Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer’s disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau “seeds” underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Keywords: Microglia, Tau, Tauopathy, Seeding, Neuroinflammation, Alzheimer’s disease
1. Introduction
Aggregation of misfolded microtubule associated protein tau (MAPT, hereafter “tau”) into abnormal cellular inclusions, both neuronal and glial, defines a molecular class of neurodegenerative disease known as tauopathies, including Alzheimer’s disease (AD), frontotemporal lobar degeneration tau (FTLD-tau), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), among others. Tau aggregates are characterized by stable morphologies (Holmes et al., 2014), differential cell tropism (Guo et al., 2016), and distinct tau proteoforms (Dujardin et al., 2020) which vary across different tauopathies. For example, the neurofibrillary tangle (NFT), an intraneuronal aggregate of misfolded tau with a flame or globose shape, is a prominent hallmark of AD and PSP (Kovacs, 2015). It is well established that aberrant post-translational modifications (predominantly hyperphosphorylation, but also acetylation, methylation, and other modifications) and/or mutations (e.g., P301S) cause tau to misfold and form aggregates. Misfolded monomeric tau aggregates to form dimers, trimers, multimers, and eventually large filamentous or fibrillar aggregates (e. g. NFTs) which accumulate within the cell. The mechanistic relationship between tau inclusions and the clinical presentation in tauopathies is still unclear. Nevertheless, there is a strong correlative relationship between the development of tau pathology in specific neuroanatomical regions and the functional deficits observed in tauopathy patients.
The phenomenon that tau lesions affect brain regions sequentially in a stereotypic progression suggests that affected brain areas are selectively vulnerable to tau pathology, or that tau pathology spreads from one brain regions to next (Mudher et al., 2017). Numerous studies support the hypothesis that tau spreads in a prion-like manner, often referred to as “tau seeding and spreading” (Clavaguera et al., 2013, 2009; Falcon et al., 2015; Frost et al., 2009; Iba et al., 2015; Kaufman et al., 2016; Wu et al., 2016). “Seeding” refers to the ability of certain conformers or strains of misfolded tau to enter recipient cells and trigger multimerization and conformational conversion of native intracellular tau, and “spreading” intercellular propagation of these tau seeds. In vitro studies have confirmed that tau can travel transynaptically from neuron to neuron (Takeda et al., 2015). In vivo mouse models also show that tau aggregates injected into the mouse brain can seed tau aggregation in connected regions (Iba et al., 2013); viral gene transfer of tau in mice also leads to rapid spreading of tau to connected neurons(Asai et al., 2015; Wegmann et al., 2015). Furthermore, expression of a human tau transgene in entorhinal cortex neurons of the mouse leads to propagation of tau aggregates into the synaptically connected hippocampus (de Calignon et al., 2012). Studies with human postmortem brain tissue show that tau seeds precede formation of NFTs along the Braak tau pathway, also suggesting that tau propagates in humans (DeVos et al., 2018a). Neuronal tau release and uptake is considered the putative mechanism of tau propagation (de Calignon et al., 2012; Liu et al., 2012; Takeda et al., 2015; Wegmann et al., 2015) and released tau in the extracellular space can be found as both a free protein and in extracellular vesicles (EVs) (Brunello et al., 2020). Notably, glia are also involved in tau seeding and spreading (Asai et al., 2015; Ferrer et al., 2019; Hopp et al., 2018; Wang and Ye, 2020; Zareba-Paslawska et al., 2021).
Microglia are the resident immune cells of the central nervous system and are derived from early myeloid progenitors that migrate to the brain during development (Ginhoux et al., 2010). Microglia constantly survey the microenvironment, and rapidly respond to injury (Nimmerjahn et al., 2005) as well as changes in neuronal activity (Liu et al., 2019; Umpierre et al., 2020). Microglia express numerous transmembrane receptors and ion channels to sense and respond to microenvironment alterations (Kettenmann et al., 2011). Microglia display a range of phenotypes or functional states which are defined by their transcriptomic signature, via expression of specific proteins, or sometimes a distinct cellular morphology. The microglial homeostatic phenotype is characterized by immune-surveillance and tightly regulated inflammatory functions which allow microglia to maintain tissue homeostasis without exacerbating the damage themselves. A progressive transition away from a microglia homeostatic phenotype and toward a dysregulated reactive phenotype correlates with the progression of cognitive deficits during tauopathies (Arriagada et al., 1992; Gerhard et al., 2006, 2004; Pascoal et al., 2021). Genetic polymorphisms that modify microglia phenotype and function are common in AD (Karch and Goate, 2015) and may represent a shared etiology amongst tauopathies. Microglia may contribute to disease progression by secretion of inflammatory factors that directly and indirectly promote neurodegeneration and protein aggregation (Cameron and Landreth, 2010). However, emerging literature suggests that microglia senescence or dampened responsiveness rather than an over-reactive response could be a contributing factor to neurodegeneration during tauopathy (Angelova and Brown, 2019). Surprisingly, microglia can also directly contribute to the progression of tau pathology by transmitting aggregate-forming tau seeds intercellularly (Asai et al., 2015; Clayton et al., 2021; Hopp et al., 2018). Conversely, microglia can also reduce tau seeding activity, which would suggest that microglia may potentially be harnessed therapeutically to restrict tau spread (Andersson et al., 2019; Hopp et al., 2018; Luo et al., 2015; Majerova et al., 2014). Cumulatively, these data establish microglia as both friend and foe during tauopathy, capable of restricting or promoting tau spread. With this complex duality in mind, the overall goal of this review is to describe how interactions between microglia and tau aggregates in AD and other tauopathies contribute to disease progression, and to demonstrate how the pivotal bidirectional interaction between microglia and tau facilitates the pathogenesis, progression, and possibly even treatment of tauopathy.
2. Progression of tau pathology is related to microglia phenotype
Tauopathies are broadly divided as either “primary”, where tau aggregates are the predominant neuropathological lesion (example: PSP), or “secondary”, where tau aggregates are observed as a co-pathology alongside another protein aggregate (example: AD) (Irwin, 2016). The former is often grouped together using the umbrella term FTLD-tau. There are many other tauopathies that are discussed in greater detail elsewhere (Kovacs, 2015; Orr et al., 2017). Typically, primary tauopathies result in behavioral and personality disturbances, cognitive impairment and/or extrapyramidal motor dysfunction. PSP and CBD often manifest as atypical Parkinsonian disorders. The progression of symptomatology correlates with the neuroanatomical distribution and burden of tau pathology which occurs in stereotypical spreading patterns, operationally defined through postmortem histopathology staging systems. For AD, Braak NFT staging classifies the progression of tau deposits from the entorhinal cortex (Braak stage I/II) to limbic regions (Braak stage III/IV) to neocortical areas (Braak stage V/VI) (Braak and Braak, 1991). For PSP, a proposed staging schema suggests six stages in which tau pathology progresses from the globus pallidus, subthalamic nuclei, and/or striatum to the frontal cortex, cerebellum, and occipital lobe while also considering whether the tau inclusions are localized to neurons, oligodendrocytes, or astrocytes (Kovacs et al., 2020). For CBD, a staging system does not exist, although generally pathology initially affects the dorsolateral prefrontal cortex and basal ganglia circuits, progressing to posterior regions later in the disease (Constantinides et al., 2019). In AD, tau-immunoreactive NFTs but not Aβ-positive plaques, positively correlate with dementia severity (Arriagada et al., 1992; Bejanin et al., 2017; Schöll et al., 2016). Dementia severity and cognitive domain specific impairment also associate with Braak NFT stage, especially in middle-to-late stage transition from medial temporal and limbic brain regions to diffuse neocortical distribution (Nelson et al., 2012). Other tauopathies, such as PSP, similarly show negative correlations between global cognitive and executive functioning with total tau burden (Koga et al., 2017). Alternatively, the NFTs and other tau aggregates themselves may not be the direct cause of neurodegeneration and cognitive decline, and some have hypothesized that NFTs could instead be a sign of protective sequestering of toxic tau (Bretteville and Planel, 2008). The species of tau responsible for neurodegeneration is still largely debated, but there is growing consensus that soluble, intermediate tau multimers (often termed “oligomers”) drive neurotoxicity (Flach et al., 2012; Fox et al., 2011; Frost et al., 2014; Gerson et al., 2014; Lasagna-Reeves et al., 2011; Niewiadomska et al., 2021; Sun et al., 2018). Overall, these data demonstrate that tau burden is associated with functional decline and generally proceeds through brain regions sequentially.
The stereotypical anatomical progression of different tauopathies lead to two competing hypotheses: (1) affected brain regions and cell subpopulations are selectively vulnerable to tau pathology and (2) that tau pathology spreads from one interconnected brain regions to another (Mudher et al., 2017). It is likely that the progression of tau pathology results from a combination of both selective vulnerability and tau seeding and spreading at different disease stages. A recent study showed that tau spreading drives early tau propagation in early-stage AD but that local replication drives late tau progression (Meisl et al., 2021). The specific factors involved in either selective regional vulnerability or tau spreading are not fully characterized, although several lines of data point to microglia as potential cellular mediators of vulnerability to and spread of tau pathology. Histopathological and positron emission tomography (PET) neuroimaging studies demonstrate that regional changes in microglia phenotype correlate with cognitive decline and/or tau pathology in AD and other tauopathies (Cherry et al., 2016; Gerhard et al., 2006, 2004; Minett et al., 2016; Parachikova et al., 2007; Pascoal et al., 2021; Perez-Nievas et al., 2013; Sanchez-Mejias et al., 2016), while RNA sequencing studies define specific molecular changes in microglia phenotype in humans as well as mouse models of tauopathy (Rexach et al., 2020; H. Wang et al., 2018).
2.1. Histopathological characterization of microglia phenotype in tauopathy
Several histopathological microglia markers are associated with the progression of tau pathology in human patients. Minett and colleagues found that higher levels of CD68 (a marker of phagocytic microglia), macrophage scavenger receptor, and Fcγ receptor I (FcγR, also known as CD64) and lower levels of ionized calcium binding adaptor molecule 1 (Iba1) were associated with worsened cognition as measured by the mini mental state exam (MMSE) (Minett et al., 2016), while Parachikova and colleagues found that higher levels of major histocompatibility complex class II (MHC-II) was associated with worsened cognition by the MMSE (Parachikova et al., 2007). Similarly, Cherry and colleagues found that CD68 + cell density could predict diagnosis of dementia in cases with repetitive head trauma (Cherry et al., 2016). Perez-Nievas and colleagues examined non-demented individuals without probable AD pathology, non-demented individuals with intermediate and high probably AD pathology (“mismatch” cases), and demented individuals with probable AD pathology to identify factors that could provide resilience to cognitive deficits caused by accumulation of amyloid β (Aβ) plaques and tau tangles. They found that mismatch cases had significantly fewer CD68-positive microglia in addition to lower levels of hyperphosphorylated tau in synapses (Perez-Nievas et al., 2013). These data are in line with data from Parachikova and colleagues showing that mismatch cases in their dataset also have lower levels of MHC-II-positive microglia compared to AD cases with cognitive deficits (Parachikova et al., 2007), since both CD68 and MHC-II are traditionally considered markers of pro-inflammatory microglia activation. Histopathological studies that focused on microglial morphological characterization rather than expression of inflammatory markers yielded very different results (Sanchez-Mejias et al., 2016; Streit et al., 2009, 2014). Indeed, these authors found very few microglia exhibiting the hypertrophic morphology characteristic of activated microglia, and instead an abundance of dystrophic microglia with fragmented cytoplasm (cytorrhexis), spheroids, and a reduced area coverage (Sanchez-Mejias et al., 2016; Streit et al., 2009, 2014). These dystrophic microglia are thought to be hypofunctional and possibly senescent rather than activated (Sanchez-Mejias et al., 2016; Streit et al., 2009, 2014). Interestingly, one of these groups observed that dystrophic microglia appear in brain regions prior to the spread of NFTs based on the stereotypical anatomical progression of AD (Streit et al., 2009). As the authors point out, this would suggest that loss of microglial protective function in a brain region renders it more susceptible to tau pathology (Streit et al., 2009). Though these data seem to present a paradoxical role of microglia in neurodegenerative disease, it is likely that each story presents a different facet of a multidimensional cell. Indeed, there is a growing consensus that both pro-inflammatory and hypofunctional microglia contribute to tau pathology and neurodegeneration in different ways (Angelova and Brown, 2019; Streit et al., 2014).
2.2. Transcriptomic characterization of microglia phenotype in tauopathy
The transcriptomic signature of microglia during neurodegenerative diseases is reviewed elsewhere (Deczkowska et al., 2018). Briefly, microglia display a unique homeostatic transcriptomic signature (Butovsky et al., 2014) that transitions to a “primed” or “neurodegenerative” signature during aging, AD, tauopathy, and other neurodegenerative processes such as amyotrophic lateral sclerosis referred to as “disease associated microglia” (DAM) or “neurodegenerative microglia” (MGnD) (Butovsky et al., 2015; Friedman et al., 2018; Holtman et al., 2015; Keren-Shaul et al., 2017; Krasemann et al., 2017); notably, this microglia phenotype is unique from that which is induced by an acute inflammatory insult such as lipopolysaccharide (LPS) (Holtman et al., 2015). The homeostatic microglial transcriptomic signature is characterized by minimal expression of pro-inflammatory cytokines and high expression of immune surveillance and clearance pathways (e. g. P2RY12 and growth-arrest specific gene 6 (Gas6), respectively) (Butovsky et al., 2014; Schuh et al., 2014). The low expression of pro-inflammatory cytokines is a sign that immune activation is resolved following clearance of the immunological insult (e. g. cell debris) (Hanisch and Kettenmann, 2007). Contrastingly, MGnDs/DAMs down-regulate immune checkpoint and “homeostatic” genes (e. g. CX3CR1 and P2RY12) and upregulate stress-related, inflammatory, phagocytic, and lipid homeostasis genes (Keren-Shaul et al., 2017; Krasemann et al., 2017)including genes that are known AD risk factors (Efthymiou and Goate, 2017; Pimenova et al., 2018). Most notably, the TREM2-APOE pathway serves as a key regulator in DAM phenotype conversion (Krasemann et al., 2017). Transcriptomic studies like those by Keren-Shaul et. al. and Krasemann et. al., pave the way for research aimed at understanding how diverse microglial subtypes interact with tau pathology.
RNA sequencing reveals a complex evolution of changing microglia phenotypes throughout tauopathy pathological stages that is reflective of changes observed in histopathological studies. Wang and colleagues recently found that rTg4510 P301L mice display little phosphorylated tau at 2 months of age and had the same number of Iba1 + microglia as WT animals at 2 months of age (H. Wang et al., 2018). In a principle component analysis of the transcriptomic data, microglia from the 2-month old rTg4510s clustered closely with WT microglia indicating that their phenotypes are undiscernible at this stage (H. Wang et al., 2018) during which these models exhibit hyperphosphorylated tau but not NFTs (Santacruz et al., 2005). A clear microglial phenotypic difference emerged at 4 months of age (H. Wang et al., 2018) coinciding with the onset of NFT pathology (Santacruz et al., 2005). They observed early changes in genes related to tumor necrosis factor (TNF) signaling, interleukin (IL)-1α and IL-1β, and colony stimulating factor 1 (CSF1) that remained elevated at all timepoints, as well as changes in AD risk genes, phagocytosis genes, complement components, scavenger receptors, and microglia polarization genes at different timepoints (H. Wang et al., 2018). Rexach and colleagues used this signature to bioinformatically identify two competing tauopathy microglia expression modules: immune activation and immune suppression (Rexach et al., 2020); these modules are also present in human AD, FTD, and PSP transcriptomic datasets. Rexach and colleagues posit that immune activation begins early during tauopathy in microglia via extracellular detection of damage signals via toll-like receptor (TLR) and NLRP3 inflammasome signaling, while immune suppression is activated later in microglia via type 1 interferon (IFN) signaling (e.g., IFN-β) with subsequent type 2 IFN response (e.g., IFN-γ) (Rexach et al., 2020). Thus, the two counteracting phenotypes cause a state of immune dysregulation where the normal homeostatic clearance functions of microglia are increasingly suppressed. Overall, these findings support observations from histopathological microglia phenotyping by confirming that there are disease-stage specific changes in microglia, progressing from pro-inflammatory activation of microglia to suppression of microglia function.
Curiously, the immune suppression microglia phenotype is characterized by upregulation of viral response genes (Rexach et al., 2020). As noted by Rexach and colleagues, studies show that tau induces transposable element expression through chromatin relaxation and PIWI-interacting RNA depletion (Guo et al., 2018; Sun et al., 2018). Endogenous retroviral double stranded RNA (dsRNA), a product of transposable element transcription, is known to drive a type of IFN response (Chuong et al., 2016). So, Rexach and colleagues hypothesize that dsRNA serves as the upstream signal for the immune suppression phenotype (Rexach et al., 2020). Considering extracellular tau itself is thought to activate microglia (Pampuscenko et al., 2020; Stancu et al., 2019), this paradigm provides a rationale for seemingly paradoxical effect of tau on microglial phenotype. Thus, perhaps blockade of type 1 IFN signaling in microglia would remove the breaks on microglial activation and promote immune clearance of pathological substrates. However, inhibition of immune suppression could prove dangerous, as it is well established that chronically activated microglia are neurotoxic (Banati et al., 1993). Though intriguing, the hypothesis presented by Rexach and colleagues requires more experimental rather than correlative evidence. Future studies may aim to determine whether microglial phenotype dysregulation in rTg4510 mice could be rescued by inhibition of the IFN response. These data illustrate the concept that microglia are both “friend and foe” in tauopathies, as both too little and too much action by microglia can be damaging to neurons.
2.3. Genetic risk variants for tauopathy regulate microglia function
In addition to transcriptomic changes in microglia phenotype associated with tauopathy, there are several variants in genes associated with microglia that modulate risk for tauopathies (Pimenova et al., 2017). The well-known AD risk factor, apolipoprotein E (ApoE), regulates microglia phenotype during AD (Krasemann et al., 2017) and the ApoE4 risk genotype is correlated with high tau seeding activity (Dujardin et al., 2020). Moreover, both microglia and microglial ApoE are necessary for tau-mediated neurodegeneration in mice (Shi et al., 2019). ApoE’s immunological function in microglia is also linked to the function of the gene encoding triggering receptor expressed on myeloid cells 2 (Trem2) (Shi and Holtzman, 2018), another microglia-specific gene harboring AD risk variants (Guerreiro et al., 2013) that influences tau burden in tauopathy mice (Gratuze et al., 2020). Together, microglial ApoE and Trem2 are thought to interact to transition microglia from a homeostatic state to a neurodegenerative disease phenotype (Krasemann et al., 2017). The gene encoding phospholipase C gamma 2 (PLCγ2) is also linked to ApoE/Trem2 signaling and a P522R mutation in this gene has been associated with reduced late onset AD (LOAD) risk (Andreone et al., 2020; Conway et al., 2018; Sims et al., 2017). Curiously, one study reported that the same PLCG2 variant was positively associated with PSP though these data lacked statistical power (Conway et al., 2018). Larger validation studies would be highly valuable given recent experimental evidence that shows Aβ pathology-dependent roles of Trem2 signaling on tau pathology (Gratuze et al., 2021). Bridging integrator 1 (BIN1), the second most prevalent LOAD risk locus behind APOE (Seshadri et al., 2010), has been shown to promote EV release by microglia (Crotti et al., 2019). Microglia-derived EVs have been implicated in the spread of tau pathology (Asai et al., 2015; Clayton et al., 2021). These are just a few examples of tauopathy risk genes linked to microglial function and phenotype. Overall, while we do not comprehensively understand how these genetic risk factors lead to aggregation and spread of misfolded tau or how this leads to neurodegeneration, microglia clearly play an important role.
2.4. Microglia phenotype in tau PET imaging
PET neuroimaging of neuroinflammation, tau, and Aβ permits assessment of pathologic processes during life. PET-based metrics of neuroinflammation are chiefly derived from microglial marker translocator protein 18 kDa (TPSO otherwise known as peripheral benzodiazepine receptor). Microglia activation, measured via [11C]-PBR28, correlates with cerebrospinal fluid (CSF) TREM2 and other inflammatory biomarkers (Pascoal et al., 2021) that capture the neuroinflammatory phenotype. PET studies on PSP, CBD, and AD cases demonstrate microglia activation generally parallels the distribution of tau deposition (Cagnin et al., 2006, 2001; Gerhard et al., 2006, 2004). Similar to histopathological studies, microglia activation correlates with declining MMSE scores, while amyloid burden does not (Edison et al., 2008). PET studies combining amyloid, tau, and microglia neuroimaging also reflect histopathological studies: increased brain binding of [11C]-PBR28 for activated microglia spatially correlates with [18F]-AV1451 NFT binding more so than [18F]-flutemetamol Aβ plaque binding (Dani et al., 2018). Regression analysis in a recent PET study using [18F]-AZD4694 (Aβ), [18F]-MK-6240 (tau), and [11C]-PBR28 revealed progression of tau pathology depended on the microglia activation network, mirrored Braak NFT staging, and that this relationship is potentiated by Aβ-mediated microglia activation (Pascoal et al., 2021). Importantly, this study demonstrates that microglia may drive tau pathology in human patients. This is supported by earlier studies suggesting that changes in microglia phenotype precede tau pathology in a similar somatotopic pattern in human (López-González et al., 2015) and mouse studies (Jaworski et al., 2011; Yoshiyama et al., 2007). Altogether, these data provide critical support for the hypothesis that microglia influence progression of tau pathology, these descriptive studies cannot fully uncover whether microglia influence the progression of tau pathology via modulation of selective vulnerability or directly via influencing tau pathology.
2.5. Manipulation of microglia phenotype can alter vulnerability to tau pathology
Several experimental studies have demonstrated that proinflammatory microglia can initiate tau pathology (Bhaskar et al., 2010; Ghosh et al., 2013; Jiang et al., 2015; Lee et al., 2015; Li et al., 2003; Maphis et al., 2016, 2015a, 2015b; Quintanilla et al., 2004). Pro-inflammatory activation of microglia with LPS induces tau hyperphosphorylation via IL-1 dependent mechanisms in WT rodent neurons in vivo and in vitro (Bhaskar et al., 2010; Lee et al., 2015; Li et al., 2003). Interestingly, IL-1β overexpression significantly increased tau hyperphosphorylation in 3xTgAD mice with P301L tau, even though Aβ plaque pathology was suppressed by 70–80 % and microglia plaque coverage was increased (Ghosh et al., 2013). Additionally, adoptive transfer of microglia from human tau transgenic mice lacking the anti-inflammatory fractalkine receptor (CX3CR1) into wild-type (WT) mice induces tau pathology in recipient mice, which is reduced with anti-IL1β treatment (Maphis et al., 2015a). Overall, these data demonstrate an important role for activated microglia in the initiation of tau pathology, particularly via pro-inflammatory IL-1β cytokine signaling. Conversely, tau pathology is ameliorated when neuroinflammation is suppressed or reduced by manipulating microglia phenotype via suppression of TNFα in 3xTgAD mice (Gabbita et al., 2015), or blockade of IL-1 signaling in 3xTgAD mice (Kitazawa et al., 2011). Additionally, Mapt knockout neurons and mice are protected from LPS induced neuroinflammatory toxicity in Cx3cr1 knockout mice (Maphis et al., 2015a) suggesting that tau mediates the neuronal toxicity of activated microglia. Overall, these data demonstrate that there is a toxic reciprocal cycle between vulnerability to tau pathology and microglia phenotype.
Over the years, studies examining TREM2 have made it increasingly clearer that TREM2 and the microglial MGnD/DAM phenotype it regulates play context specific roles in AD. In mouse models of Aβ plaque pathology, TREM2 deficiency exacerbates plaque-associated toxicity most likely due to the loss of the protective plaque compaction performed by MGnD/DAM microglia (Jay et al., 2017; Ulrich et al., 2014; Wang et al., 2016; Yuan et al., 2016). But studies using double transgenic tauopathy-TREM2 deficient models suggest a very different and more nuanced role of TREM2 in the context of tau pathology (Bemiller et al., 2017; Gratuze et al., 2020; Jiang et al., 2015; Leyns et al., 2017, 2019; Sayed et al., 2018). Deletion of murine TREM2 (mTREM2) reduces the brain atrophy and microgliosis in P301S mice without affecting tau phosphorylation (Leyns et al., 2017; Sayed et al., 2018). However, 5-month-old P301S mice intracerebrally injected with lentivirus expressing a TREM2 silencing cassette displayed heightened tau phosphorylation and microgliosis at 2 months post-injection (Jiang et al., 2015). This is concordant with what was observed in hemizygous mTREM2 knockout P301S mice (Sayed et al., 2018). Strikingly, TREM2 overexpression in P301S mice, attenuated microglial expression of pro-inflammatory cytokines, neuronal loss, tau phosphorylation, and decline in spatial memory (Jiang et al., 2016). More recently, studies have utilized human TREM2 knock-in mice with either R47H mutation (LOAD risk variant) or common variant crossed with P301S mice (Gratuze et al., 2020; Sayed et al., 2021). Gratuze and colleagues observed that the homozygous R47H-hTREM2;P301S genotype attenuated microglial reactivity, brain atrophy, and expression of the MGnD/DAM signature (Gratuze et al., 2020). Sayed and colleagues reported that the heterozygous R47H-hTREM2;P301S genotype had the opposite effect (Sayed et al., 2021), thus recapitulating what was observed in mTREM2 suppressed (Jiang et al., 2015) and hemizygous knockout mice (Sayed et al., 2018). Together these studies suggest that dampened or haploinsufficient expression of TREM2 exacerbates tauopathies. It is still unclear which downstream signaling mechanisms underpin this phenomenon. Both pro- and anti-inflammatory signaling pathways have been linked to TREM2 activation (Forabosco et al., 2013). In accordance with these observations, transcriptomic studies have teased apart “pro-inflammatory” and “anti-inflammatory” subsets of MGnD/DAM phenotype (Rangaraju et al., 2018). This could partially explain the observations that deletion and overexpression have the same outcome with respect to pro-inflammatory cytokine expression. Curiously, TREM2 hemizygosity has no impact on pro-inflammatory cytokine expression in Aβ plaque models (Ulrich et al., 2014). These context-dependent differences might be explained by the differences in extracellular concentration of putative ligands of TREM2 ligands or other parallel immune receptors that influence microglial phenotype. Studies which focused on tau spread in the context of Aβ pathology reported loss of TREM2-dependent MGnD/DAM signaling exacerbated tau-spread to peri-plaque dystrophic neurites (Gratuze et al., 2021; Leyns et al., 2019). Based on these data one may speculate that the MGnD/DAM phenotype is protective during the early stages of AD by mitigating plaque-associated toxicity and tau spread, but concurrent chronic inflammation has deleterious repercussions on the progression of tau pathology. These studies underscore the importance of further investigating microglia-tau interaction as improper timing of TREM2-targeted treatments could have serious consequences in a patient population.
Despite the experimental findings that show suppressing certain pro-inflammatory signaling pathways ameliorates tau pathology, one should avoid making the generalization that inflammation is intrinsically harmful in tauopathies. Indeed, microglial surveillance and clearance functions are essential in neurodegenerative disease where extracellular protein aggregates and cell debris are rampant. For example, microglial activation and clearance of neuronal TDP-43 protein aggregates was shown to be necessary for recovery of motor neuron function in a reversibly inducible TDP-43 expression model of amyotrophic lateral sclerosis (Spiller et al., 2018). Important to the topic of this review, TDP-43 aggregates can be a feature of secondary tauopathies, including AD (Huang et al., 2020). Further, Rexach and colleagues show that an immune suppression rather than activation phenotypic module is correlated with tau pathological progression (Rexach et al., 2020). Therefore, studies aiming to generate immunomodulatory treatments for tauopathies should consider the importance of preserving the homeostatic immune function of microglia.
Another important microglial phenotype with emerging mechanistic relevance to tauopathies is senescence. Cellular senescence is characterized by cell cycle arrest (Hayflick and Moorhead, 1961) and the senescence-associated secretory phenotype (SASP) (Coppé et al., 2008). As reviewed elsewhere, the number of senescent and/or dystrophic microglia in the brain increases with advanced age and neurodegenerative disease (Angelova and Brown, 2019; Streit et al., 2014). Microglia senescence is characterized by cytorrhexis, shortened processes, reduced motility, reduced phagocytic capacity, and increases in pro-inflammatory cytokines and reactive oxygen species (Angelova and Brown, 2019). Microglia with dystrophic morphology, which may represent senescence, accumulate in brain regions with high tau burden in human AD (Sanchez-Mejias et al., 2016; Streit et al., 2009, 2014). Notably, senescent microglia and astrocytes, characterized by p16INK4A expression, accumulate in P301S tauopathy mice; genetic or pharmacological elimination of senescent microglia (and astrocytes) from these mice reduces tau phosphorylation, neurodegeneration, and cognitive decline (Bussian et al., 2018), although a conflicting study showed that pharmacological elimination of senescent cells in rTg4510 tauopathy mice was protective by eliminating senescent NFT-containing neurons rather than senescent glia (Musi et al., 2018). More recently, it was shown that after ingesting tau aggregate-bearing neurons, SASP microglia secrete tau extracellularly and exhibit reduced phagocytic capacity (Brelstaff et al., 2021). Taken together, these data suggest that tau pathology contributes to microglial senescence, but that microglia senescence also contributes to tau pathology in a feedforward cycle. It is currently unclear what, if any, overlapping characteristics are shared between senescent microglia and MGnD/DAM microglia. A recent transcriptomic study of nuclei from human AD brain identified subpopulations of senescent cells that corresponded with tau pathology, although excitatory neurons but not microglia or other glia expressed markers of senescence (Dehkordi et al., 2021). However, single nuclei transcriptomic studies show poor sensitivity for detection of microglia phenotype in human brain (Thrupp et al., 2020), warranting further investigation into the intersection of the MGnD/DAM phenotype and senescence to determine whether they are distinct or overlapping microglia phenotypes.
3. Microglia internalize tau
Tau is released into the extracellular space of the brain as a mixture of free tau and vesicular tau (Brunello et al., 2020). Several studies have observed microglia take up free tau in vitro (Andersson et al., 2019; Asai et al., 2015; Bolós et al., 2015; Funk et al., 2015; Luo et al., 2015; Majerova et al., 2014; Sanchez-Mejias et al., 2016; Zilkova et al., 2020) and when injected directly into the mouse brain (Bolós et al., 2015). Additionally, microglia can internalize neurons that contain tau in vitro (Sanchez-Mejias et al., 2016). There is also evidence that this uptake occurs with endogenously expressed tau in vivo: tau is observed within microglia in human cases of AD(Bolós et al., 2015; Ghoshal et al., 2001; Odawara et al., 1995), mouse models of tauopathy (Asai et al., 2015; van Olst et al., 2020), and aged Tupaia belangeri (tree shrews) which develop tau pathology naturally with age (Rodriguez-Callejas et al., 2020). Microglia do not express the tau gene Mapt (Hopp et al., 2018; Y. Zhang et al., 2014); however, microglia isolated from the rTg4510 mouse model of tauopathy do contain tau protein(Hopp et al., 2018). Several receptors facilitating tau uptake in neurons and astrocytes have been identified (De La-Rocque et al., 2020; Martini-Stoica et al., 2018; Wang and Ye, 2020), and there may be therapeutic potential in targeting them to halt the spread of tau. There is a relative dearth of information on the mechanisms by which microglia internalize tau. Microglial heparan sulphate proteoglycan (HSPG) (Funk et al., 2015), CX3CR1 (Bolós et al., 2017), and P2RY12 (Das and Chinnathambi, 2021) have all been shown to bind tau, albeit only blockade of CX3CR1 and HSPGs was shown to inhibit uptake of tau. HSPGs have also been shown to mediate tau uptake in neurons (Holmes et al., 2013) and astrocytes (Martini-Stoica et al., 2018). Additionally, low density lipoprotein receptor related protein 1 (LRP1), an important immune regulator in microglia (Chuang et al., 2016), has been shown to mediate tau uptake in neurons (Rauch et al., 2020). Therefore, it will be important to verify experimentally whether microglial LRP1 also mediates tau uptake, as future therapies that target these receptors to block neuronal tau propagation may also affect microglial uptake of tau. Given microglial uptake of tau represents a beneficial clearance mechanism, consideration of microglia-specific tau internalization mechanisms is warranted.
3.1. Phagocytosis
Classical work, such as that of Gey et al. in 1954 and W.H. Lewis in 1931, distinguished phagocytosis (cell “eating”) from pinocytosis (cell “drinking”) (Gey et al., 1954; Lewis, 1931). Yet, in the fields of tauopathy and microglia, the term “phagocytosis” is frequently and inaccurately used to describe microglial uptake of extracellular, soluble tau multimers. The biomechanics of phagocytosis dictate why this process is better suited for uptake of large particles (≥0.5 μm in diameter) such as foreign bodies (e. g. bacteria) or cell debris (e. g. apoptotic bodies) over solutes, including tau multimers. Indeed, phagocytosis initiates upon the cooperative activation of multiple phagocytic receptors which bind to different sites on the surface of the particle directly or indirectly (Rosales and Uribe-Querol, 2017). More receptors are recruited to the phagocytic cup to facilitate the anchoring of the membrane protrusion as the membrane progressively covers the surface of the particle (Jaumouillé and Waterman, 2020). Mathematical modeling complemented by experimental observations indicate that particles with a diameter of 2–3 μm are most efficiently internalized via phagocytosis (Champion et al., 2008). Therefore, microglia utilize phagocytosis to clear apoptotic neurons (Grommes et al., 2007) and prune synapses (Paolicelli et al., 2011) while employing liquid-phase pinocytic mechanisms for uptake of small protein aggregates (Mandrekar et al., 2009). We speculate microglial phagocytosis of tau seeds is restricted to engulfment of tau seed containing synapses and whole neurons, rather than small soluble extracellular aggregates.
Opsonic phagocytosis involves binding of opsins (e. g. complement and immunoglobulin G (IgG)) to a particle before it can be internalized (Flannagan et al., 2012). Immunoreactivity of complement has been observed near Aβ-plaques, dystrophic neurites, neuropil threads, and NFTs in histological sections of AD brains (Loeffler et al., 2008). Synapse pruning by microglia is carried out using the complement system (Stephan et al., 2012). Some have postulated that phagocytosis of tau-containing synapses tagged with complement component 1q (C1q) is a means by which microglia internalize tau (Clayton et al., 2021). Interestingly, C1q is known to interact with LRP1 (Duus et al., 2010), a receptor that has been implicated in the uptake of tau by neurons (De La-Rocque et al., 2020). Another major opsonic receptor is FcγR which mediates phagocytosis of IgG coated “meals” (Flannagan et al., 2012). Both Aβ- and tau-targeting IgG antibodies have been in development as potential immunotherapies to treat AD for over a decade (Cummings et al., 2014). However, it is unlikely that anti-tau monoclonal antibodies (mAbs) targeting a single specific region of tau, such as Semorinemab (which binds the N-terminus of tau), can coat the surface of a tau multimer. Since only large, opsonized particles are internalized via FcγR-mediated phagocytosis, these smaller immune complexes are more likely to be internalized by a different form of FcγR-mediated endocytosis (Huang et al., 2006; Tse et al., 2003).
Apoptotic cell receptors allow phagocytes to clear apoptotic cells while suppressing the immune response (Szondy et al., 2017). Examples include (αvβ3/5 integrin and Axl/Mer which bind to milk-fat globule epidermal growth factor 8 (MFG-E8) and Gas6, respectively (Grommes et al., 2007; Neniskyte and Brown, 2013). Bridging proteins MFG-E8 and Gas6 bind to phosphatidylserines (PS), which are normally found on the inner leaflet of the membrane but are flipped to the outer leaflet in apoptotic cells (Bratton et al., 1997). Thus, PS provide an “eat me” signal which helps microglia distinguish living from dying neurons. Experimental studies suggest that tau-filled living neurons erroneously express these “eat me” signal, and this results in their aberrant phagocytosis by microglia via MFG-E8 bridging protein (Brelstaff et al., 2018, 2021). Intriguingly, microglia displaying a senescence-like phenotype, become hypophagocytic, and secrete tau after ingesting tau filled neurons in vitro (Brelstaff et al., 2021).
3.2. Macropinocytosis
Macropinocytosis is a form of pinocytosis (cellular “drinking”) in which membrane ruffles protrude from the surface and then join to form a nascent macropinosome (diameter ≈0.25–5 μm) (Canton, 2018). Like phagocytosis, macropinocytosis is actin dependent (Canton, 2018). However, unlike phagocytosis, macropinocytic membrane ruffles (e. g. lamellipodia, circular dorsal ruffles) do not adhere to the surface of the engulfed particle(s), and thus the size of the macropinosome is not dependent on the size of the particle (Jaumouillé and Waterman, 2020). In most cell types, macropinocytosis is triggered by some intra- or extracellular stimulus, but it can also occur constitutively in certain myeloid lineage cells (Canton, 2018; Lim and Gleeson, 2011). For example, macrophages (Bohdanowicz et al., 2013; Steinman et al., 1976; Yao et al., 2009), and immature dendritic cells (Garrett et al., 2000; Norbury et al., 1997) employ constitutive macropinocytosis to survey the extracellular milieu (Doodnauth et al., 2019). Homeostatic microglia, like macrophages, serve as sentinels monitoring their local environment through continuous sampling via pinocytosis (Booth and Thomas, 1991; Canton, 2018; Canton et al., 2016; Fitzner et al., 2011; Z. Liu et al., 2020; Mandrekar et al., 2009; Ranson and Thomas, 1991; Redka et al., 2018). In fact, these surveying, ramified microglia are the most highly pinocytic cells in the brain (Booth and Thomas, 1991; Ranson and Thomas, 1991). Such propensity for liquid phase endocytosis, suggests that microglia activate macropinocytosis constitutively like their myeloid kin (macrophages). Further, a recent publication showed that microglial macropinocytosis involves the same signaling pathways that mediate macrophage and dendritic cell macropinocytosis (Z. Liu et al., 2020). Microglial macropinocytosis has been shown to mediate the uptake of soluble Aβ (Mandrekar et al., 2009), and most likely contributes to uptake of soluble tau (Funk et al., 2015). Microglial macropinocytosis has been shown to mediate uptake of oligodendrocyte derived exosomes (Fitzner et al., 2011). Tau containing exosomes have been shown to contribute to tau spread (Asai et al., 2015; Clayton et al., 2021). Further, it has been shown that exosomes permeabilize the endolysosomal compartment, providing tau a means to escape degradation (Polanco et al., 2021). Therefore, it would be valuable to investigate whether neuron-derived exosomes containing tau are also taken up via macropinocytosis. Moreover, considering the fundamental importance of macropinocytosis to myeloid cells, more studies characterizing the role of macropinocytosis in microglia are warranted.
Tau aggregates have been shown to bind to HSPGs on the surface of neurons (Holmes et al., 2013), astrocytes (Martini-Stoica et al., 2018) and microglia (Funk et al., 2015). HSPGs are ubiquitous glycoproteins present on the surface of cells and the constitute an important component of the extracellular matrix and are internalized via macropinocytosis (Sarrazin et al., 2011). Internalization of HSPGs occurs constitutively, so HSPG-mediated tau internalization may be a passive process of tau simply binding to HSPGs and “hitching a ride” to the endosomal compartment (Sarrazin et al., 2011); however, there is evidence that HSPGs can act as a receptor to induce endocytosis of specific ligands (Wittrup et al., 2009). Ultrastructural analysis via electron microscopy images revealed that tau bound to the neuronal surface HSPGs enters the cell via lamellipodia-like projections that form large non-specific vesicles resembling macropinosomes (Holmes et al., 2013). In microglia, one group showed similar lamellipodia-like structures form in response to extracellular tau in vitro (Das et al., 2020; Desale and Chinnathambi, 2021; Uhlemann et al., 2016). The same group also demonstrated that treatment with dietary supplement α-linolenic acid (ALA) mimicked these effects and promoted tau endocytosis by microglia in vitro (Desale and Chinnathambi, 2021). The authors posit that ALA could be used to slow tauopathy progression by promoting microglial tau clearance. But, if ALA indeed induces tau endocytosis via lamellipodia projection and formation of macropinosomes, then it would likely also promote neuronal (Holmes et al., 2013) and astroglial (Martini-Stoica et al., 2018) tau uptake. This underscores the importance of recognizing that microglia, despite being the “phagocytes of the brain,” utilize non-phagocytic forms of endocytosis that overlap with those used by other cells.
3.3. Micropinocytosis
Micropinocytosis, which includes clathrin-mediated, caveolae-dependent, and clathrin/caveolae-independent endocytosis, is a form of endocytosis in which smaller vesicles take up liquid-phase particles through either receptor-mediated or receptor-independent processes (Iversen et al., 2011; Saha et al., 2013; Solé-Domènech et al., 2016). Neuronal micropinocytic mechanisms of tau internalization include clathrin-mediated endocytosis (CME) via muscarinic acetylcholine receptors and LRP1, and clathrin-independent endocytosis via flotillin-dependent and flotillin-independent lipid microdomains (De La-Rocque et al., 2020). To the best of our knowledge, no studies have specifically examined the role of any of these receptors or endocytic mechanisms in microglial uptake of tau. Like neurons, microglia undergo various forms of micropinocytosis (Minami et al., 2012; Solé-Domènech et al., 2016) and express some of the same cell surface receptors (e. g. LRP1) as neurons (Rauch et al., 2020).
Chemokine receptors are internalized either constitutively or upon ligand binding via CME or caveolae-dependent endocytosis (Bonecchi et al., 2010). Bolos and colleagues showed that knockout of microglial fractalkine receptor CX3CR1 reduced microglial tau uptake in vitro and in vivo, and that monomeric tau has high affinity for CX3CR1, albeit reduced with tau phosphorylation by the kinase glycogen synthase 3 β (GSK3β) (Bolós et al., 2017). Further, they showed that endogenous ligand CX3CL1 competitively inhibits binding of monomeric tau to CX3CR1 (Bolós et al., 2017). The biological relevance of CX3CR1 as a microglia receptor of tau is unclear, since neurons, the main producer of tau protein in the brain, more efficiently release tau dimers and multimers compared to tau monomers (Wegmann et al., 2016) and high-molecular weight phosphorylated tau strains are thought to be the main tau strains involved in propagation of tau seeds (Takeda et al., 2015). Nevertheless, the findings of Bolós and colleagues are intriguing considering CX3CR1 activation by endogenous ligand CX3CL1 results in suppression of the immune activation (Biber et al., 2007). Bolós and colleagues also found that coincubation of CX3CL1 and tau monomers reduces binding affinity of tau monomers to CX3CR1 (Bolós et al., 2017) which might suggest that tau monomers bind to the orthosteric site of CX3CR1. Other studies have shown that aggregated tau induces microglial pro-inflammatory activation (Das and Chinnathambi, 2021; Morales et al., 2013). Thus, it would be informative to investigate how tau monomers and aggregates modulate dose-dependent responding of CX3CR1 to CX3CL1. Tau acting as either an antagonist or inverse agonist at this receptor would provide a mechanistic explanation for induction of microglial activation by tau. Future work is needed to determine which receptors bind and promote the internalization of tau and the other downstream pathways are affected, particularly in the context of microglial activation state.
FcγR-mediated phagocytosis is restricted to the engulfment of large, IgG coated particles (Huang et al., 2006; Tse et al., 2003). Smaller antigen-antibody complexes and aggregates of IgG are internalized via FcγR-mediated actin-independent micropinocytic mechanisms, such as CME (Huang et al., 2006; Tse et al., 2003). In mouse brain slice cultures expressing human tau treated with fluorescently labeled 4E6G7 anti-tau mAb, pharmacological inhibition of clathrin but not actin blocked antibody uptake (Congdon et al., 2013). Similar results were observed in primary culture neurons treated with the same antibody (Congdon et al., 2013). Others have found that anti-tau antibodies robustly enhance uptake of small, soluble tau aggregates by microglia (Andersson et al., 2019; Funk et al., 2015; Zilkova et al., 2020) most likely via actin-independent micropinocytosis rather than phagocytosis (Huang et al., 2006; Tse et al., 2003). The same pharmacological inhibitors used in Congdon et. al. could be applied to cultured microglia in the presence of tau antibodies to implicate or disqualify actin-independent micropinocytosis involvement.
The protein product of LOAD risk gene BIN1 has been shown to be a negative regulator of CME in neurons (Calafate et al., 2016). The neuronal isoform of the BIN1 protein (Bin1) is unique in that it contains a clathrin and AP-2-binding (CLAP) domain which has been shown to mediate the interaction between clathrin and its adaptor protein, AP2 (Micheva et al., n.d.; Ramjaun and McPherson, 1998; Slepnev et al., n.d.; Wigge et al., 1997). In an in vitro model of neuronal tau spread, overexpression and knockdown of BIN1 decreased and increased tau spread, respectively (Calafate et al., 2016). Interestingly, the microglial isoform of BIN1 has been shown to promote the release of tau in EVs (Crotti et al., 2019) which may protect it from lysosomal degradation (Polanco et al., 2021). The role of CME and the various clathrin-independent micropinocytic mechanisms have not been explored for microglial uptake of tau. Indeed, CME is implicated in both microglia and tau pathology during AD: PICALM, a clathrin adaptor protein, is localized to both microglia and tau during AD, and polymorphisms in PICALM are risk factors for AD (Ando et al., 2013). Since micropinocytic mechanisms have been identified to mediate neuronal uptake of tau, it will be important to determine whether the same mechanisms in microglia facilitate tau uptake and whether this leads to tau clearance or spread.
4. Microglia release tau seeds
Both mouse and human microglia release seed-competent tau into their conditioned media ex vivo (Hopp et al., 2018), suggesting that microglia release tau that can cause tau aggregation in recipient cells. Tau protein and bioactive tau seeds are still present in conditioned media from microglia isolated from tauopathy mice up to at least 6 days in vitro. This is longer than the previous reported half-life of tau in vitro which ranges from 3- to 20-hours (David et al., 2002; Shimura et al., 2004), indicating (1) that seed competent tau strains are surprisingly long lived within microglia compared to previous in vitro half-life data; (2) that microglia are not fully processing tau into non-toxic forms; and (3) that seed competent tau strains are released by microglia. Microglia may release tau seeds via intracellular tau mediated microglia toxicity (Sanchez-Mejias et al., 2016) leading to release of not-yet-degraded tau seeds. There may be common mechanisms mediating endosomal-lysosomal dysfunction across various amyloid aggregates (Aβ, NFTs, α-synuclein), including but not limited to intracellular vesicle rupture (Flavin et al., 2017; Polanco et al., 2021). Several studies suggest that microglia secrete tau-containing exosomes in vitro (Crotti et al., 2019) and in vivo (Clayton et al., 2021) which may be how microglia propagate spread of tau directly. However, it still unknown whether the microglia-derived exosomes contain seed-competent aggregates, and it is unclear how much tau-containing exosomes contribute to tau spread in vivo. Additionally, microglia secrete factors in addition to tau or tau-containing exosomes that may exacerbate tau propagation in neurons. For example, neurons express a variety of cytokine receptors that trigger specific intracellular signaling cascades that may confer specific vulnerability to the combination of tau and cytokines release by microglia, since cytokines themselves cause tau seeding (Gorlovoy et al., 2009) and phosphorylation (Maphis et al., 2015b).
Exosomes are EVs derived from the endosomal comparment which are used to dispose of waste or send long-distance signals (Colombo et al., 2014). Internalized exosomes are trafficked to the late endosome where they may either by transported to the cell membrane for exocytosis or fuse with the lysosome for degradation (Colombo et al., 2014). However, the endolysosome does not efficiently degrade the exosomes and can rupture following exosome uptake (Polanco et al., 2021). This suggest that exosomal-tau has higher potential for spread than exosome-free tau. Blockade of exosome release and depletion of microglia can mitigate tau spread in mouse models of tau seeding and coadministration of LPS and ATP can stimulate exosome release by microglia (Asai et al., 2015). The authors of this study postulate that LPS stimulation drives ubiquitination via TLR4 activation, ATP activates P2X7 receptor (P2X7R) to promote release of exosomes, and ubiquitinated proteins are incorporated in multivesicular bodies which are later released as exosomes (Asai et al., 2015). Indeed, in the same study they were able to show that LPS priming of microglia led to ubiquitination of tau (Asai et al., 2015), and P2X7R activation has been shown elsewhere to induce exosome release (Dubyak, 2012). More recent work from the same groups has shown that Aβ, a putative TLR4 ligand (Y. Liu et al., 2020), is associated with microglial exosome release in a mouse model of Aβ-plaque pathology and tau spread (Clayton et al., 2021). Thus, the coincidence of extracellular ATP released by stressed neurons and Aβ-plaques in the microenvironment may serve as a trigger for microglial exosome release. Given the recent results from PET imaging of human AD patients demonstrating that microglia drive the progression of tau pathology via potentiation by Aβ (Pascoal et al., 2021), these animal studies suggest a fascinating mechanism by which Aβ and tau pathology synergize via microglia dysfunction.
BIN1, the second most prevalent LOAD risk locus (Seshadri et al., 2010) plays a key role in microglia exosome release (Crotti et al., 2019) in addition to its aforementioned role in CME. BIN1 human isoform 6 is predominantly expressed in microglia and shares 95 % sequence similarity with the murine microglia-predominant variant, murine isoform 2 (Crotti et al., 2019). Human embryonic kidney 293 T (HEK293T) cells transfected with plasmid encoding the sequences of tau and murine isoform 2 resulted in greater release of tau-containing exosomes than HEK293T transfected with tau alone (Crotti et al., 2019). Further, microglia-specific knockout of BIN1 in PS19 mice reduces tau spread in male but not female mice (Crotti et al., 2019). Further studies are warranted to determine the cause of these sex differences. Nevertheless, these findings taken together with the in vitro findings that neuronal BIN1 loss potentiates tau spread (Calafate et al., 2016) presents a divergent role of microglial and neuronal BIN1. Although only the neuronal isoforms of Bin1 expresses a CLAP domain which interacts with the CME machinery (Micheva et al., n.d.; Ramjaun and McPherson, 1998; Slepnev et al., n.d.; Wigge et al., 1997), all isoforms of Bin1 contain a BAR domain which senses membrane curvature and an SH3 domain which interacts with dynamin. This presents evidence of crosstalk between the endocytic pathways of tau internalization and tau-containing exosome release in microglia.
It is important to point out that two recent studies modeling tau propagation in the context of Aβ pathology have reported seemingly contrasting effects of ablating microglia on tau spread (Clayton et al., 2021; Gratuze et al., 2021). Gratuze et. al. reported that ablation of microglia increased tau spread, and Clayton et. al. reported that ablation of microglia reduced tau spread. In both studies, tau pathology was seeded via stereotaxic injection in a discrete brain region and a CSF1 receptor (CSF1R) inhibitor was used to ablate microglia (Clayton et al., 2021; Gratuze et al., 2021). However, there are several differences in their experimental design that could account for these contradictory observations. For instance, Gratuze et. al. injected human AD derived tau aggregates into the hippocampus and cortex of a 5XFAD mouse, whereas Clayton et. al. injected a viral vector driving the expression of P301L tau into the entorhinal cortex of a humanized amyloid precursor protein (APP) knock-in APPNL-G-F mouse. Further, Clayton et. al. used a more CSF1R-selective microglia depleting drug (PLX5562) than the one used by Gratuze et. al. (PLX3397), which also inhibits c-Kit (DeNardo et al., 2011). Notably, the two Aβ pathology mouse models differ in that transgenic 5XFAD mice overexpress human APP and presenilin 1 (PS1) with familial AD mutations under the Thy1 promoter, while the knock-in model, APPNL-G-F, expresses human familial AD mutant APP at lower levels and does not express human PS1. Thus, it is possible that regional, mouse, or pharmacological differences may affect the role that microglia play in tau spread. Also, it should be noted that not all microglia are ablated by CSF1R inhibitor treatment. Indeed, Gratuze et. al. reported that approximately 20 % of microglia survived ablation and 80 % of these treatment-resistant microglia were associated with Aβ-plaques. Ablation-resistant microglia have been characterized via single-cell RNA sequencing and were shown to have a highly similar transcriptomic profile to microglial progenitor cells (Zhan et al., 2020). This explains why microglia rapidly repopulate after withdrawal of CSF1R inhibition (Elmore et al., 2014; Gratuze et al., 2021; Zhan et al., 2020). Curiously, ablation-resistant progenitor-like microglia highly express Mac2, also known as Galectin-3 (Zhan et al., 2020) a purported marker of the plaque-associated microglia implicated in the spread of tau pathology (Clayton et al., 2021). Given this information, one might postulate that these plaque-associated, Mac2-positive microglia implicated in tau spread constitute a population of microglial progenitor-like cells. Future studies are warranted to characterize the transcriptome of the microglial population implicated tau spread to determine if distinct subpopulations of microglia respectively promote and mitigate tau spread. Nevertheless, data from Gratuze et al. highlight an important protective role for microglia and microglial TREM2 in suppression of tau spread. Microglial degradation of tau is likely an important mechanism by which microglia exert protective functions during tauopathy.
5. Microglia degrade tau
Early studies in human tissues suggest that microglia may process tau into specific forms based on immunohistochemical labeling with conformation-specific anti-tau antibodies. For example, the anti-tau antibody Tau-2 labels a subset of microglia-like cells in AD and dementia with Lewy bodies (Odawara et al., 1995) as well as microglia-like cells surrounding ischemic foci (Uchihara et al., 2005, 2004, 2000). The Tau-2 antibody also labels NFTs and neuropil threads in AD tissue (Odawara et al., 1995; Uchihara et al., 2005, 2004, 2000). However, unlike NFTs, microglia-like Tau-2 positive cells did not show labeling with other anti-tau antibodies, were devoid of argyrophilic fibrillar characteristics, and underwent a reversible conformation change upon exposure to detergent (Uchihara et al., 2005, 2004, 2000). Similarly, the anti-tau antibody Tau-66 labels a subset of microglia in AD but not control brains which were not labeled by other anti-tau antibodies such as Tau5 (total tau, aa210–241) (Ghoshal et al., 2001). The Tau-66 antibody also labels diffuse reticulated plaques as well as NFTs and neuronal cell bodies in both a punctate and ribosomal-like pattern. Notably, the ribosomal-like staining is like that which Tau-2 stains in neurons. The Tau-66 antibody recognizes a discontinuous epitope of tau containing residues aa155–244 and aa305–314 (Ghoshal et al., 2001) and is associated with loss of the N- and C- termini in late-stage conformational changes in AD tau pathology (Ibarra-Bracamontes et al., 2020) but does not label PSP or CBD tau pathology (Berry et al., 2004). More recently, Iba1-labeled microglia were found to colocalize with tau-containing structures labeled with phospho-specific (PHF-1 (pS396, pS404), AT100 (pT212, pS214_, Tau396 (pS396), AT180 (pT231), Tau404 (pS404), and AT8 (pS202, pT205)) and total tau antibodies (Tau-1 (aa162–210) and HT7 (aa159–163)) (Bolós et al., 2015). However, colocalization or apposition between microglia and these phospho- and total-tau antibodies was partial (suggestive of active engulfment), whereas the Tau-66 and Tau-2 labeling showed microglia that were filled with cytoplasmic tau (perhaps suggestive of failed degradation). Overall, these observations confirm that microglia internalize tau in vivo, and suggest that microglia may process tau by cleaving at specific epitopes, leading to specific conformational changes recognized by Tau-2 and Tau-66. Currently, little is known about the fragmentation, conformation, phosphorylation, or other post-translational modifications that microglia perform on tau during degradative processing or preparation for release in exosomes and whether these modifications represent an increase or decrease in tau toxicity or propensity for spread.
Several studies have shown that microglia are capable of degrading tau under different conditions in vitro (Andersson et al., 2019; Luo et al., 2015; Majerova et al., 2014). Majerova and colleagues found that heparin-induced recombinant tau oligomers (tau151–391) were internalized and degraded by primary rat microglia, rat primary blood monocyte derived macrophages, immortalized microglia-like mouse BV2 cells, and immortalized macrophage-like mouse TIB67 cells, but macrophages were more efficient than microglia and microglia degradation of tau was only efficient after LPS exposure (Majerova et al., 2014). Luo and colleagues found that human sarkosyl-insoluble tau fibrils from human AD cases were internalized and degraded by primary mouse microglia and that this was enhanced by the MC1 anti-tau antibody in an Fc-dependent manner (Luo et al., 2015). Andersson and colleagues found that rTg4510-derived sarkosyl-insoluble tau was internalized by primary mouse microglia in an Fcγ-dependent fashion. They also found that inhibition of lysosomal acidification in microglia with chloroquine or bafilomycin A1 reduced sarkosyl-insoluble hyperphosphorylated tau degradation, while inhibition of the proteosome did not (Andersson et al., 2019). Lysosomal acidification is important for pH-sensitive proteolytic cleavage. Notably, microglia express a specific cysteine protease, asparaginyl endopeptidase (AEP, otherwise known as legumain) that has been implicated in the formation of aggregate-prone tau fragments as well as physiological tau degradation, pointing to a dual role of microglia-mediated tau proteolysis (Behrendt et al., 2019). Legumain cleaves tau at four different sites (N167, N255, N296 and N368)(Schlegel et al., 2019). Previous studies indicated that legumain knockout mice show increased AT8 tau inclusions and that legumain-resistant tau prevents synaptic dysfunction (Z. Zhang et al., 2014). Studies point to both increased (Behrendt et al., 2019; Z. Zhang et al., 2014) or unchanged (Schlegel et al., 2019) legumain activity in human AD brains; the study that found unchanged levels controlled better for vascular issues which can drive legumain activity. Further, follow-up studies have not found accumulation of N368 tau fragments in tau aggregates in AD brains (Schlegel et al., 2019). Notably, microglia are enriched for legumain in the brain compared to other cell types and microglia are able to degrade tau without releasing cleaved fragments (Behrendt et al., 2019). When microglia are depleted from the brain via CSF1R inhibition, there is a significant reduction in legumain expression (Spangenberg et al., 2019). Cathepsin (in Greek: “to digest”) is a family of widely expressed proteases which are found within the endosomal/lysosomal compartments during homeostatic conditions and may also be secreted upon induction (Cavallo-Medved et al., 2011), such as through pro-inflammatory activation in the case of microglia (Lowry and Klegeris, 2018). Cathepsins are differentially classified based on the amino acid residue within the proteolytic site which confers their function as serine, cysteine, or aspartic proteases (Siklos et al., 2015; Turk et al., 2012). The role of microglial cathepsins in neurodegenerative disease is dependent upon context (Lowry and Klegeris, 2018). Microglial cathepsins serve not only immune clearance functions, but are also immune regulatory roles (Lowry and Klegeris, 2018). For example, intracellular cysteine protease cathepsin B has been implicated in tau processing (Bendiske and Bahr, 2003; Farizatto et al., 2017), but has also been shown to enhance the NF-κB and caspase-1 activation (Halle et al., 2008; Ni et al., 2015; Zhou et al., 2016). Pharmacological enhancement of cathepsin B has been shown to reduce hyperphosphorylated tau accumulation in cultured hippocampal slices (Bendiske and Bahr, 2003; Farizatto et al., 2017). However, interpretation of this data is difficult as cathepsin B is also highly expressed in neurons (Bernstein et al., 1990) and astrocytes (Oberstein et al., 2021). Like legumain, cathepsin B is pH sensitive, but it differs in that lower pH favors its exopeptidase activity while higher (neutral/alkaline) pH favors its endopeptidase activity (Cavallo-Medved et al., 2011). Thus, cathepsin B processing of tau may differ depending upon its location intra- or extracellularly and the acidification state of the lysosomal compartment. However, enhancement of mature cathepsin B expression via treatment with Z-Phe-Ala-diazomethylketone (PADK) was capable of reversing hyperphosphorylated tau accumulation induced by chloroquine treatment, despite the lysosome deacidifying property of chloroquine (Bendiske and Bahr, 2003). Further, PADK treatment has been shown to prevent hyperphosphorylated tau accumulation in the context of proteosome inhibition (Farizatto et al., 2017). Thus, it is possible that cathepsin B can degrade certain pathological forms of tau in both acidic and non-acidic contexts. However, due to the differing function of exo- or endopeptidases, it is likely that its cleavage products are dependent upon these two contexts. Importantly, extracellular cathepsin B has been shown to be neurotoxic independent of tau pathology (Kingham and Pocock, 2001). In addition to cathepsin B, cathepsin D (Bednarski and Lynch, 1996; Johnson et al., 1991) and cathepsin S (Nübling et al., 2017) have been shown to cleave tau. Notably, incubation of tau with cathepsin S alone results in tau oligomer formation (Nübling et al., 2017), suggesting that partial degradation of tau can be deleterious. Within the lysosome, cathepsins and other lysosomal proteases most likely work in concert to degrade tau into its constituent amino acids. Overall, the role of microglial cathepsins in the context of tauopathy remains poorly understood. Thus, further work is warranted to determine how modulation of cathepsin activity may affect microglial tau processing and tauopathy neurodegeneration.
6. Role of microglia phenotype in tau processing
Microglia phenotype changes throughout the course of tauopathy (Rexach et al., 2020; H. Wang et al., 2018), which may affect microglial processing of tau. When assessing the role of the MGnD/DAM and inflammatory phenotypes in tauopathy, it is important to remember that neither phenotype represents a monolith. Indeed, their constituent subpopulations may affect tau pathology in different ways. This may explain why experimental models of these two phenotypes have been shown to both restrict and propagate tau pathology under different contexts. For example, ApoE, an AD risk factor that is crucial for triggering the MGnD/DAM phenotype during AD (Krasemann et al., 2017), is correlated with high tau seeding activity in human cases of tauopathy (Dujardin et al., 2020). Ablation of microglia or microglia-specific knockout of ApoE reduces NFT-mediated neurodegeneration in tauopathy model mice suggesting that MGnD/DAM contribute to tau-mediated neurodegeneration (Shi et al., 2019). Additionally, microglia activation triggers tau release by microglia (Asai et al., 2015; Clayton et al., 2021). Similarly, activated microglia may contribute to tau pathology by exacerbating tau pathological phosphorylation, aggregation, and expression in mouse and cellular models (Bhaskar et al., 2010; Ghosh et al., 2013; Kitazawa et al., 2005; Lee et al., 2015; Li et al., 2003; Quintanilla et al., 2004). However, activated microglia can also restrict tau seeding and spreading: ablation of microglia or suppression of microglia activation by knockout of TREM2 in the context of Aβ pathology enhances tau spreading (Gratuze et al., 2021). This is also consistent with data showing that microglia need to be activated to internalize and degrade tau (Andersson et al., 2019; Luo et al., 2015; Majerova et al., 2014), but these studies themselves are not entirely consistent with each other. For example, Majerova and colleagues observed that microglia activation with LPS modestly enhances the proportion of microglia that internalize tau pre-formed fibrils from 26.4 % to 42.6 % with 24 h of LPS pretreatment with no effect with a 2–6 h LPS pretreatment (Majerova et al., 2014), while Andersson and colleagues did not note any significant change in intracellular tau levels with LPS treatment at an undescribed timepoint (Andersson et al., 2019). Notably, peripheral macrophages and monocytes are more effective than microglia at internalization and degradation of tau, suggesting there are specific properties of different myeloid cells that make them more efficient at tau degradation (Majerova et al., 2014). This could be due to reduced lysosomal acidity in microglia compared to other macrophages (Majumdar et al., 2007). Moreover, in the chronic inflammatory environment of AD, microglia lose key degradative enzymes (Hickman et al., 2008). Additionally, inflammatory activation is known to alter endocytosis in myeloid-lineage cells (Redka et al., 2018). These observations warrant a deeper examination into which cellular processes drive tau degradation without also causing improper processing and packaging and release of tau into extracellular vesicles. Such findings would not only help to reconcile contradictions on the influence of phenotype on tau processing, but also provide therapeutic targets for promoting tau clearance.
7. Strategies to promote tau clearance
The overall mechanism of tau clearance from the brain is unknown, although clearance into the glymphatic system (Patel et al., 2019), and vasculature (J. Wang et al., 2018) are involved. Notably, tau pathology induces vascular dysfunction that may affect clearance (Bennett et al., 2018). It is not known whether tau pathology leads to glymphatic dysfunction (Silva et al., 2021), but in mouse models of aging (Kress et al., 2014) and Aβ pathology (Peng et al., 2016) the glymphatic system is impaired as a result of astrogliosis and mislocalization of astrocytic aquaporin 4 (AQP4). Loss of AQP4 results in increased tau aggregation after traumatic brain injury (Iliff et al., 2014). Importantly, microglia compensate clearance of extracellular aggregates when the glymphatic system is impaired (Feng et al., 2020). Pre-plaque AQP4-deficient APP/PS1 mice exhibited more intraneuronal accumulation of Aβ and enhanced microglia activation compared to AQP4-intact APP/PS1 mice, but only loss of AqP4 combined with local depletion of microglia via clodronate liposomes resulted in plaque deposition (Feng et al., 2020). Whether microglia would compensate similarly with extracellular tau is unknown. However, it was shown that intrahippocampally injected monomeric tau is cleared from the brain parenchyma and deposited into the systemic circulation much more efficiently in mice with an intact glymphatic system than in mice in which the glymphatic system was ablated genetically (Patel et al., 2019). Future studies will need to investigate the role of microglia in extracellular tau clearance in the context of glymphatic and vascular dysfunction during tauopathy.
Therapeutics that target tau protein, such as anti-tau mAbs, are also in clinical trials (e. g. NCT03828747). Tau aggregates are reduced in tauopathy models after treatment with anti-tau mAbs (Ayalon et al., 2021; Castillo-Carranza et al., 2014; Yanamandra et al., 2013; Zilkova et al., 2020). Studies have shown that microglia are important for antibody-mediated tau clearance putatively via FcγR effector function (Andersson et al., 2019; Funk et al., 2015; Zilkova et al., 2020). However, a group at Genentech reported that microglial activation via effector function of anti-pS409-tau immunotherapy negates its therapeutic efficacy (Lee et al., 2016). The authors noted that only effectorless mutant anti-pS409-tau antibodies were able to block intraneuronal tau fragmentation induced by exogenously applied tau multimers when neurons were cocultured with microglia (Lee et al., 2016). They also observed that effectorless mutant anti-pS409-tau antibodies were modestly more effective in blocking tau spread in a mouse model of tauopathy than WT anti-pS409-tau antibodies, albeit both were significantly more effective than IgG control (Lee et al., 2016). Based on these findings, they conclude that FcγR effector function attenuates the therapeutic efficacy of tau immunotherapies by causing neurotoxicity via pro-inflammatory activation of microglia (Lee et al., 2016). Thus, passive immunotherapies with an IgG4 backbone which confers reduced effector function relative to other IgG isotypes (e. g. IgG1, IgG3) (Vidarsson et al., 2014) have dominated clinical trials (NCT03289143, NCT03828747, NCT03068468, NCT02880956). Thus far, most of these therapies have since been scrapped due to a failure to demonstrate disease modifying benefits except for one (Semorinemab) for which clinical trials are ongoing (NCT03828747). Lee and colleagues’ rationale for choosing the IgG4 backbone is contested by a more recent study reporting anti-tau antibody IgG isotype had no impact on pro-inflammatory cytokine secretion by primary human microglia (Zilkova et al., 2020). The same study also found that IgG1 isotype more effectively promoted tau uptake by human microglia than the IgG4 isotype (Zilkova et al., 2020). Furthermore, several experimental studies have shown that antibodies promote microglial degradative capacity for tau (Andersson et al., 2019; Funk et al., 2015; Luo et al., 2015). Although antibodies might also facilitate tau degradation by neuronal lysosomes (Congdon et al., 2013), it may be more favorable to design therapies that direct tau aggregates away from neurons and toward microglia. More work is required to elucidate the overall therapeutic mechanism of anti-tau mAbs, so that more effective treatments may be developed.
Therapeutics that reduce Mapt gene expression, such as antisense oligonucleotides (ASOs), are currently in clinical trials (e.g., NCT03186989). Mapt-targeting ASO treatment in tauopathy mice reduces existing tau aggregate pathology (DeVos et al., 2017). Theoretically these therapies may act primarily intracellularly to restrict replication of tau multimers which one study suggests may play a larger role than spread in late-stage tau pathological progression (Meisl et al., 2021). However, microglial clearance of extracellular tau may also contribute to its therapeutic efficacy. Notably, when the Mapt gene is silenced in the rTg4510 mouse model of tauopathy, the presence of Aβ pathology attenuates tau clearance after tau gene silencing compared to mice that only exhibit tau pathology (DeVos et al., 2018b) which could be due to disrupted microglia endocytosis due to Aβ pathology (Hickman et al., 2008). These data suggest that microglia may play a role in therapeutic tau clearance after Mapt gene silencing. Future studies will need to investigate the role of microglial endocytosis in tau clearance after Mapt gene silencing.
8. Concluding remarks
Herein we have discussed the importance of microglia in tauopathies and ways in which these cells may help or harm in the context of future tau-targeted therapies. There are currently no disease-modifying treatments for tauopathies, and halting tau-spread may be one of the most effective strategies for preventing widespread neurodegeneration. Ultimately, tau-based therapeutic strategies aimed at eliminating or mitigating cognitive impairment will be most efficacious in prodromal, asymptomatic, or early disease phases, before the spread and degree of tau pathology leads to irreparable cell loss and atrophy. Therefore, understanding factors that influence the propagation or restriction of tau pathology, such as microglia, is vital for developing effective treatments for tauopathy. Theoretically, promoting tau clearance by inducing microglial endocytosis and degradation of tau could be an effective therapeutic strategy. However, microglial endocytosis of tau may not always be protective, as improper processing of tau could enhance its propensity for spread (Asai et al., 2015; Clayton et al., 2021; Hopp et al., 2018). Future therapies will want to avoid triggering microglial pro-inflammatory cytokine cascades, improper proteolysis of tau, and exosome release. Anti-tau mAbs afford a means to enhance microglial degradation of extracellular tau (Andersson et al., 2019; Funk et al., 2015; Luo et al., 2015), but the microglial contribution to tau clearance therein is still unclear. Determining outcomes that potentiate anti-tau therapeutic efficacy, via microglia or other mechanisms, is paramount to successful design of future therapies. We present a model for microglia-tau interactions in Fig. 1, acknowledging unanswered questions remain. Our current understanding suggests that microglia act as both friend and foe within the context of tauopathy. Overall, promoting microglia beneficial functions will be key to harnessing their therapeutic potential in tauopathy. In adapting microglia’s therapeutic potential, the approach of handling these cells may be akin to a common approach in life: ‘to keep your friends close, but your enemies closer’.
Fig. 1.
Possible pathways by which tau seeds are internalized and processed by microglia. Black arrows: Tau seeding and spread pathway – Tau seeds are released from the axon terminal as either free or EV tau. Released tau is taken up by the postsynaptic neuron and internalized seeds serve as a structural template for native tau to misfold and aggregate. Blue arrows: Tau endocytic pathways – Free tau, EV tau, mAb-bound tau, or tau within dendritic spines can be taken up by various endocytic mechanisms. Single immune complexes of tau and anti-tau mAb are endocytosed via FcγRs and most likely involve CME. C1q, Axl/Mer and Gas6 have been implicated in the phagocytosis of dendritic spines. Tau binds microglial HSPGs which are internalized via macropinocytosis. Microglia internalize EVs through macropinocytosis. The endocytosed tau seeds are transported via the nascent endocytic vesicle to the EE. The EE membrane invaginates to form a MVB and matures to form an LE. Green arrows: Tau degradative pathways – Tau may be degraded extracellularly by CatB released by a secretory lysosome or intracellularly upon fusion of the MVB/LE with a LY containing legumain. Red arrows: Tau endosomal escape/exosome release – Tau has been shown to escape ELs. Co-activation of TLR4 and P2X7R, and the downstream inflammasome pathway induces the release of exosomes from the MVB/LE. This causes the release of tau-containing exosomes in the context of tau pathology. Abbreviations: Bin1 = bridging integrator 1; CatB = cathepsin B; C1q = complement component 1 q; CME = clathrin-mediated endocytosis; EE = early endosome; EL = endolysosome; EV = extracellular vesicle; Exo = exosome, FcγR = Fc gamma receptor; HSPG = heparan sulfate proteoglycan; LE = late endosome; LY = lysosome; Macro = macropinocytosis; MVB = multivesieular body; NLRP3 = NOD-, LRR-, and pyrin domain-containing protein 3; P2X7R = purinergic P2X7 ionotropic receptor; Phago = phagocytosis; TLR4 = toll-like receptor 4.
Funding sources
KFO is supported by funding from the National Institutes of Health [T32 NS082145]. SCH is supported by the National Institutes of Health [K01AG066747, R21AG072423], the Brain and Behavior Research Foundation [28970], the Alzheimer’s Association [AARG-21-846012], and the Dan & Kelley McNamara Alzheimer’s Research Fund. KFB is supported by funding from the National Institute on Aging (P30AG066546-01A1, R01AG062348, R01AG070214), the Michael J. Fox Foundation (MJFF020301), the Texas Alzheimer’s Research and Care Consortium, the J.M.R. Barker Foundation, the Bill and Rebecca Reed Center for Precision Therapies and Palliative Care, and the Bartell and Mollie Zachry Endowment for Alzheimer’s Research and Patient Care.
Abbreviations:
- MAPT
Microtubule associated protein tau
- AD
Alzheimer’s disease
- FTLD-tau
frontotemporal lobar degeneration tau
- CBD
corticobasal degeneration
- PSP
progressive supranuclear palsy
- NFT
neurofibrillary tangle
- EVs
extracellular vesicles
- Aβ
amyloid β
- PET
positron emission tomography
- Iba1
ionized calcium binding adaptor molecule 1
- MMSE
mini mental state exam
- DAM
disease associated microglia
- MGnD
neurodegenerative microglia
- Gas6
growth-arrest specific gene 6
- TNF
tumor necrosis factor
- IL
interleukin
- CSF1
colony stimulating factor 1
- TLR
toll-like receptor
- IFN
interferon
- dsRNA
double stranded RNA
- ApoE
apolipoprotein E
- Trem2
triggering receptor expressed on myeloid cells 2
- PLCγ2
phospholipase C gamma 2
- LOAD
late onset AD
- BIN1
Bridging integrator 1
- TPSO
translocator protein 18 kDa
- CSF
cerebrospinal fluid
- LPS
lipopolysaccharide
- WT
wild-type
- mTREM2
murine TREM2
- SASP
senescence-associated secretory phenotype
- HSPG
heparan sulphate proteoglycan
- LRP1
low density lipoprotein receptor related protein 1
- IgG
immunoglobulin G
- C1q
complement component 1q
- mAb
monoclonal antibody
- MFG-E8
milk-fat globule epidermal growth factor 8
- PS
phosphatidylserines
- ALA
α-linolenic acid
- CME
clathrin-mediated endocytosis
- GSK3β
glycogen synthase 3 β
- CLAP
clathrin and AP-2-binding
- APP
amyloid precursor protein
- PS1
presenilin 1
- HEK293T
Human embryonic kidney 293T
- CSF1R
CSF1 receptor
- AEP
asparaginyl endopeptidase
- AQP4
astrocytic aquaporin 4
- ASOs
antisense oligonucleotides
References
- Andersson CR, Falsig J, Stavenhagen JB, Christensen S, Kartberg F, Rosenqvist N, Finsen B, Pedersen JT, 2019. Antibody-mediated clearance of tau in primary mouse microglial cultures requires Fcγ-receptor binding and functional lysosomes. Sci. Rep 9. 10.1038/s41598-019-41105-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ando K, Brion JP, Stygelbout V, Suain V, Authelet M, Dedecker R, Chanut A, Lacor P, Lavaur J, Sazdovitch V, Rogaeva E, Potier MC, Duyckaerts C, 2013. Clathrin adaptor CALM/PICALM is associated with neurofibrillary tangles and is cleaved in Alzheimer’s brains. Acta Neuropathol. 125, 861–878. 10.1007/s00401-013-1111-z. [DOI] [PubMed] [Google Scholar]
- Andreone BJ, Przybyla L, Llapashtica C, Rana A, Davis SS, van Lengerich B, Lin K, Shi J, Mei Y, Astarita G, Di Paolo G, Sandmann T, Monroe KM, Lewcock JW, 2020. Alzheimer’s-associated PLCγ2 is a signaling node required for both TREM2 function and the inflammatory response in human microglia. Nat. Neurosci 23, 927–938. 10.1038/s41593-020-0650-6. [DOI] [PubMed] [Google Scholar]
- Angelova DM, Brown DR, 2019. Microglia and the aging brain: are senescent microglia the key to neurodegeneration. J. Neurochem 10.1111/jnc.14860. [DOI] [PubMed] [Google Scholar]
- Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT, 1992. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 42, 631–639. 10.1212/wnl.42.3.631. [DOI] [PubMed] [Google Scholar]
- Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, Wolozin B, Butovsky O, Kügler S, Ikezu T, 2015. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci 18, 1584–1593. 10.1038/nn.4132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayalon G, Lee SH, Adolfsson O, Foo-Atkins C, Atwal JK, Blendstrup M, Booler H, Bravo J, Brendza R, Brunstein F, Chan R, Chandra P, Couch JA, Datwani A, Demeule B, DiCara D, Erickson R, Ernst JA, Foreman O, He D, Hötzel I, Keeley M, Kwok MCM, Lafrance-Vanasse J, Lin H, Lu Y, Luk W, Manser P, Muhs A, Ngu H, Pfeifer A, Pihlgren M, Rao GK, Scearce-Levie K, Schauer SP, Smith WB, Solanoy H, Teng E, Wildsmith KR, Yadav DB, Ying Y, Fuji RN, Kerchner GA, 2021. Antibody semorinemab reduces tau pathology in a transgenic mouse model and engages tau in patients with Alzheimer’s disease. Sci. Transl. Med 13 10.1126/scitranslmed.abb2639. [DOI] [PubMed] [Google Scholar]
- Banati RB, Gehrmann J, Schubert P, Kreutzberg GW, 1993. Cytotoxicity of microglia. Glia 7, 111–118. 10.1002/GLIA.440070117. [DOI] [PubMed] [Google Scholar]
- Bednarski E, Lynch G, 1996. Cytosolic proteolysis of τ by cathepsin D in hippocampus following suppression of cathepsins B and L. J. Neurochem 67, 1846–1855. 10.1046/j.1471-4159.1996.67051846.x. [DOI] [PubMed] [Google Scholar]
- Behrendt A, Bichmann M, Ercan-Herbst E, Haberkant P, Schöndorf DC, Wolf M, Fahim SA, Murolo E, Ehrnhoefer DE, 2019. Asparagine endopeptidase cleaves tau at N167 after uptake into microglia. Neurobiol. Dis 130. 10.1016/j.nbd.2019.104518. [DOI] [PubMed] [Google Scholar]
- Bejanin A, Schonhaut DR, La Joie R, Kramer JH, Baker SL, Sosa N, Ayakta N, Cantwell A, Janabi M, Lauriola M, O’Neil JP, Gorno-Tempini ML, Miller ZA, Rosen HJ, Miller BL, Jagust WJ, Rabinovici GD, 2017. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 140, 3286–3300. 10.1093/brain/awx243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bemiller SM, McCray TJ, Allan K, Formica SV, Xu G, Wilson G, Kokiko-Cochran ON, Crish SD, Lasagna-Reeves CA, Ransohoff RM, Landreth GE, Lamb BT, 2017. TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy. Mol. Neurodegener 12, 1–12. 10.1186/s13024-017-0216-6/FIGURES/4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bendiske J, Bahr BA, 2003. Lysosomal activation is a compensatory response against protein accumulation and associated synaptopathogenesis—an approach for slowing Alzheimer disease. J. Neuropathol. Exp. Neurol 62, 451–463. 10.1093/JNEN/62.5.451. [DOI] [PubMed] [Google Scholar]
- Bennett RE, Robbins AB, Hu M, Cao X, Betensky RA, Clark T, Das S, Hyman BT, 2018. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in P301L transgenic mice and human Alzheimer’s disease. Proc. Natl. Acad. Sci. U. S. A 115, E1289–E1298. 10.1073/pnas.1710329115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernstein HG, Kirschke H, Wiederanders B, Schmidt D, Rinne A, 1990. Antigenic expression of cathepsin B in aged human brain. Brain Res. Bull 24, 543–549. 10.1016/0361-9230(90)90157-U. [DOI] [PubMed] [Google Scholar]
- Berry RW, Sweet AP, Clark FA, Lagalwar S, Lapin BR, Wang T, Topgi S, Guillozet-Bongaarts AL, Cochran EJ, Bigio EH, Binder LI, 2004. Tau epitope display in progressive supranuclear palsy and corticobasal degeneration. J. Neurocytol 333 (33), 287–295. 10.1023/B:NEUR.0000044190.96426.B9. [DOI] [PubMed] [Google Scholar]
- Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT, 2010. Regulation of tau pathology by the microglial fractalkine receptor. Neuron 68, 19–31. 10.1016/j.neuron.2010.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biber K, Neumann H, Inoue K, Boddeke HWGM, 2007. Neuronal “On” and “Off” signals control microglia. Trends Neurosci. 10.1016/j.tins.2007.08.007. [DOI] [PubMed] [Google Scholar]
- Bohdanowicz M, Schlam D, Hermansson M, Rizzuti D, Fairn GD, Ueyama T, Somerharju P, Du G, Grinstein S, 2013. Phosphatidic acid is required for the constitutive ruffling and macropinocytosis of phagocytes. Mol. Biol. Cell 24, 1700–1712. 10.1091/mbc.E12-11-0789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolós M, Llorens-Martín M, Jurado-Arjona J, Hernández F, Rábano A, Avila J, 2015. Direct evidence of internalization of tau by microglia in vitro and in vivo. J. Alzheimer’s Dis 50, 77–87. 10.3233/JAD-150704. [DOI] [PubMed] [Google Scholar]
- Bolós M, Llorens-Martín M, Perea JR, Jurado-Arjona J, Rábano A, Hernández F, Avila J, 2017. Absence of CX3CR1 impairs the internalization of Tau by microglia. Mol. Neurodegener 12, 59. 10.1186/s13024-017-0200-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonecchi R, Savino B, Borroni EM, Mantovani A, Locati M, 2010. Chemokine decoy receptors: structure–function and biological properties. Chemokine Syst. Exp. Clin. Hematol 15–36. 10.1007/82_2010_19. [DOI] [PubMed] [Google Scholar]
- Booth PL, Thomas WE, 1991. Evidence for motility and pinocytosis in ramified microglia in tissue culture. Brain Res. 548, 163–171. 10.1016/0006-8993(91)91118-K. [DOI] [PubMed] [Google Scholar]
- Braak H, Braak E, 1991. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259. [DOI] [PubMed] [Google Scholar]
- Bratton DL, Fadok VA, Richter DA, Kailey JM, Guthrie LA, Henson PM, 1997. Appearance of phosphatidylserine on apoptotic cells requires calcium- mediated nonspecific flip-flop and is enhanced by loss of the aminophospholipid translocase. J. Biol. Chem 272, 26159–26165. 10.1074/jbc.272.42.26159. [DOI] [PubMed] [Google Scholar]
- Brelstaff J, Tolkovsky AM, Ghetti B, Goedert M, Spillantini MG, 2018. Living neurons with tau filaments aberrantly expose phosphatidylserine and are phagocytosed by microglia. Cell Rep. 24, 1939–1948. 10.1016/j.celrep.2018.07.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brelstaff JH, Mason M, Katsinelos T, McEwan WA, Ghetti B, Tolkovsky AM, Spillantini MG, 2021. Microglia become hypofunctional and release metalloproteases and tau seeds when phagocytosing live neurons with P301S tau aggregates. Sci. Adv 7. 10.1126/sciadv.abg4980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bretteville A, Planel E, 2008. Tau aggregates: toxic, inert, or protective species. J. Alzheimer’s Dis 14, 431–436. 10.3233/JAD-2008-14411. [DOI] [PubMed] [Google Scholar]
- Brunello CA, Merezhko M, Uronen RL, Huttunen HJ, 2020. Mechanisms of secretion and spreading of pathological tau protein. Cell. Mol. Life Sci 10.1007/s00018-019-03349-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ, 2018. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582. 10.1038/s41586-018-0543-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP, Weiner HL, 2014. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci 17, 131–143. 10.1038/nn.3599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butovsky O, Jedrychowski MP, Cialic R, Krasemann S, Murugaiyan G, Fanek Z, Greco DJ, Wu PM, Doykan CE, Kiner O, Lawson RJ, Frosch MP, Pochet N, El Fatimy R, Krichevsky AM, Gygi SP, Lassmann H, Berry J, Cudkowicz ME, Weiner HL, 2015. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol 77, 75–99. 10.1002/ana.24304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, Jones T, Banati RB, 2001. In-vivo measurement of activated microglia in dementia. Lancet 358, 461–467. 10.1016/S0140-6736(01)05625-2. [DOI] [PubMed] [Google Scholar]
- Cagnin A, Kassiou M, Meikle SR, Banati RB, 2006. In vivo evidence for microglial activation in neurodegenerative dementia. Acta Neurol. Scand. Suppl 185, 107–114. 10.1111/j.1600-0404.2006.00694.x. [DOI] [PubMed] [Google Scholar]
- Calafate S, Flavin W, Verstreken P, Moechars D, 2016. Loss of Bin1 promotes the propagation of tau pathology. Cell Rep. 17, 931–940. 10.1016/J.CELREP.2016.09.063. [DOI] [PubMed] [Google Scholar]
- de Calignon A, Polydoro M, Suárez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, Pitstick R, Sahara N, Ashe KH, Carlson GA, Spires-Jones TL, Hyman BT, 2012. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 73, 685–697. 10.1016/j.neuron.2011.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron B, Landreth GE, 2010. Inflammation, microglia, and Alzheimer’s disease. Neurobiol. Dis 37, 503–509. 10.1016/j.nbd.2009.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canton J, 2018. Macropinocytosis: New insights into its underappreciated role in innate immune cell surveillance. Front. Immunol 10.3389/fimmu.2018.02286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canton J, Schlam D, Breuer C, Gütschow M, Glogauer M, Grinstein S, 2016. Calcium-sensing receptors signal constitutive macropinocytosis and facilitate the uptake of NOD2 ligands in macrophages. Nat. Commun 7, 1–12. 10.1038/ncomms11284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castillo-Carranza DL, Gerson JE, Sengupta U, Guerrero-Muñoz MJ, Lasagna-Reeves CA, Kayed R, 2014. Specific targeting of tau oligomers in Htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds. J. Alzheimer’s Dis 40. 10.3233/JAD-132477. [DOI] [PubMed] [Google Scholar]
- Cavallo-Medved D, Moin K, Sloane B, 2011. Cathepsin B: basis sequence: mouse. AFCS-Nat. Mol 2011. [PMC free article] [PubMed] [Google Scholar]
- Champion JA, Walker A, Mitragotri S, 2008. Role of particle size in phagocytosis of polymeric microspheres. Pharm. Res 25, 1815–1821. 10.1007/s11095-008-9562-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherry JD, Tripodis Y, Alvarez VE, Huber B, Kiernan PT, Daneshvar DH, Mez J, Montenigro PH, Solomon TM, Alosco ML, Stern RA, McKee AC, Stein TD, 2016. Microglial neuroinflammation contributes to tau accumulation in chronic traumatic encephalopathy. Acta Neuropathol. Commun 4, 112. 10.1186/s40478-016-0382-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chuang TY, Guo Y, Seki SM, Rosen AM, Johanson DM, Mandell JW, Lucchinetti CF, Gaultier A, 2016. LRP1 expression in microglia is protective during CNS autoimmunity. Acta Neuropathol. Commun 4, 68. 10.1186/s40478-016-0343-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chuong EB, Elde NC, Feschotte C, 2016. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Sci. (80-). 351, 1083–1087. 10.1126/science.aad5497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, Fraser G, Stalder AK, Beibel M, Staufenbiel M, Jucker M, Goedert M, Tolnay M, 2009. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol 11, 909–913. 10.1038/ncb1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clavaguera F, Akatsu H, Fraser G, Crowther RA, Frank S, Hench J, Probst A, Winkler DT, Reichwald J, Staufenbiel M, Ghetti B, Goedert M, Tolnay M, 2013. Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc. Natl. Acad. Sci. U. S. A 110, 9535–9540. 10.1073/pnas.1301175110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clayton K, Delpech JC, Herron S, Iwahara N, Ericsson M, Saito T, Saido TC, Ikezu S, Ikezu T, 2021. Plaque associated microglia hyper-secrete extracellular vesicles and accelerate tau propagation in a humanized APP mouse model. Mol. Neurodegener 16, 1–16. 10.1186/s13024-021-00440-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombo M, Raposo G, Théry C, 2014. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol 10.1146/annurev-cellbio-101512-122326. [DOI] [PubMed] [Google Scholar]
- Congdon EE, Gu J, Sait HBR, Sigurdsson EM, 2013. Antibody uptake into neurons occurs primarily via clathrin-dependent fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J. Biol. Chem 288, 35452. 10.1074/JBC.M113.491001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Constantinides VC, Paraskevas GP, Paraskevas PG, Stefanis L, Kapaki E, 2019. Corticobasal degeneration and corticobasal syndrome: a review. Clin. Park. Relat. Disord 1, 66–71. 10.1016/j.prdoa.2019.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conway OJ, Carrasquillo MM, Wang X, Bredenberg JM, Reddy JS, Strickland SL, Younkin CS, Burgess JD, Allen M, Lincoln SJ, Nguyen T, Malphrus KG, Soto AI, Walton RL, Boeve BF, Petersen RC, Lucas JA, Ferman TJ, Cheshire WP, Van Gerpen JA, Uitti RJ, Wszolek ZK, Ross OA, Dickson DW, Graff-Radford NR, Ertekin-Taner N, 2018. ABI3 and PLCG2 missense variants as risk factors for neurodegenerative diseases in Caucasians and African Americans 11 Medical and Health Sciences 1109 Neurosciences 06 Biological Sciences 0604 Genetics. Mol. Neurodegener 13, 1–12. 10.1186/s13024-018-0289-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J, 2008. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, e301 10.1371/journal.pbio.0060301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crotti A, Sait HR, McAvoy KM, Estrada K, Ergun A, Szak S, Marsh G, Jandreski L, Peterson M, Reynolds TL, Dalkilic-Liddle I, Cameron A, Cahir-McFarland E, Ransohoff RM, 2019. BIN1 favors the spreading of Tau via extracellular vesicles. Sci. Rep 9, 1–20. 10.1038/s41598-019-45676-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cummings JL, Morstorf T, Zhong K, 2014. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res. Ther 6, 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dani M, Wood M, Mizoguchi R, Fan Z, Walker Z, Morgan R, Hinz R, Biju M, Kuruvilla T, Brooks DJ, Edison P, 2018. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain 141, 2740–2754. 10.1093/BRAIN/AWY188. [DOI] [PubMed] [Google Scholar]
- Das R, Chinnathambi S, 2021. Microglial remodeling of actin network by Tau oligomers, via G protein-coupled purinergic receptor, P2Y12R-driven chemotaxis. Traffic 22, 153–170. 10.1111/tra.12784. [DOI] [PubMed] [Google Scholar]
- Das R, Balmik AA, Chinnathambi S, 2020. Phagocytosis of full-length Tau oligomers by actin-remodeling of activated microglia. J. Neuroinflamm 17, 1–15. 10.1186/s12974-019-1694-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- David DC, Layfield R, Serpell L, Narain Y, Goedertà M, Spillantini MG, 2002. Proteasomal degradation of tau protein. J. Neurochem 83, 176–185. [DOI] [PubMed] [Google Scholar]
- De La-Rocque S, Moretto E, Butnaru I, Schiavo G, 2020. Knockin’ on heaven’s door: Molecular mechanisms of neuronal tau uptake. J. Neurochem 10.1111/jnc.15144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I, 2018. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell. Cell. Press. [DOI] [PubMed] [Google Scholar]
- Dehkordi SK, Walker J, Sah E, Bennett E, Atrian F, Frost B, Woost B, Bennett RE, Orr TC, Zhou Y, Andhey PS, Colonna M, Sudmant PH, Xu P, Wang M, Zhang B, Zare H, Orr ME, 2021. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat. Aging 112 (1), 1107–1116. 10.1038/s43587-021-00142-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA, Rugo HS, Shelley Hwang E, Jirström K, West BL, Coussens LM, 2011. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Disco 1, 54. 10.1158/2159-8274.CD-10-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desale SE, Chinnathambi S, 2021. α-Linolenic acid induces clearance of Tau seeds via Actin-remodeling in Microglia. Mol. Biomed 2. 10.1186/s43556-021-00028-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeVos SL, Miller RL, Schoch KM, Holmes BB, Kebodeaux CS, Wegener AJ, Chen G, Shen T, Tran H, Nichols B, Zanardi TA, Kordasiewicz HB, Swayze EE, Bennett CF, Diamond MI, Miller TM, 2017. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci. Transl. Med 10.1126/scitranslmed.aag0481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeVos SL, Corjuc BT, Commins C, Dujardin S, Bannon RN, Corjuc D, Moore BD, Bennett RE, Jorfi M, Gonzales JA, Dooley PM, Roe AD, Pitstick R, Irimia D, Frosch MP, Carlson GA, Hyman BT, 2018a. Tau reduction in the presence of amyloid-β prevents tau pathology and neuronal death in vivo. Brain 141, 2194–2212. 10.1093/brain/awy117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeVos SL, Corjuc BT, Oakley DH, Nobuhara CK, Bannon RN, Chase A, Commins C, Gonzalez JA, Dooley PM, Frosch MP, Hyman BT, 2018b. Synaptic tau seeding precedes tau pathology in human Alzheimer’s disease brain. Front. Neurosci 12, 267. 10.3389/fnins.2018.00267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doodnauth SA, Grinstein S, Maxson ME, 2019. Constitutive and stimulated macropinocytosis in macrophages: Roles in immunity and in the pathogenesis of atherosclerosis. Philos. Trans. R. Soc. B Biol. Sci 10.1098/rstb.2018.0147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubyak GR, 2012. P2X7 receptor regulation of non-classical secretion from immune effector cells. Cell. Microbiol 14, 1697–1706. 10.1111/cmi.12001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dujardin S, Commins C, Lathuiliere A, Beerepoot P, Fernandes AR, Kamath TV, De Los Santos MB, Klickstein N, Corjuc DL, Corjuc BT, Dooley PM, Viode A, Oakley DH, Moore BD, Mullin K, Jean-Gilles D, Clark R, Atchison K, Moore R, Chibnik LB, Tanzi RE, Frosch MP, Serrano-Pozo A, Elwood F, Steen JA, Kennedy ME, Hyman BT, 2020. Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease. Nat. Med 26, 1256–1263. 10.1038/s41591-020-0938-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duus K, Hansen EW, Tacnet P, Frachet P, Arlaud GJ, Thielens NM, Houen G, 2010. Direct interaction between CD91 and C1q. FEBS J. 277, 3526–3537. 10.1111/J.1742-4658.2010.07762.X. [DOI] [PubMed] [Google Scholar]
- Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, Hammers A, Tai YF, Fox N, Kennedy A, Rossor M, Brooks DJ, 2008. Microglia, amyloid, and cognition in Alzheimer’s disease: an [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol. Dis 32, 412–419. 10.1016/j.nbd.2008.08.001. [DOI] [PubMed] [Google Scholar]
- Efthymiou AG, Goate AM, 2017. Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener 12, 43. 10.1186/s13024-017-0184-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmore MRP, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, Kitazawa M, Matusow B, Nguyen H, West BL, Green KN, 2014. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380–397. 10.1016/j.neuron.2014.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falcon B, Cavallini A, Angers R, Glover S, Murray TK, Barnham L, Jackson S, O’Neill MJ, Isaacs AM, Hutton ML, Szekeres PG, Goedert M, Bose S, 2015. Conformation determines the seeding potencies of native and recombinant Tau aggregates. J. Biol. Chem 290, 1049–1065. 10.1074/jbc.M114.589309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farizatto KLG, Ikonne US, Almeida MF, Ferrari MFR, Bahr BA, 2017. Aβ42-mediated proteasome inhibition and associated tau pathology in hippocampus are governed by a lysosomal response involving cathepsin B: evidence for protective crosstalk between protein clearance pathways. PLoS One 12, e0182895. 10.1371/JOURNAL.PONE.0182895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng W, Zhang Y, Wang Z, Xu H, Wu T, Marshall C, Gao J, Xiao M, 2020. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer’s disease mouse model with suppression of glymphatic clearance. Alzheimer’s Res. Ther. 2020 121 (12), 1–15. 10.1186/S13195-020-00688-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrer I, García MA, Carmona M, Andrés-Benito P, Torrejón-Escribano B, Garcia-Esparcia P, Del Rio JA, 2019. Involvement of oligodendrocytes in tau seeding and spreading in tauopathies. Front. Aging Neurosci 11, 112. 10.3389/fnagi.2019.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitzner D, Schnaars M, Van Rossum D, Krishnamoorthy G, Dibaj P, Bakhti M, Regen T, Hanisch UK, Simons M, 2011. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J. Cell Sci 124, 447–458. 10.1242/jcs.074088. [DOI] [PubMed] [Google Scholar]
- Flach K, Hilbrich I, Schiffmann A, Gärtner U, Krüger M, Leonhardt M, Waschipky H, Wick L, Arendt T, Holzer M, 2012. Tau oligomers impair artificial membrane integrity and cellular viability. J. Biol. Chem 287, 43223–43233. 10.1074/jbc.M112.396176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flannagan RS, Jaumouillé V, Grinstein S, 2012. The cell biology of phagocytosis. Annu. Rev. Pathol. Mech. Dis 10.1146/annurev-pathol-011811132445. [DOI] [PubMed] [Google Scholar]
- Flavin WP, Bousset L, Green ZC, Chu Y, Skarpathiotis S, Chaney MJ, Kordower JH, Melki R, Campbell EM, 2017. Endocytic vesicle rupture is a conserved mechanism of cellular invasion by amyloid proteins. Acta Neuropathol. 134, 629–653. 10.1007/s00401-017-1722-x. [DOI] [PubMed] [Google Scholar]
- Forabosco P, Ramasamy A, Trabzuni D, Walker R, Smith C, Bras J, Levine AP, Hardy J, Pocock JM, Guerreiro R, Weale ME, Ryten M, 2013. Insights into TREM2 biology by network analysis of human brain gene expression data. Neurobiol. Aging 34, 2699–2714. 10.1016/J.NEUROBIOLAGING.2013.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fox LM, William CM, Adamowicz DH, Pitstick R, Carlson GA, Spires-Jones TL, Hyman BT, 2011. Soluble tau species, not neurofibrillary aggregates, disrupt neural system integration in a tau transgenic model. J. Neuropathol. Exp. Neurol 70, 588–595. 10.1097/NEN.0B013E318220A658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman BA, Srinivasan K, Ayalon G, Meilandt WJ, Lin H, Huntley MA, Cao Y, Lee S-H, Haddick PCG, Ngu H, Modrusan Z, Larson JL, Kaminker JS, van der Brug MP, Hansen DV, 2018. Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Rep. 22, 832–847. 10.1016/j.celrep.2017.12.066. [DOI] [PubMed] [Google Scholar]
- Frost B, Jacks RL, Diamond MI, 2009. Propagation of tau misfolding from the outside to the inside of a cell. J. Biol. Chem 284, 12845–12852. 10.1074/JBC.M808759200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frost B, Hemberg M, Lewis J, Feany MB, 2014. Tau promotes neurodegeneration through global chromatin relaxation. Nat. Neurosci 17, 357–366. 10.1038/nn.3639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Funk KE, Mirbaha H, Jiang H, Holtzman DM, Diamond MI, 2015. Distinct therapeutic mechanisms of tau antibodies: promoting microglial clearance versus blocking neuronal uptake. J. Biol. Chem 290, 21652–21662. 10.1074/jbc.M115.657924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabbita SP, Johnson MF, Kobritz N, Eslami P, Poteshkina A, Varadarajan S, Turman J, Zemlan F, Harris-White ME, 2015. Oral TNFα modulation alters neutrophil infiltration, improves cognition and diminishes tau and amyloid pathology in the 3xTgAD mouse model. PLoS One 10, e0137305. 10.1371/journal.pone.0137305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garrett WS, Chen LM, Kroschewski R, Ebersold M, Turley S, Trombetta S, Galán JE, Mellman I, 2000. Developmental control of endocytosis in dendritic cells by Cdc42. Cell 102, 325–334. 10.1016/S0092-8674(00)00038-6. [DOI] [PubMed] [Google Scholar]
- Gerhard A, Watts J, Trender-Gerhard I, Turkheimer F, Banati RB, Bhatia K, Brooks DJ, 2004. In vivo imaging of microglial activation with [11C](R -PK11195 PET in corticobasal degeneration. Mov. Disord 19, 1221–1226. 10.1002/mds.20162. [DOI] [PubMed] [Google Scholar]
- Gerhard A, Trender-Gerhard I, Turkheimer F, Quinn NP, Bhatia KP, Brooks DJ, 2006. In vivo imaging of microglial activation with [11C]-PK11195 PET progresive supranuclear palsy. Mov. Disord 21, 89–93. 10.1002/mds.20668. [DOI] [PubMed] [Google Scholar]
- Gerson JE, Castillo-Carranza DL, Kayed R, 2014. Advances in therapeutics for neurodegenerative tauopathies: moving toward the specific targeting of the most toxic tau species. ACS Chem. Neurosci 5, 752–769. 10.1021/CN500143N. [DOI] [PubMed] [Google Scholar]
- Gey GO, Shapras P, Borysko E, 1954. Activities and responses of living cells and their components as recorded by cinephase microscopy and electron microscopy. Ann. N. Y. Acad. Sci 58, 1089–1109. 10.1111/j.1749-6632.1954.tb45895.x. [DOI] [PubMed] [Google Scholar]
- Ghosh S, Wu MD, Shaftel SS, Kyrkanides S, LaFerla FM, Olschowka JA, O’Banion MK, 2013. Sustained interleukin-1 overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J. Neurosci 33, 5053–5064. 10.1523/JNEUROSCI.4361-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghoshal N, García-Sierra F, Fu Y, Beckett LA, Mufson EJ, Kuret J, Berry RW, Binder LI, 2001. Tau-66: evidence for a novel tau conformation in Alzheimer’s disease. J. Neurochem 77, 1372–1385. 10.1046/j.1471-4159.2001.00346.x. [DOI] [PubMed] [Google Scholar]
- Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M, 2010. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845. 10.1126/science.1194637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorlovoy P, Larionov S, Pham TTH, Neumann H, 2009. Accumulation of tau induced in neurites by microglial proinflammatory mediators. FASEB J. 23, 2502–2513. 10.1096/fj.08-123877. [DOI] [PubMed] [Google Scholar]
- Gratuze M, Leyns CEG, Sauerbeck AD, St-Pierre MK, Xiong M, Kim N, Serrano JR, Tremblay MÈ, Kummer TT, Colonna M, Ulrich JD, Holtzman DM, 2020. Impact of TREM2R47Hvariant on tau pathology-induced gliosis and neurodegeneration. J. Clin. Invest 130, 4954–4968. 10.1172/JCI138179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gratuze M, Chen Y, Parhizkar S, Jain N, Strickland MR, Serrano JR, Colonna M, Ulrich JD, Holtzman DM, 2021. Activated microglia mitigate Aβ-associated tau seeding and spreading. J. Exp. Med 218. 10.1084/jem.20210542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grommes C, Lee CYD, Wilkinson BL, Jiang Q, Koenigsknecht-Talboo JL, Varnum B, Landreth GE, 2007. Regulation of microglial phagocytosis and inflammatory gene expression by Gas6 acting on the Axl/Mer family of tyrosine kinases. J. Neuroimmune Pharmacol 32 (3), 130–140. 10.1007/S11481-007-9090-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JSK, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert J-C, Amouyel P, Goate A, Rademakers R, Morgan K, Powell St. J, George-Hyslop P, Singleton A, Hardy J, 2013. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med 368, 117–127. 10.1056/nejmoa1211851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo C, Jeong HH, Hsieh YC, Klein HU, Bennett DA, De Jager PL, Liu Z, Shulman JM, 2018. Tau activates transposable elements in Alzheimer’s disease. Cell Rep. 23, 2874–2880. 10.1016/J.CELREP.2018.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo JL, Narasimhan S, Changolkar L, He Z, Stieber A, Zhang B, Gathagan RJ, Iba M, McBride JD, Trojanowski JQ, Lee VMY, 2016. Unique pathological tau conformers from Alzheimer’s brains transmit tau pathology in nontransgenic mice. J. Exp. Med 213, 2635–2654. 10.1084/JEM.20160833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, Golenbock DT, 2008. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat. Immunol 9, 857–865. 10.1038/ni.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanisch UK, Kettenmann H, 2007. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci 2007 1011 (10), 1387–1394. 10.1038/nn1997. [DOI] [PubMed] [Google Scholar]
- Hayflick L, Moorhead PS, 1961. The serial cultivation of human diploid cell strains. Exp. Cell Res 25, 585–621. 10.1016/0014-4827(61)90192-6. [DOI] [PubMed] [Google Scholar]
- Hickman SE, Allison EK, El Khoury J, 2008. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J. Neurosci 28, 8354–8360. 10.1523/JNEUROSCI.0616-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes BB, DeVos SL, Kfoury N, Li M, Jacks R, Yanamandra K, Ouidja MO, Brodsky FM, Marasa J, Bagchi DP, Kotzbauer PT, Miller TM, Papy-Garcia D, Diamond MI, 2013. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proc. Natl. Acad. Sci. U. S. A 110, E3138–E3147. 10.1073/pnas.1301440110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, Belaygorod L, Cairns NJ, Holtzman DM, Diamond MI, 2014. Proteopathic tau seeding predicts tauopathy in vivo. Proc. Natl. Acad. Sci. U. S. A 111, E4376–85. 10.1073/pnas.1411649111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holtman IR, Raj DD, Miller JA, Schaafsma W, Yin Z, Brouwer N, Wes PD, Möller T, Orre M, Kamphuis W, Hol EM, Boddeke EWGM, Eggen BJL, 2015. Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: a co-expression meta-analysis. Acta Neuropathol. Commun 3, 31. 10.1186/s40478-015-0203-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hopp SC, Lin Y, Oakley D, Roe AD, DeVos SL, Hanlon D, Hyman BT, 2018. The role of microglia in processing and spreading of bioactive tau seeds in Alzheimer’s disease. J. Neuroinflamm 15, 269. 10.1186/s12974-018-1309-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang W, Zhou Y, Tu L, Ba Z, Huang J, Huang N, Luo Y, 2020. TDP-43: from Alzheimer’s disease to limbic-predominant age-related TDP-43 encephalopathy. Front. Mol. Neurosci 13, 26. 10.3389/FNMOL.2020.00026/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang Z-Y, Barreda DR, Worth RG, Indik ZK, Kim M-K, Chien P, Schreiber AD, 2006. Differential kinase requirements in human and mouse Fc-gamma receptor phagocytosis and endocytosis. J. Leukoc. Biol 80, 1553–1562. 10.1189/jlb.0106019. [DOI] [PubMed] [Google Scholar]
- Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM-Y, 2013. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J. Neurosci 33, 1024–1037. 10.1523/JNEUROSCI.2642-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iba M, McBride JD, Guo JL, Zhang B, Trojanowski JQ, Lee VMY, 2015. Tau pathology spread in PS19 tau transgenic mice following locus coeruleus (LC) injections of synthetic tau fibrils is determined by the LC’s afferent and efferent connections. Acta Neuropathol. 130, 349–362. 10.1007/s00401015-1458-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibarra-Bracamontes VJ, Escobar-Herrera J, Kristofikova Z, Rípova D, Florán-Garduño B, Garcia-Sierra F, 2020. Early but not late conformational changes of tau in association with ubiquitination of neurofibrillary pathology in Alzheimer’s disease brains. Brain Res. 1744. 10.1016/j.brainres.2020.146953. [DOI] [PubMed] [Google Scholar]
- Iliff JJ, Chen MJ, Plog BA, Zeppenfeld DM, Soltero M, Yang L, Singh I, Deane R, Nedergaard M, 2014. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J. Neurosci 34, 16180–16193. 10.1523/JNEUROSCI.3020-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irwin DJ, 2016. Tauopathies as clinicopathological entities. Park. Relat. Disord 22, S29–S33. 10.1016/j.parkreldis.2015.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iversen TG, Skotland T, Sandvig K, 2011. Endocytosis and intracellular transport of nanoparticles: present knowledge and need for future studies. Nano Today. 10.1016/j.nantod.2011.02.003. [DOI] [Google Scholar]
- Jaumouillé V, Waterman CM, 2020. Physical constraints and forces involved in phagocytosis. Front. Immunol 11, 1097. 10.3389/FIMMU.2020.01097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaworski T, Lechat B, Demedts D, Gielis L, Devijver H, Borghgraef P, Duimel H, Verheyen F, Kügler S, Van Leuven F, 2011. Dendritic degeneration, neurovascular defects, and inflammation precede neuronal loss in a mouse model for tau-mediated neurodegeneration. Am. J. Pathol 179, 2001–2015. 10.1016/j.ajpath.2011.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE, Ransohoff RM, Lamb BT, Landreth GE, 2017. Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J. Neurosci 37, 637–647. 10.1523/JNEUROSCI.2110-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang T, Tan L, Zhu XC, Zhou JS, Cao L, Tan MS, Wang HF, Chen Q, Zhang YD, Yu JT, 2015. Silencing of TREM2 exacerbates tau pathology, neurodegenerative changes, and spatial learning deficits in P301S tau transgenic mice. Neurobiol. Aging 36, 3176–3186. 10.1016/J.NEUROBIOLAGING.2015.08.019. [DOI] [PubMed] [Google Scholar]
- Jiang T, Zhang Y-D, Chen Q, Gao Q, Zhu X-C, Zhou J-S, Shi J-Q, Lu H, Tan L, Yu J-T, 2016. TREM2 modifies microglial phenotype and provides neuroprotection in P301S tau transgenic mice. Neuropharmacology 105, 196–206. 10.1016/j.neuropharm.2016.01.028. [DOI] [PubMed] [Google Scholar]
- Johnson GVW, Litersky JM, Whitaker JN, 1991. Proteolysis of microtubule-associated protein 2 and tubulin by cathepsin D. J. Neurochem 57, 1577–1583. 10.1111/j.1471-4159.1991.tb06354.x. [DOI] [PubMed] [Google Scholar]
- Karch CM, Goate AM, 2015. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 77, 43–51. 10.1016/j.biopsych.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaufman SK, Sanders DW, Thomas TL, Ruchinskas AJ, Vaquer-Alicea J, Sharma AM, Miller TM, Diamond MI, 2016. Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92, 796–812. 10.1016/j.neuron.2016.09.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, Itzkovitz S, Colonna M, Schwartz M, Amit I, 2017. A unique microglia type associated with restricting development of Alzheimer’s disease. e17 Cell 169, 1276–1290. 10.1016/J.CELL.2017.05.018. [DOI] [PubMed] [Google Scholar]
- Kettenmann H, Hanisch U-K, Noda M, Verkhratsky A, 2011. Physiology of microglia. Physiol. Rev 91, 461–553. 10.1152/physrev.00011.2010. [DOI] [PubMed] [Google Scholar]
- Kingham PJ, Pocock JM, 2001. Microglial secreted cathepsin B induces neuronal apoptosis. J. Neurochem 76, 1475–1484. 10.1046/j.1471-4159.2001.00146.x. [DOI] [PubMed] [Google Scholar]
- Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM, 2005. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J. Neurosci 25, 8843–8853. 10.1523/JNEUROSCI.2868-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, Cribbs DH, LaFerla FM, 2011. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal β-catenin pathway function in an Alzheimer’s disease model. J. Immunol 187, 6539–6549. 10.4049/jimmunol.1100620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koga S, Parks A, Kasanuki K, Sanchez-Contreras M, Baker MC, Josephs KA, Ahlskog JE, Uitti RJ, Graff-Radford N, van Gerpen JA, Wszolek ZK, Rademakers R, Dickson DW, 2017. Cognitive impairment in progressive supranuclear palsy is associated with tau burden. Mov. Disord 32, 1772–1779. 10.1002/mds.27198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovacs GG, 2015. Invited review: neuropathology of tauopathies: principles and practice. Neuropathol. Appl. Neurobiol 41, 3–23. 10.1111/NAN.12208. [DOI] [PubMed] [Google Scholar]
- Kovacs GG, Lukic MJ, Irwin DJ, Arzberger T, Respondek G, Lee EB, Coughlin D, Giese A, Grossman M, Kurz C, McMillan CT, Gelpi E, Compta Y, van Swieten JC, Laat LD, Troakes C, Al-Sarraj S, Robinson JL, Roeber S, Xie SX, Lee VMY, Trojanowski JQ, Höglinger GU, 2020. Distribution patterns of tau pathology in progressive supranuclear palsy. Acta Neuropathol. 140, 99–119. 10.1007/s00401-020-02158-2. [DOI] [PMC free article] [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 DJJ, Smith STT, Tweet G, Humulock Z, Zrzavy T, Conde-Sanroman P, Gacias M, Weng Z, Chen H, Tjon E, Mazaheri F, Hartmann K, Madi A, Ulrich JDD, Glatzel M, Worthmann A, Heeren J, Budnik B, Lemere C, Ikezu T, Heppner FLL, Litvak V, Holtzman DMM, Lassmann H, Weiner HLL, Ochando J, Haass C, Butovsky O, O’Loughlin E, Xu Y, Fanek Z, Greco DJJ, Smith STT, Tweet G, Humulock Z, Zrzavy T, Conde-Sanroman P, Gacias M, Weng Z, Chen H, Tjon E, Mazaheri F, Hartmann K, Madi A, Ulrich JDD, Glatzel M, Worthmann A, Heeren J, Budnik B, Lemere C, Ikezu T, Heppner FLL, Litvak V, Holtzman DMM, Lassmann H, Weiner HLL, Ochando J, Haass C, Butovsky O, 2017. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. e9 Immunity 47, 566–581. 10.1016/j.immuni.2017.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kress B, Iliff J, Xia M, Wang M, H.W.-A. of, 2014, undefined, 2014. Impairment of paravascular clearance pathways in the aging brain. Wiley Online Libr. 76, 845–861. 10.1002/ana.24271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Clos AL, Jackson GR, Kayed R, 2011. Tau oligomers impair memory and induce synaptic and mitochondrial dysfunction in wild-type mice. Mol. Neurodegener 6, 1–14. 10.1186/1750-1326-6-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee M, McGeer E, McGeer PL, 2015. Activated human microglia stimulate neuroblastoma cells to upregulate production of beta amyloid protein and tau: implications for Alzheimer’s disease pathogenesis. Neurobiol. Aging 36, 42–52. 10.1016/j.neurobiolaging.2014.07.024. [DOI] [PubMed] [Google Scholar]
- Lee SH, Le Pichon CE, Adolfsson O, Gafner V, Pihlgren M, Lin H, Solanoy H, Brendza R, Ngu H, Foreman O, Chan R, Ernst JA, DiCara D, Hotzel I, Srinivasan K, Hansen DV, Atwal J, Lu Y, Bumbaca D, Pfeifer A, Watts RJ, Muhs A, Scearce-Levie K, Ayalon G, 2016. Antibody-mediated targeting of tau in vivo does not require effector function and microglial engagement. Cell Rep. 16, 1690–1700. 10.1016/j.celrep.2016.06.099. [DOI] [PubMed] [Google Scholar]
- Lewis WH, 1931. Pinocytosis. Bull Johns. Hopkins Hosp. 49, 17–26. [Google Scholar]
- Leyns CEG, Ulrich JD, Finn MB, Stewart FR, Koscal LJ, Serrano JR, Robinson GO, Anderson E, Colonna M, Holtzman DM, 2017. TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc. Natl. Acad. Sci. U. S. A 114, 11524–11529. 10.1073/PNAS.1710311114/-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leyns CEG, Gratuze M, Narasimhan S, Jain N, Koscal LJ, Jiang H, Manis M, Colonna M, Lee VMY, Ulrich JD, Holtzman DM, 2019. TREM2 function impedes tau seeding in neuritic plaques. Nat. Neurosci 228 (22), 1217–1222. 10.1038/s41593-019-0433-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Liu L, Barger SW, Griffin WST, 2003. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J. Neurosci 23, 1605–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim JP, Gleeson PA, 2011. Macropinocytosis: an endocytic pathway for internalising large gulps. Immunol. Cell Biol 89, 836–843. 10.1038/ICB.2011.20. [DOI] [PubMed] [Google Scholar]
- Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland C, Duff K, 2012. Trans-synaptic spread of tau pathology in vivo. PLoS One 7, e31302. 10.1371/journal.pone.0031302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Dai Y, Li Q, Chen C, Chen H, Song Y, Hua F, Zhang Z, 2020. Beta-amyloid activates NLRP3 inflammasome via TLR4 in mouse microglia. Neurosci. Lett 736, 135279 10.1016/J.NEULET.2020.135279. [DOI] [PubMed] [Google Scholar]
- Liu YU, Ying Y, Li Y, Eyo UB, Chen T, Zheng J, Umpierre AD, Zhu J, Bosco DB, Dong H, Wu LJ, 2019. Neuronal network activity controls microglial process surveillance in awake mice via norepinephrine signaling. Nat. Neurosci 22, 1771–1781. 10.1038/s41593-019-0511-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Z, Xu E, Zhao HT, Cole T, West AB, 2020. LRRK2 and Rab10 coordinate macropinocytosis to mediate immunological responses in phagocytes. EMBO J. 39, e104862 10.15252/embj.2020104862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loeffler DA, Camp DM, Bennett DA, 2008. Plaque complement activation and cognitive loss in Alzheimer’s disease. J. Neuroinflamm 5, 1–8. 10.1186/1742-2094-5-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- López-González I, Schlüter A, Aso E, Garcia-Esparcia P, Ansoleaga B, LLorens F, Carmona M, Moreno J, Fuso A, Portero-Otin M, Pamplona R, Pujol A, Ferrer I, 2015. Neuroinflammatory signals in Alzheimer disease and APP/PS1 transgenic mice: correlations with plaques, tangles, and oligomeric species. J. Neuropathol. Exp. Neurol 74, 319–344. 10.1097/NEN.0000000000000176. [DOI] [PubMed] [Google Scholar]
- Lowry JR, Klegeris A, 2018. Emerging roles of microglial cathepsins in neurodegenerative disease. Brain Res. Bull 139, 144–156. 10.1016/J.BRAINRESBULL.2018.02.014. [DOI] [PubMed] [Google Scholar]
- Luo W, Liu W, Hu X, Hanna M, Caravaca A, Paul SM, 2015. Microglial internalization and degradation of pathological tau is enhanced by an anti-tau monoclonal antibody. Sci. Rep 5, 11161. 10.1038/srep11161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Majerova P, Zilkova M, Kazmerova Z, Kovac A, Paholikova K, Kovacech B, Zilka N, Novak M, 2014. Microglia display modest phagocytic capacity for extracellular tau oligomers. J. Neuroinflamm 11, 161. 10.1186/s12974-014-0161-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Majumdar A, Cruz D, Asamoah N, Buxbaum A, Sohar I, Lobel P, Maxfield FR, 2007. Activation of microglia acidifies lysosomes and leads to degradation of Alzheimer amyloid fibrils. Mol. Biol. Cell 18, 1490–1496. 10.1091/mbc.e06-10-0975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mandrekar S, Jiang Q, Lee CYD, Koenigsknecht-Talboo J, Holtzman DM, Landreth GE, 2009. Microglia mediate the clearance of soluble aβ through fluid phase macropinocytosis. J. Neurosci 29, 4252–4262. 10.1523/JNEUROSCI.5572-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maphis N, Xu G, Kokiko-Cochran ON, Cardona AE, Ransohoff RM, Lamb BT, Bhaskar K, 2015a. Loss of tau rescues inflammation-mediated neurodegeneration. Front. Neurosci 9, 196. 10.3389/fnins.2015.00196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, Lamb BT, Bhaskar K, 2015b. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain 138, 1738–1755. 10.1093/brain/awv081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maphis N, Jiang S, Xu G, Kokiko-Cochran ON, Roy SM, Van Eldik LJ, Watterson DM, Lamb BT, Bhaskar K, 2016. Selective suppression of the α isoform of p38 MAPK rescues late-stage tau pathology. Alzheimers Res. Ther 8, 54. 10.1186/s13195-016-0221-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martini-Stoica H, Cole AL, Swartzlander DB, Chen F, Wan Y-W, Bajaj L, Bader DA, Lee VMY, Trojanowski JQ, Liu Z, Sardiello M, Zheng H, 2018. TFEB enhances astroglial uptake of extracellular tau species and reduces tau spreading. J. Exp. Med 215, 2355–2377. 10.1084/jem.20172158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meisl G, Hidari E, Allinson K, Rittman T, DeVos SL, Sanchez JS, Xu CK, Duff KE, Johnson KA, Rowe JB, Hyman BT, Knowles TPJ, Klenerman D, 2021. In vivo rate-determining steps of tau seed accumulation in Alzheimer’s disease. Sci. Adv 7. 10.1126/sciadv.abh1448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Micheva K, Ramjaun A, Kay B, letters PM-F, 1997. undefined, n.d. SH3 Domain-dependent Interactions of Endophilin with Amphiphysin. Elsevier. [DOI] [PubMed] [Google Scholar]
- Minami SS, Sun B, Popat K, Kauppinen T, Pleiss M, Zhou Y, Ward ME, Floreancig P, Mucke L, Desai T, 2012. Selective targeting of microglia by quantum dots. J. Neuroinflamm 9, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minett T, Classey J, Matthews FE, Fahrenhold M, Taga M, Brayne C, Ince PG, Nicoll JAR, Boche D, CFAS MRC, 2016. Microglial immunophenotype in dementia with Alzheimer’s pathology. J. Neuroinflamm 13, 135. 10.1186/s12974-016-0601-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morales I, Jiménez JM, Mancilla M, Maccioni RB, 2013. Tau oligomers and fibrils induce activation of microglial cells. J. Alzheimers Dis 37, 849–856. 10.3233/JAD-131843. [DOI] [PubMed] [Google Scholar]
- Mudher A, Colin M, Dujardin S, Medina M, Dewachter I, Alavi Naini SM, Mandelkow EM, Mandelkow E, Buée L, Goedert M, Brion JP, 2017. What is the evidence that tau pathology spreads through prion-like propagation? Acta Neuropathol. Commun 10.1186/s40478-017-0488-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, Orr ME, 2018. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17, 12840. 10.1111/ACEL.12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ, Castellani RJ, Crain BJ, Davies P, Tredici K., Del, Duyckaerts C, Frosch MP, Haroutunian V, Hof PR, Hulette CM, Hyman BT, Iwatsubo T, Jellinger KA, Jicha GA, Kövari E, Kukull WA, Leverenz JB, Love S, MacKenzie IR, Mann DM, Masliah E, McKee AC, Montine TJ, Morris JC, Schneider JA, Sonnen JA, Thal DR, Trojanowski JQ, Troncoso JC, Wisniewski T, Woltjer RL, Beach TG, 2012. Correlation of alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J. Neuropathol. Exp. Neurol 10.1097/NEN.0b013e31825018f7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neniskyte U, Brown GC, 2013. Lactadherin/MFG-E8 is essential for microglia-mediated neuronal loss and phagoptosis induced by amyloid β. J. Neurochem 126, 312–317. 10.1111/jnc.12288. [DOI] [PubMed] [Google Scholar]
- Ni J, Wu Z, Peterts C, Yamamoto K, Qing H, Nakanishi H, 2015. The critical role of proteolytic relay through cathepsins B and E in the phenotypic change of microglia/macrophage. J. Neurosci 35, 12488–12501. 10.1523/JNEUROSCI.1599-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niewiadomska G, Niewiadomski W, Steczkowska M, Gasiorowska A, 2021. Tau oligomers neurotoxicity. Life 11, 28. 10.3390/LIFE11010028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nimmerjahn A, Kirchhoff F, Helmchen F, 2005. Neuroscience: resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Sci. (80-. ) 308, 1314–1318. 10.1126/science.1110647. [DOI] [PubMed] [Google Scholar]
- Norbury CC, Chambers BJ, Prescott AR, Ljunggren HG, Watts C, 1997. Constitutive macropinocytosis allows TAP-dependent major histocompatibility compex class I presentation of exogenous soluble antigen by bone marrow-derived dendritic cells. Eur. J. Immunol 27, 280–288. 10.1002/eji.1830270141. [DOI] [PubMed] [Google Scholar]
- Nübling G, Schuberth M, Feldmer K, Giese A, Holdt LM, Teupser D, Lorenzl S, 2017. Cathepsin S increases tau oligomer formation through limited cleavage, but only IL-6, not cathespin S serum levels correlate with disease severity in the neurodegenerative tauopathy progressive supranuclear palsy. Exp. Brain Res 235, 2407–2412. 10.1007/s00221-017-4978-4. [DOI] [PubMed] [Google Scholar]
- Oberstein TJ, Utz J, Spitzer P, Klafki HW, Wiltfang J, Lewczuk P, Kornhuber J, Maler JM, 2021. The role of cathepsin B in the degradation of Aβ and in the production of Aβ peptides starting with Ala2 in cultured astrocytes. Front. Mol. Neurosci 0, 250. 10.3389/FNMOL.2020.615740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odawara T, Iseki E, Kosaka K, Akiyama H, Ikeda K, Yamamoto T, 1995. Investigation of tau-2 positive microglia-like cells in the subcortical nuclei of human neurodegenerative disorders. Neurosci. Lett 192, 145–148. 10.1016/0304-3940(95)11595-N. [DOI] [PubMed] [Google Scholar]
- Orr ME, Sullivan AC, Frost B, 2017. A brief overview of tauopathy: causes, consequences, and therapeutic strategies. Trends Pharmacol. Sci 10.1016/j.tips.2017.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pampuscenko K, Morkuniene R, Sneideris T, Smirnovas V, Budvytyte R, Valincius G, Brown GC, Borutaite V, 2020. Extracellular tau induces microglial phagocytosis of living neurons in cell cultures. J. Neurochem 154, 316–329. 10.1111/JNC.14940. [DOI] [PubMed] [Google Scholar]
- Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross CT, 2011. Synaptic pruning by microglia is necessary for normal brain development. Sci. (80-.) 333, 1456–1458. 10.1126/science.1202529. [DOI] [PubMed] [Google Scholar]
- Parachikova A, Agadjanyan MG, Cribbs DH, Blurton-Jones M, Perreau V, Rogers J, Beach TG, Cotman CW, 2007. Inflammatory changes parallel the early stages of Alzheimer disease. Neurobiol. Aging 28, 1821–1833. 10.1016/j.neurobiolaging.2006.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, Savard M, Lussier FZ, Tissot C, Karikari TK, Ottoy J, Mathotaarachchi S, Stevenson J, Massarweh G, Schöll M, de Leon MJ, Soucy J-P, Edison P, Blennow K, Zetterberg H, Gauthier S, Rosa-Neto P, 2021. Microglial activation and tau propagate jointly across Braak stages. Nat. Med 2021, 1–8. 10.1038/s41591-021-01456-w. [DOI] [PubMed] [Google Scholar]
- Patel TK, Habimana-Griffin L, Gao X, Xu B, Achilefu S, Alitalo K, McKee CA, Sheehan PW, Musiek ES, Xiong C, Coble D, Holtzman DM, 2019. Dural lymphatics regulate clearance of extracellular tau from the CNS. Mol. Neurodegener 14, 11. 10.1186/s13024-019-0312-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng W, Achariyar TM, Li B, Liao Y, Mestre H, Hitomi E, Regan S, Kasper T, Peng S, Ding F, Benveniste H, Nedergaard M, Deane R, 2016. Suppression of glymphatic fluid transport in a mouse model of Alzheimer’s disease. Neurobiol. Dis 93, 215–225. 10.1016/J.NBD.2016.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez-Nievas BG, Stein TD, Tai H-C, Dols-Icardo O, Scotton TC, Barroeta-Espar I, Fernandez-Carballo L, de Munain EL, Perez J, Marquie M, Serrano-Pozo A, Frosch MP, Lowe V, Parisi JE, Petersen RC, Ikonomovic MD, López OL, Klunk W, Hyman BT, Gómez-Isla T, 2013. Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 136, 2510–2526. 10.1093/brain/awt171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pimenova AA, Marcora E, Goate AM, 2017. A tale of two genes: microglial apoe and trem2. Immunity. 10.1016/j.immuni.2017.08.015. [DOI] [PubMed] [Google Scholar]
- Pimenova AA, Raj T, Goate AM, 2018. Untangling genetic risk for Alzheimer’s disease. Biol. Psychiatry 10.1016/j.biopsych.2017.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polanco JC, Hand GR, Briner A, Li C, Götz J, 2021a. Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol. Acta Neuropathol. 141, 235–256. 10.1007/s00401020-02254-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB, 2004. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp. Cell Res 295, 245–257. 10.1016/j.yexcr.2004.01.002. [DOI] [PubMed] [Google Scholar]
- Ramjaun AR, McPherson PS, 1998. Multiple amphiphysin II splice variants display differential clathrin binding: Identification of two distinct clathrin-binding sites. J. Neurochem 70, 2369–2376. 10.1046/J.1471-4159.1998.70062369.X. [DOI] [PubMed] [Google Scholar]
- Rangaraju S, Dammer EB, Raza SA, Rathakrishnan P, Xiao H, Gao T, Duong DM, Pennington MW, Lah JJ, Seyfried NT, Levey AI, 2018. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer’s disease. Mol. Neurodegener 13, 1–25. 10.1186/S13024-018-0254-8/FIGURES/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ranson PA, Thomas WE, 1991. Pinocytosis as a select marker of ramified microglia in vivo and in vitro. J. Histochem. Cytochem 39, 853–858. 10.1177/39.6.2033242. [DOI] [PubMed] [Google Scholar]
- Rauch JN, Luna G, Guzman E, Audouard M, Challis C, Sibih YE, Leshuk C, Hernandez I, Wegmann S, Hyman BT, Gradinaru V, Kampmann M, Kosik KS, 2020. LRP1 is a master regulator of tau uptake and spread. Nature 580, 381–385. 10.1038/s41586-020-2156-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redka DS, Gütschow M, Grinstein S, Canton J, 2018. Differential ability of proinflammatory and anti-inflammatory macrophages to perform macropinocytosis. Mol. Biol. Cell 29, 53–65. 10.1091/mbc.E17-06-0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rexach JE, Polioudakis D, Yin A, Swarup V, Chang TS, Nguyen T, Sarkar A, Chen L, Huang J, Lin LC, Seeley W, Trojanowski JQ, Malhotra D, Geschwind DH, 2020. Tau pathology drives dementia risk-associated gene networks toward chronic inflammatory states and immunosuppression. Cell Rep. 33. 10.1016/j.celrep.2020.108398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C, 2020. Increased oxidative stress, hyperphosphorylation of tau, and dystrophic microglia in the hippocampus of aged Tupaia belangeri. Glia 68, 1775–1793. 10.1002/glia.23804. [DOI] [PubMed] [Google Scholar]
- Rosales C, Uribe-Querol E, 2017. Phagocytosis: a fundamental process in immunity. Biomed. Res. Int 2017. 10.1155/2017/9042851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saha K, Kim ST, Yan B, Miranda OR, Alfonso FS, Shlosman D, Rotello PVM, 2013. Surface functionality of nanoparticles determines cellular uptake mechanisms in mammalian cells. Small 9, 300. 10.1002/sMLL.201201129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez-Mejias E, Navarro V, Jimenez S, Sanchez-Mico M, Sanchez-Varo R, Nuñez-Diaz C, Trujillo-Estrada L, Davila JC, Vizuete M, Gutierrez A, Vitorica J, 2016. Soluble phospho-tau from Alzheimer’s disease hippocampus drives microglial degeneration. Acta Neuropathol. 132, 897–916. 10.1007/s00401-016-1630-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McCowan E, Forster C, Yue M, Orne J, Janus C, Mariash A, Kuskowski M, Hyman B, Hutton M, Ashe KH, 2005. Medicine: tau suppression in a neurodegenerative mouse model improves memory function. Sci. (80-.) 309, 476–481. 10.1126/science.1113694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarrazin S, Lamanna WC, Esko JD, 2011. Heparan sulfate proteoglycans. Cold Spring Harb. Perspect. Biol 3, 1–33. 10.1101/cshperspect.a004952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sayed FA, Telpoukhovskaia M, Kodama L, Li Y, Zhou Y, Le D, Hauduc A, Ludwig C, Gao F, Clelland C, Zhan L, Cooper YA, Davalos D, Akassoglou K, Coppola G, Gan L, 2018. Differential effects of partial and complete loss of TREM2 on microglial injury response and tauopathy. Proc. Natl. Acad. Sci. U. S. A 115, 10172–10177. 10.1073/pnas.1811411115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sayed FA, Kodama L, Fan L, Carling GK, Udeochu JC, Le D, Li Q, Zhou L, Wong MY, Horowitz R, Ye P, Mathys H, Wang M, Niu X, Mazutis L, Jiang X, Wang X, Gao F, Brendel M, Telpoukhovskaia M, Tracy TE, Frost G, Zhou Y, Li Y, Qiu Y, Cheng Z, Yu G, Hardy J, Coppola G, Wang F, DeTure MA, Zhang B, Xie L, Trajnowski JQ, Lee VMY, Gong S, Sinha SC, Dickson DW, Luo W, Gan L, 2021. AD-linked R47H-TREM2 mutation induces disease-enhancing microglial states via AKT hyperactivation. Sci. Transl. Med 13, eabe3947. 10.1126/scitranslmed.abe3947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schlegel K, Awwad K, Heym RG, Holzinger D, Doell A, Barghorn S, Jahn TR, Klein C, Mordashova Y, Schulz M, Gasparini L, 2019. N368-Tau fragments generated by legumain are detected only in trace amount in the insoluble Tau aggregates isolated from AD brain. Acta Neuropathol. Commun 7, 177. 10.1186/s40478-019-0831-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schöll M, Lockhart SN, Schonhaut DR, O’Neil JP, Janabi M, Ossenkoppele R, Baker SL, Vogel JW, Faria J, Schwimmer HD, Rabinovici GD, Jagust WJ, 2016. PET imaging of tau deposition in the aging human brain. Neuron 89, 971–982. 10.1016/j.neuron.2016.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schuh C, Wimmer I, Hametner S, Haider L, Van Dam AM, Liblau RS, Smith KJ, Probert L, Binder CJ, Bauer J, Bradl M, Mahad D, Lassmann H, 2014. Oxidative tissue injury in multiple sclerosis is only partly reflected in experimental disease models. Acta Neuropathol. 128, 247–266. 10.1007/s00401014-1263-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seshadri S, Fitzpatrick AL, Ikram MA, DeStefano AL, Gudnason V, Boada M, Bis JC, Smith AV, Carassquillo MM, Lambert JC, Harold D, Schrijvers EMC, Ramirez-Lorca R, Debette S, Longstreth WT, Janssens ACJW, Pankratz VS, Dartigues JF, Hollingworth P, Aspelund T, Hernandez I, Beiser A, Kuller LH, Koudstaal PJ, Dickson DW, Tzourio C, Abraham R, Antunez C, Du Y, Rotter JI, Aulchenko YS, Harris TB, Petersen RC, Berr C, Owen MJ, Lopez-Arrieta J, Varadarajan BN, Becker JT, Rivadeneira F, Nalls MA, Graff-Radford NR, Campion D, Auerbach S, Rice K, Hofman A, Jonsson PV, Schmidt H, Lathrop M, Mosley TH, Au R, Psaty BM, Uitterlinden AG, Farrer LA, Lumley T, Ruiz A, Williams J, Amouyel P, Younkin SG, Wolf PA, Launer LJ, Lopez OL, Van Duijn CM, Breteler MMB, 2010. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA - J. Am. Med. Assoc 303, 1832–1840. 10.1001/jama.2010.574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Holtzman DM, 2018. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol 10.1038/s41577-018-0051-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Manis M, Long J, Wang K, Sullivan PM, Serrano JR, Hoyle R, Holtzman DM, 2019. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J. Exp. Med 216, 2546–2561. 10.1084/jem.20190980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimura H, Schwartz D, Gygi SP, Kosik KS, 2004. CHIP-Hsc70 complex ubiquitinates phosphorylated tau and enhances cell survival. J. Biol. Chem 279, 4869–4876. 10.1074/jbc.M305838200. [DOI] [PubMed] [Google Scholar]
- Siklos M, BenAissa M, Thatcher GRJ, 2015. Cysteine proteases as therapeutic targets: Does selectivity matter? A systematic review of calpain and cathepsin inhibitors. Acta Pharm. Sin. B 10.1016/j.apsb.2015.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silva I, Silva J, Ferreira R, Trigo D, 2021. Glymphatic system, AQP4, and their implications in Alzheimer’s disease. Neurol. Res. Pr 3. 10.1186/S42466-021-00102-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sims R, Van Der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, Kunkle BW, Boland A, Raybould R, Bis JC, Martin ER, Grenier-Boley B, Heilmann-Heimbach S, Chouraki V, Kuzma AB, Sleegers K, Vronskaya M, Ruiz A, Graham RR, Olaso R, Hoffmann P, Grove ML, Vardarajan BN, Hiltunen M, Nöthen MM, White CC, Hamilton-Nelson KL, Epelbaum J, Maier W, Choi SH, Beecham GW, Dulary C, Herms S, Smith AV, Funk CC, Derbois C, Forstner AJ, Ahmad S, Li H, Bacq D, Harold D, Satizabal CL, Valladares O, Squassina A, Thomas R, Brody JA, Qu L, Sánchez-Juan P, Morgan T, Wolters FJ, Zhao Y, Garcia FS, Denning N, Fornage M, Malamon J, Naranjo MCD, Majounie E, Mosley TH, Dombroski B, Wallon D, Lupton MK, Dupuis J, Whitehead P, Fratiglioni L, Medway C, Jian X, Mukherjee S, Keller L, Brown K, Lin H, Cantwell LB, Panza F, McGuinness B, Moreno-Grau S, Burgess JD, Solfrizzi V, Proitsi P, Adams HH, Allen M, Seripa D, Pastor P, Cupples LA, Price ND, Hannequin D, Frank-García A, Levy D, Chakrabarty P, Caffarra P, Giegling I, Beiser AS, Giedraitis V, Hampel H, Garcia ME, Wang X, Lannfelt L, Mecocci P, Eiriksdottir G, Crane PK, Pasquier F, Boccardi V, Henández I, Barber RC, Scherer M, Tarraga L, Adams PM, Leber M, Chen Y, Albert MS, Riedel-Heller S, Emilsson V, Beekly D, Braae A, Schmidt R, Blacker D, Masullo C, Schmidt H, Doody RS, Spalletta G Jr., Fairchild WTL, Bossù TJ, Lopez P, Frosch OL, Sacchinelli MP, Ghetti E, Yang B, Huebinger Q, Jessen RM, Li F, Kamboh S, Morris MI, Sotolongo-Grau JC, Katz O, Corcoran MJ, Dunstan C, Braddel M, Thomas A, Meggy C, Marshall A, Gerrish R, Chapman A, Aguilar J, Taylor M, Hill S, Fairén M, Hodges MD, Vellas A, Soininen B, Kloszewska H, Daniilidou I, Uphill M, Patel J, Hughes Y, Lord JT, Turton J, Hartmann J, Cecchetti AM, Fenoglio R, Serpente C, Arcaro M, Caltagirone M, Orfei C, Ciaramella MD, Pichler A, Mayhaus S, Gu M, Lleó W, Fortea A, Blesa J, Barber R, Brookes IS, Cupidi K, Maletta C, Carrell RG, Sorbi D, Moebus S, Urbano S, Pilotto M, Kornhuber A, Bosco J, Todd P, Craig S, Johnston D, Gill J, Lawlor M, Lynch B, Fox A, Hardy NC, Albin J, Apostolova RL, Arnold LG, Asthana SE, Atwood S, Baldwin CS, Barnes CT, Barral LL, Beach S, Becker TG, Bigio JT, Bird EH, Boeve TD, Bowen BF, Boxer JD, Burke A, Burns JR, Buxbaum JM, Cairns JD, Cao NJ, Carlson C, Carlsson CS, Carney CM, Carrasquillo RM, Carroll MM, Diaz SL, Chui CC, Clark HC, Cribbs DG, Crocco DH, Decarli EA, Dick C, Duara M, Evans R, Faber DA, Fallon KM, Fardo KB, Farlow DW, Ferris MR, Foroud S, Galasko TM, Gearing DR, Geschwind M, Gilbert DH, Graff-Radford JR, Green NR, Growdon RC, Hamilton JH, Harrell RL, Honig LE, Huentelman LS, Hulette MJ, Hyman CM, Jarvik BT, Abner GP, Jin E, Jun LW, Karydas G, Kaye A, Kim JA, Kowall R, Kramer NW, Laferla JH, Lah FM, Leverenz JJ, Levey JB, Li AI, Lieberman G, Lunetta AP, Lyketsos KL, Marson CG, Martiniuk DC, Mash F, Masliah DC, McCormick E, McCurry WC, McDavid SM, McKee AN, Mesulam AC, Miller M, Miller BL, Miller CA, Murrell JW, Myers JR, O’Bryant AJ, Olichney S, Pankratz JM, Parisi VS, Paulson JE, Perry HL, Peskind W, Pierce E, Poon A, Potter WW, Quinn H, Raj JF, Raskind A, Reisberg M, Reitz B, Ringman C, Roberson JM, Rogaeva ED, Rosen E, Rosenberg HJ, Sager RN, Saykin MA, Schneider AJ, Schneider JA, Seeley LS, Smith WW, Sonnen AG, Spina JA, Stern S, Swerdlow RA, Tanzi RH, Thornton-Wells RE, Trojanowski TA, Troncoso JQ, Van Deerlin JC, Van Eldik VM, Vinters LJ, Vonsattel HV, Weintraub JP, Welsh-Bohmer S, Wilhelmsen KA, Williamson KC, Wingo J, Woltjer TS, Wright RL, Yu CB, Yu CE, Garzia L, Golamaully F, Septier F, Engelborghs G, Vandenberghe S, De Deyn R, Fernadez PP, Benito CM, Thonberg YA, Forsell H, Lilius C, Kinhult-Stählbom L, Kilander A, Brundin L, Concari R, Helisalmi L, Koivisto S, Haapasalo AM, Dermecourt A, Fievet V, Hanon N, Dufouil O, Brice C, Ritchie A, Dubois K, Himali B, Keene JJ, Tschanz CD, Fitzpatrick J, Kukull AL, Norton WA, Aspelund M, Larson T, Munger EB, Rotter R, Lipton JI, Bullido RB, Hofman MJ, Montine A, Coto TJ, Boerwinkle E, Petersen E, Alvarez RC, Rivadeneira V, Reiman F, Gallo EM, O’Donnell M, Reisch CJ, Brani JS, Royall AC, Dichgans DR, Sano M, Galimberti M, St George-Hyslop D, Scarpini P, Tsuang E, Mancuso DW, Bonuccelli M, Winslow U, Daniele AR, Wu A, Peters CK, Nacmias O, Riemenschneider B, Heun M, Brayne R, Rubinsztein C, Bras DC, Guerreiro J, Al-Chalabi R, Shaw A, Collinge CE, Tsolaki J, Clarimón M, Sussams J, Lovestone R, O’Donovan S, Owen MC, Behrens MJ, Mead TW, Uitterlinden S, Holmes AG, Cruchaga C, Ingelsson C, Bennett M, Powell DA, Golde J, Graff TE, De Jager C, Morgan PL, Ertekin-Taner K, Combarros N, Psaty O, Passmore BM, Younkin P, Berr SG, Gudnason C, Rujescu V, Dickson D, Dartigues DW, Destefano JF, Ortega-Cubero AL, Hakonarson S, Campion H, Boada D, Kauwe M, Farrer JK, Van Broeckhoven LA, Ikram C, Jones MA, Haines L, Tzourio JL, Tzourio C, Escott-Price C, Mayeux V, Deleuze R, Amin JF, Goate N, Pericak-Vance AM, Amouyel MA, Van Duijn P, Ramirez CM, Wang A, Lambert LS, Seshadri JC, Williams S, Schellenberg J,D,G, 2017. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. In: Nat. Genet, 499, pp. 1373–1384. 10.1038/ng.3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slepnev V, Ochoa G, … J of B MB., 2000, undefined, n.d. Tandem arrangement of the clathrin and AP-2 binding domains in amphiphysin 1 and disruption of clathrin coat function by amphiphysin fragments comprising these. ASBMB. [DOI] [PubMed] [Google Scholar]
- Solé-Domènech S, Cruz DL, Capetillo-Zarate E, Maxfield FR, 2016. The endocytic pathway in microglia during health, aging and Alzheimer’s disease. Ageing Res. Rev 10.1016/j.arr.2016.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spangenberg E, Severson PL, Hohsfield LA, Crapser J, Zhang J, Burton EA, Zhang Y, Spevak W, Lin J, Phan NY, Habets G, Rymar A, Tsang G, Walters J, Nespi M, Singh P, Broome S, Ibrahim P, Zhang C, Bollag G, West BL, Green KN, 2019. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat. Commun 10, 3758. 10.1038/s41467-019-11674-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spiller KJ, Restrepo CR, Khan T, Dominique MA, Fang TC, Canter RG, Roberts CJ, Miller KR, Ransohoff RM, Trojanowski JQ, Lee VMY, 2018. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci 21, 329–340. 10.1038/s41593-018-0083-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stancu I-CC, Cremers N, Vanrusselt H, Couturier J, Vanoosthuyse A, Kessels S, Lodder C, Brône B, Huaux F, Octave J-NN, Terwel D, Dewachter I, 2019. Aggregated Tau activates NLRP3–ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 137, 599–617. 10.1007/s00401-018-01957-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinman RM, Brodie SE, Cohn ZA, 1976. Membrane flow during pinocytosis: a stereologic analysis. J. Cell Biol 68, 665–687. 10.1083/jcb.68.3.665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stephan AH, Barres BA, Stevens B, 2012. The complement system: an unexpected role in synaptic pruning during development and disease. Annu. Rev. Neurosci 10.1146/annurev-neuro-061010-113810. [DOI] [PubMed] [Google Scholar]
- Streit WJ, Braak H, Xue QS, Bechmann I, 2009. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 118, 475–485. 10.1007/s00401-009-0556-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streit WJ, Xue QS, Tischer J, Bechmann I, 2014. Microglial pathology. Acta Neuropathol. Commun 2, 1–17. 10.1186/s40478-014-0142-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun W, Samimi H, Gamez M, Zare H, Frost B, 2018. Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies. Nat. Neurosci 21, 1038–1048. 10.1038/s41593-018-0194-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szondy Z, Sarang Z, Kiss B, Garabuczi É, Köröskényi K, 2017. Anti-inflammatory mechanisms triggered by apoptotic cells during their clearance. Front. Immunol 8, 909. 10.3389/FIMMU.2017.00909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takeda S, Wegmann S, Cho H, DeVos SL, Commins C, Roe AD, Nicholls SB, Carlson GA, Pitstick R, Nobuhara CK, Costantino I, Frosch MP, Müller DJ, Irimia D, Hyman BT, 2015. Neuronal uptake and propagation of a rare phosphorylated high-molecular-weight tau derived from Alzheimer’s disease brain. Nat. Commun 6, 8490. 10.1038/ncomms9490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thrupp N, Sala Frigerio C, Wolfs L, Skene NG, Fattorelli N, Poovathingal S, Fourne Y, Matthews PM, Theys T, Mancuso R, de Strooper B, Fiers M, 2020. Single-nucleus RNA-Seq is not suitable for detection of microglial activation genes in humans. Cell Rep. 32. 10.1016/J.CELREP.2020.108189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tse SML, Furuya W, Gold E, Schreiber AD, Sandvig K, Inman RD, Grinstein S, 2003. Differential role of actin, clathrin, and dynamin in Fcγ receptor-mediated endocytosis and phagocytosis. J. Biol. Chem 278, 3331–3338. 10.1074/JBC.M207966200. [DOI] [PubMed] [Google Scholar]
- Turk V, Stoka V, Vasiljeva O, Renko M, Sun T, Turk B, Turk D, 2012. Cysteine cathepsins: from structure, function and regulation to new frontiers. Biochim. Biophys. Acta - Proteins Proteom 10.1016/j.bbapap.2011.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uchihara T, Tsuchiya K, Nakamura A, Ikeda K, 2000. Appearance of tau-2 immunoreactivity in glial cells in human brain with cerebral infarction. Neurosci. Lett 286, 99–102. 10.1016/S0304-3940(00)01103-4. [DOI] [PubMed] [Google Scholar]
- Uchihara T, Nakamura A, Arai T, Ikeda K, Tsuchiya K, 2004. Microglial tau undergoes phosphorylation-independent modification after ischemia. Glia 45, 180–187. 10.1002/GLIA.10318. [DOI] [PubMed] [Google Scholar]
- Uchihara T, Duyckaerts C, Seilhean D, Nakamura A, Lazarini F, Hauw JJ, 2005. Exclusive induction of tau2 epitope in microglia/macrophages in inflammatory lesions-tautwopathy distinct from degenerative tauopathies. Acta Neuropathol. 109, 159–164. 10.1007/S00401-004-0922-3. [DOI] [PubMed] [Google Scholar]
- Uhlemann R, Gertz K, Boehmerle W, Schwarz T, Nolte C, Freyer D, Kettenmann H, Endres M, Kronenberg G, 2016. Actin dynamics shape microglia effector functions. Brain Struct. Funct 221, 2717–2734. 10.1007/s00429-015-1067-y. [DOI] [PubMed] [Google Scholar]
- Ulrich JD, Finn MB, Wang Y, Shen A, Mahan TE, Jiang H, Stewart FR, Piccio L, Colonna M, Holtzman DM, 2014. Altered microglial response to Aβ plaques in APPPS1-21 mice heterozygous for TREM2. Mol. Neurodegener 9, 1–9. 10.1186/1750-1326-9-20/FIGURES/5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Umpierre AD, Bystrom LL, Ying Y, Liu YU, Worrell G, Wu LJ, 2020. Microglial calcium signaling is attuned to neuronal activity in awake mice. Elife 9, 1–24. 10.7554/eLife.56502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Olst L, Verhaege D, Franssen M, Kamermans A, Roucourt B, Carmans S, Ytebrouck E, van der Pol SMA, Wever D, Popovic M, Vandenbroucke RE, Sobrino T, Schouten M, de Vries HE, 2020. Microglial activation arises after aggregation of phosphorylated-tau in a neuron-specific P301S tauopathy mouse model. Neurobiol. Aging 89, 89–98. 10.1016/j.neurobiolaging.2020.01.003. [DOI] [PubMed] [Google Scholar]
- Vidarsson G, Dekkers G, Rispens T, 2014. IgG subclasses and allotypes: from structure to effector functions. Front. Immunol 5, 520. 10.3389/fimmu.2014.00520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Li Y, Ryder JW, Hole JT, Ebert PJ, Airey DC, Qian HR, Logsdon B, Fisher A, Ahmed Z, Murray TK, Cavallini A, Bose S, Eastwood BJ, Collier DA, Dage JL, Miller BB, Merchant KM, O’Neill MJ, Demattos RB, 2018. Genome-wide RNAseq study of the molecular mechanisms underlying microglia activation in response to pathological tau perturbation in the rTg4510 tau transgenic animal model. Mol. Neurodegener 13. 10.1186/s13024-018-0296-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Jin WS, Bu X., Le, Zeng F, Huang ZL, Li WW, Shen LL, Zhuang ZQ, Fang Y, Sun BL, Zhu J, Yao XQ, Zeng GH, Dong ZF, Yu JT, Hu Z, Song W, Zhou HD, Jiang JX, Liu YH, Wang YJ, 2018. Physiological clearance of tau in the periphery and its therapeutic potential for tauopathies. Acta Neuropathol. 136, 525–536. 10.1007/s00401-018-1891-2. [DOI] [PubMed] [Google Scholar]
- Wang P, Ye Y, 2020. An integrin receptor complex mediates filamentous Tau-induced activation of primary astrocytes. bioRxiv 2020.05.07.083295. 10.1101/2020.05.07.083295. [DOI] [Google Scholar]
- Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, Yuan P, Mahan TE, Shi Y, Gilfillan S, Cella M, Grutzendler J, DeMattos RB, Cirrito JR, Holtzman DM, Colonna M, 2016. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J. Exp. Med 213, 667–675. 10.1084/JEM.20151948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegmann S, Maury EA, Kirk MJ, Saqran L, Roe A, DeVos SL, Nicholls S, Fan Z, Takeda S, Cagsal-Getkin O, William CM, Spires-Jones TL, Pitstick R, Carlson GA, Pooler AM, Hyman BT, 2015. Removing endogenous tau does not prevent tau propagation yet reduces its neurotoxicity. EMBO J. 34, 3028–3041. 10.15252/embj.201592748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegmann S, Nicholls S, Takeda S, Fan Z, Hyman BT, 2016. Formation, release, and internalization of stable tau oligomers in cells. J. Neurochem 139, 1163–1174. 10.1111/jnc.13866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wigge P, Köhler K, Vallis Y, Doyle CA, Owen D, Hunt SP, McMahon HT, 1997. Amphiphysin heterodimers: Potential role in clathrin-mediated endocytosis. Mol. Biol. Cell 8, 2003–2015. 10.1091/MBC.8.10.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wittrup A, Zhang SH, ten Dam GB, van Kuppevelt TH, Bengtson P, Johansson M, Welch J, Mörgelin M, Belting M, 2009. ScFv antibody-induced translocation of cell-surface heparan sulfate proteoglycan to endocytic vesicles: evidence for heparan sulfate epitope specificity and role of both syndecan and glypican. J. Biol. Chem 284, 32959–32967. 10.1074/jbc.M109.036129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu JW, Hussaini SA, Bastille IM, Rodriguez GA, Mrejeru A, Rilett K, Sanders DW, Cook C, Fu H, Boonen RACM, Herman M, Nahmani E, Emrani S, Figueroa YH, Diamond MI, Clelland CL, Wray S, Duff KE, 2016. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat. Neurosci 19, 1085–1092. 10.1038/nn.4328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE, Wozniak DF, Diamond MI, Holtzman DM, 2013. Anti-Tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 80, 402–414. 10.1016/j.neuron.2013.07.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao W, Li K, Liao K, 2009. Macropinocytosis contributes to the macrophage foam cell formation in RAW264.7 cells. Acta Biochim. Biophys. Sin. (Shanghai) 41, 773–780. 10.1093/abbs/gmp066. [DOI] [PubMed] [Google Scholar]
- Yoshiyama Y, Higuchi M, Zhang B, Huang S-M, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VM-Y, 2007. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351. 10.1016/j.neuron.2007.01.010. [DOI] [PubMed] [Google Scholar]
- Yuan P, Condello C, Keene CD, Wang Y, Bird TD, Paul SM, Luo W, Colonna M, Baddeley D, Grutzendler J, 2016. TREM2 Haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90, 724–739. 10.1016/J.NEURON.2016.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zareba-Paslawska J, Patra K, Kluzer L, Revesz T, Svenningsson P, 2021. Tau isoform-driven CBD pathology transmission in oligodendrocytes in humanized tau mice. Front. Neurol 11. 10.3389/fneur.2020.589471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan L, Fan L, Kodama L, Sohn PD, Wong MY, Mousa GA, Zhou Y, Li Y, Gan L, 2020. A mac2-positive progenitor-like microglial population is resistant to csf1r inhibition in adult mouse brain. Elife 9, 1–22. 10.7554/eLife.51796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, Deng S, Liddelow SA, Zhang C, Daneman R, Maniatis T, Barres BA, Wu JQ, 2014. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, Seyfried NT, Hu WT, Liu Z, Wang JZ, Cheng L, Sun YE, Yu SP, Levey AI, Ye K, 2014. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat. Med 20, 1254–1262. 10.1038/nm.3700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Lu M, Du RH, Qiao C, Jiang CY, Zhang KZ, Ding JH, Hu G, 2016. MicroRNA-7 targets Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease. Mol. Neurodegener 11, 1–15. 10.1186/s13024-016-0094-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zilkova M, Nolle A, Kovacech B, Kontsekova E, Weisova P, Filipcik P, Skrabana R, Prcina M, Hromadka T, Cehlar O, Rolkova GP, Maderova D, Novak M, Zilka N, Hoozemans JJM, 2020. Humanized tau antibodies promote tau uptake by human microglia without any increase of inflammation. Acta Neuropathol. Commun 81 (8), 1–19. 10.1186/S40478-020-00948-Z. [DOI] [PMC free article] [PubMed] [Google Scholar]