., Dickson D.W.W., Dartigues J.-F., DeStefano A.L., Ortega-Cubero S., Hakonarson H., Campion D., Boada M., Kauwe J.K., Farrer L.A., Van Broeckhoven C., Ikram M.A., Jones L., Haines J.L., Tzourio C., Launer L.J., Escott-Price V., Mayeux R., Deleuze J.-F., Amin N., Holmans P.A., Pericak-Vance M.A., Amouyel P., van Duijn C.M., Ramirez A., Wang L.-S., Lambert J.-C., Seshadri S., Williams J., Schellenberg G.D.D. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer's disease. Nat. Genet. 2017;49:1373–1384. doi: 10.1038/ng.3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sipe G.O., Lowery R.L., Tremblay M.-È., Kelly E.A., Lamantia C.E., Majewska A.K. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat. Commun. 2016;7 doi: 10.1038/ncomms10905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith M.A., Makino S., Kvetnansky R., Post R.M. Stress and glucocorticoids affect the expression of brain-derived neurotrophic factor and neurotrophin-3 mRNAs in the hippocampus. J. Neurosci. 1995;15:1768–1777. doi: 10.1523/JNEUROSCI.15-03-01768.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith S.M., Vale W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006;8:383–395. doi: 10.31887/DCNS.2006.8.4/ssmith. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solito E., Sastre M. Microglia function in Alzheimer's disease. Front. Pharmacol. 2012;3:14. doi: 10.3389/fphar.2012.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spires-Jones T.L., Hyman B.T. The intersection of amyloid beta and tau at synapses in Alzheimer's disease. Neuron. 2014;82:756–771. doi: 10.1016/j.neuron.2014.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stephan A.H., Madison D.V., Mateos J.M., Fraser D.A., Lovelett E.A., Coutellier L., Kim L., Tsai H.-H., Huang E.J., Rowitch D.H., Berns D.S., Tenner A.J., Shamloo M., Barres B.A. A dramatic increase of C1q protein in the CNS during normal aging. J. Neurosci. 2013;33:13460–13474. doi: 10.1523/JNEUROSCI.1333-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stevens B., Allen N.J., Vazquez L.E., Howell G.R., Christopherson K.S., Nouri N., Micheva K.D., Mehalow A.K., Huberman A.D., Stafford B., Sher A., Litke A.M., Lambris J.D., Smith S.J., John S.W.M., Barres B.A. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–1178. doi: 10.1016/j.cell.2007.10.036. [DOI] [PubMed] [Google Scholar]
- Sugama S., Fujita M., Hashimoto M., Conti B. Stress induced morphological microglial activation in the rodent brain: involvement of interleukin-18. Neuroscience. 2007;146:1388–1399. doi: 10.1016/j.neuroscience.2007.02.043. [DOI] [PubMed] [Google Scholar]
- Sugama S., Takenouchi T., Fujita M., Kitani H., Hashimoto M. Cold stress induced morphological microglial activation and increased IL-1β expression in astroglial cells in rat brain. J. Neuroimmunol. 2011;233:29–36. doi: 10.1016/j.jneuroim.2010.11.002. [DOI] [PubMed] [Google Scholar]
- Suri D., Vaidya V.A. Glucocorticoid regulation of brain-derived neurotrophic factor: relevance to hippocampal structural and functional plasticity. Neuroscience. 2013;239:196–213. doi: 10.1016/j.neuroscience.2012.08.065. [DOI] [PubMed] [Google Scholar]
- Swaab D.F., Raadsheer F.C., Endert E., Hofman M.A., Kamphorst W., Ravid R. Increased cortisol levels in aging and Alzheimer's disease in postmortem cerebrospinal fluid. J. Neuroendocrinol. 1994;6:681–687. doi: 10.1111/j.1365-2826.1994.tb00635.x. [DOI] [PubMed] [Google Scholar]
- Swanwick G.R., Kirby M., Bruce I., Buggy F., Coen R.F., Coakley D., Lawlor B.A. Hypothalamic-pituitary-adrenal axis dysfunction in Alzheimer's disease: lack of association between longitudinal and cross-sectional findings. Am. J. Psychiatr. 1998;155:286–289. doi: 10.1176/ajp.155.2.286. [DOI] [PubMed] [Google Scholar]
- Takahashi K., Rochford C.D.P., Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J. Exp. Med. 2005;201:647–657. doi: 10.1084/jem.20041611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka J., Fujita H., Matsuda S., Toku K., Sakanaka M., Maeda N. Glucocorticoid- and mineralocorticoid receptors in microglial cells: the two receptors mediate differential effects of corticosteroids. Glia. 1997;20:23–37. [PubMed] [Google Scholar]
- Tay T.L., Savage J.C., Hui C.W., Bisht K., Tremblay M.-È. Microglia across the lifespan: from origin to function in brain development, plasticity and cognition. J. Physiol. 2017;595:1929–1945. doi: 10.1113/JP272134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian L., Hui C.W., Bisht K., Tan Y., Sharma K., Chen S., Zhang X., Tremblay M.-E. Microglia under psychosocial stressors along the aging trajectory: consequences on neuronal circuits, behavior, and brain diseases. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2017;79:27–39. doi: 10.1016/j.pnpbp.2017.01.007. [DOI] [PubMed] [Google Scholar]
- Trajkovska V., Vinberg M., Aznar S., Knudsen G.M., Kessing L.V. Whole blood BDNF levels in healthy twins discordant for affective disorder: association to life events and neuroticism. J. Affect. Disord. 2008;108:165–169. doi: 10.1016/j.jad.2007.09.004. [DOI] [PubMed] [Google Scholar]
- Tran T.T., Srivareerat M., Alkadhi K.A. Chronic psychosocial stress accelerates impairment of long-term memory and late-phase long-term potentiation in an at-risk model of Alzheimer's disease. Hippocampus. 2011;21:724–732. doi: 10.1002/hipo.20790. [DOI] [PubMed] [Google Scholar]
- Tsankova N.M., Berton O., Renthal W., Kumar A., Neve R.L., Nestler E.J. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat. Neurosci. 2006;9:519–525. doi: 10.1038/nn1659. [DOI] [PubMed] [Google Scholar]
- van der Flier W.M., Scheltens P. Epidemiology and risk factors of dementia. J. Neurol. Neurosurg. Psychiatry. 2005;76(Suppl. 5):v2–7. doi: 10.1136/jnnp.2005.082867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varki A. Sialic acids in human health and disease. Trends Mol. Med. 2008;14:351–360. doi: 10.1016/j.molmed.2008.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varki A., Angata T. Siglecs–the major subfamily of I-type lectins. Glycobiology. 2006;16:1R–27R. doi: 10.1093/glycob/cwj008. [DOI] [PubMed] [Google Scholar]
- Venegas C., Kumar S., Franklin B.S., Dierkes T., Brinkschulte R., Tejera D., Vieira-Saecker A., Schwartz S., Santarelli F., Kummer M.P., Griep A., Gelpi E., Beilharz M., Riedel D., Golenbock D.T., Geyer M., Walter J., Latz E., Heneka M.T. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer's disease. Nature. 2017;552:355–361. doi: 10.1038/nature25158. [DOI] [PubMed] [Google Scholar]
- Vyas S., Rodrigues A.J., Silva J.M., Tronche F., Almeida O.F.X., Sousa N., Sotiropoulos I. Chronic stress and glucocorticoids: from neuronal plasticity to neurodegeneration. Neural Plast. 2016;2016 doi: 10.1155/2016/6391686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H.-T., Huang F.-L., Hu Z.-L., Zhang W.-J., Qiao X.-Q., Huang Y.-Q., Dai R.-P., Li F., Li C.-Q. Early-life social isolation-induced depressive-like behavior in rats results in microglial activation and neuronal histone methylation that are mitigated by minocycline. Neurotox. Res. 2017;31:505–520. doi: 10.1007/s12640-016-9696-3. [DOI] [PubMed] [Google Scholar]
- Wang H.-X., Wahlberg M., Karp A., Winblad B., Fratiglioni L. Psychosocial stress at work is associated with increased dementia risk in late life. Alzheimer's Dementia. 2012;8:114–120. doi: 10.1016/j.jalz.2011.03.001. [DOI] [PubMed] [Google Scholar]
- Wang J., Xiong S., Xie C., Markesbery W.R., Lovell M.A. Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer's disease. J. Neurochem. 2005;93:953–962. doi: 10.1111/j.1471-4159.2005.03053.x. [DOI] [PubMed] [Google Scholar]
- Wielgat P., Braszko J.J. The participation of sialic acids in microglia-neuron interactions. Cell. Immunol. 2012;273:17–22. doi: 10.1016/j.cellimm.2011.12.002. [DOI] [PubMed] [Google Scholar]
- Wildsmith K.R., Holley M., Savage J.C., Skerrett R., Landreth G.E. Evidence for impaired amyloid β clearance in Alzheimer's disease. Alzheimer's Res. Ther. 2013;5:33. doi: 10.1186/alzrt187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkinson B.L., Landreth G.E. The microglial NADPH oxidase complex as a source of oxidative stress in Alzheimer's disease. J. Neuroinflammation. 2006;3:30. doi: 10.1186/1742-2094-3-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Winkler Z., Kuti D., Ferenczi S., Gulyás K., Polyák Á., Kovács K.J. Impaired microglia fractalkine signaling affects stress reaction and coping style in mice. Behav. Brain Res. 2017;334:119–128. doi: 10.1016/j.bbr.2017.07.023. [DOI] [PubMed] [Google Scholar]
- Wohleb E.S., McKim D.B., Sheridan J.F., Godbout J.P. Monocyte trafficking to the brain with stress and inflammation: a novel axis of immune-to-brain communication that influences mood and behavior. Front. Neurosci. 2015;9:1–17. doi: 10.3389/fnins.2014.00447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wohleb E.S., Terwilliger R., Duman C.H., Duman R.S. Stress-induced neuronal colony stimulating factor 1 provokes microglia-mediated neuronal remodeling and depressive-like behavior. Biol. Psychiatr. 2018;83:38–49. doi: 10.1016/j.biopsych.2017.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woodruff T.M., Ager R.R., Tenner A.J., Noakes P.G., Taylor S.M. The role of the complement system and the activation fragment C5a in the central nervous system. NeuroMolecular Med. 2010;12:179–192. doi: 10.1007/s12017-009-8085-y. [DOI] [PubMed] [Google Scholar]
- Woolley C.S., Gould E., McEwen B.S. Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. 1990;531:225–231. doi: 10.1016/0006-8993(90)90778-a. [DOI] [PubMed] [Google Scholar]
- Yuan P., Condello C., Keene C.D., Wang Y., Bird T.D., Paul S.M., Luo W., Colonna M., Baddeley D., Grutzendler J. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron. 2016;90:724–739. doi: 10.1016/j.neuron.2016.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zanjani H., Finch C.E., Kemper C., Atkinson J., McKeel D., Morris J.C., Price J.L. Complement activation in very early Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2005;19:55–66. doi: 10.1097/01.wad.0000165506.60370.94. [DOI] [PubMed] [Google Scholar]
- Zhao Y., Zhao B. Oxidative stress and the pathogenesis of Alzheimer's disease. Oxid. Med. Cell. Longev. 2013;2013 doi: 10.1155/2013/316523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng H., Jia L., Liu C.-C., Rong Z., Zhong L., Yang L., Chen X.-F., Fryer J.D., Wang X., Zhang Y.-W., Xu H., Bu G. TREM2 promotes microglial survival by activating wnt/β-catenin pathway. J. Neurosci. 2017;37:1772–1784. doi: 10.1523/JNEUROSCI.2459-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhong L., Zhang Z.-L., Li X., Liao C., Mou P., Wang T., Wang Z., Wang Z., Wei M., Xu H., Bu G., Chen X.-F. TREM2/DAP12 complex regulates inflammatory responses in microglia via the JNK signaling pathway. Front. Aging Neurosci. 2017;9:204. doi: 10.3389/fnagi.2017.00204. [DOI] [PMC free article] [PubMed] [Google Scholar]
Further reading
- Andorfer C., Kress Y., Espinoza M., de Silva R., Tucker K.L., Barde Y.-A., Duff K., Davies P. Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms. J. Neurochem. 2003;86:582–590. doi: 10.1046/j.1471-4159.2003.01879.x. [DOI] [PubMed] [Google Scholar]
- Chishti M.A., Yang D.S., Janus C., Phinney A.L., Horne P., Pearson J., Strome R., Zuker N., Loukides J., French J., Turner S., Lozza G., Grilli M., Kunicki S., Morissette C., Paquette J., Gervais F., Bergeron C., Fraser P.E., Carlson G.A., George-Hyslop P.S., Westaway D. Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695. J. Biol. Chem. 2001;276:21562–21570. doi: 10.1074/jbc.M100710200. [DOI] [PubMed] [Google Scholar]
- Hsiao K., Chapman P., Nilsen S., Eckman C., Harigaya Y., Younkin S., Yang F., Cole G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
- Jankowsky J.L., Slunt H.H., Ratovitski T., Jenkins N.A., Copeland N.G., Borchelt D.R. Co-expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomol. Eng. 2001;17:157–165. doi: 10.1016/s1389-0344(01)00067-3. [DOI] [PubMed] [Google Scholar]
- Lambourne S.L., Sellers L.A., Bush T.G., Choudhury S.K., Emson P.C., Suh Y., Wilkinson L.S. Increased tau phosphorylation on mitogen-activated protein kinase consensus sites and cognitive decline in transgenic models for Alzheimer's disease and FTDP-17: evidence for distinct molecular processes underlying tau abnormalities. Mol. Cell Biol. 2005;25:278–293. doi: 10.1128/MCB.25.1.278-293.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mucke L., Masliah E., Yu G.Q., Mallory M., Rockenstein E.M., Tatsuno G., Hu K., Kholodenko D., Johnson-Wood K., McConlogue L. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 2000;20:4050–4058. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oakley H., Cole S.L., Logan S., Maus E., Shao P., Craft J., Guillozet-Bongaarts A., Ohno M., Disterhoft J., Van Eldik L., Berry R., Vassar R. 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;26:10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oddo S., Caccamo A., Shepherd J.D., Murphy M.P., Golde T.E., Kayed R., Metherate R., Mattson M.P., Akbari Y., LaFerla F.M. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
- Radde R., Bolmont T., Kaeser S.A., Coomaraswamy J., Lindau D., Stoltze L., Calhoun M.E., Jäggi F., Wolburg H., Gengler S., Haass C., Ghetti B., Czech C., Hölscher C., Mathews P.M., Jucker M. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006;7:940–946. doi: 10.1038/sj.embor.7400784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richards J.G., Higgins G.A., Ouagazzal A.-M., Ozmen L., Kew J.N.C., Bohrmann B., Malherbe P., Brockhaus M., Loetscher H., Czech C., Huber G., Bluethmann H., Jacobsen H., Kemp J.A. PS2APP transgenic mice, coexpressing hPS2mut and hAPPswe, show age-related cognitive deficits associated with discrete brain amyloid deposition and inflammation. J. Neurosci. 2003;23:8989–9003. doi: 10.1523/JNEUROSCI.23-26-08989.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshiyama Y., Higuchi M., Zhang B., Huang S.-M., Iwata N., Saido T.C., Maeda J., Suhara T., Trojanowski J.Q., Lee V.M.-Y. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337–351. doi: 10.1016/j.neuron.2007.01.010. [DOI] [PubMed] [Google Scholar]